Next Article in Journal
A Retrospective Review of Calcineurin Inhibitors’ Impact on Cytomegalovirus Infections in Lung Transplant Recipients
Next Article in Special Issue
Development of a Porcine Slaughterhouse Kidney Perfusion Model
Previous Article in Journal
Laparoscopic Living-Donor Nephrectomy of a Horseshoe Kidney: A Case Report and Review of the Literature
Previous Article in Special Issue
Ex Vivo Lung Perfusion: A Platform for Donor Lung Assessment, Treatment and Recovery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Hypothermic and Normothermic Perfusion in Kidney Transplantation

by
Thomas B. Smith
*,
Michael L. Nicholson
* and
Sarah A. Hosgood
*
Department of Surgery, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK
*
Authors to whom correspondence should be addressed.
Transplantology 2021, 2(4), 460-477; https://doi.org/10.3390/transplantology2040044
Submission received: 14 August 2021 / Revised: 26 September 2021 / Accepted: 2 November 2021 / Published: 17 November 2021
(This article belongs to the Special Issue Machine Perfusion in Organ Transplantation)

Abstract

:
Hypothermic and normothermic machine perfusion in kidney transplantation are purported to exert a beneficial effect on post-transplant outcomes compared to the traditionally used method of static cold storage. Kidney perfusion techniques provide a window for organ reconditioning and quality assessment. However, how best to deliver these preservation methods or improve organ quality has not yet been conclusively defined. This review summarises the promising advances in machine perfusion science in recent years, which have the potential to further improve early graft function and prolong graft survival.

1. Introduction

Kidney transplantation is the most economical [1] and effective [2,3,4,5] therapy for patients with end-stage renal disease (ESRD). However, there is a worldwide shortage of suitable kidneys for transplantation [6]. Over the next decade, the incidence of chronic kidney disease (CKD) and ESRD is expected to increase considerably, with CKD due to become the fifth leading cause of death by 2040 [7]. Strategies that increase the number of kidneys available for transplantation or improve transplant success rates and outcomes are likely to have a considerable effect on global health.
Machine perfusion technologies have emerged as an important tool in tackling critical problems intrinsic to transplantation, such as ischaemia reperfusion injury (IRI) [7,8,9], poor post-transplant graft function [10,11,12] and reduced graft survival [10]. Understanding how machine perfusion ameliorates these problems and optimising these methods will likely further improve patient outcomes. Although the central goal of this research (i.e., increasing the availability and quality of transplant kidneys) is uniform, the means by which this could potentially be achieved differs. Optimisation of machine perfusion technologies may improve kidney transplantation in three ways:
-
Improvement of transplant outcomes through delivery of therapeutic agents to repair and regenerate kidneys.
-
Reduction in the number of discarded kidneys by developing robust techniques of organ assessment.
-
Reduction in ischaemic injury during the preservation interval to improve the ‘shelf life’ of donated kidneys and increase the number available for transplant.
This review focuses on the current understanding of the biological factors that negatively affect kidney quality and the physiological effects of hypothermic and normothermic techniques. Recent advances that further our understanding of how we can adapt machine perfusion to improve outcomes or assess kidney quality will also be discussed.

2. Why Do We Need Organ Preservation? What Are the Factors Diminishing Kidney Quality?

Modifiable factors that have a key impact on pre-transplant kidney quality are the periods of ischaemia that occur prior to transplant and the reperfusion injury that occurs following transplant. These are illustrated in Figure 1 and described below.

3. Warm Ischaemia

Preservation techniques maintain organ viability from the time of retrieval until transplantation. These techniques are required to counteract the destructive processes initiated during warm ischaemia. In general, warm ischaemia arises prior to donation [13] and results in impaired delivery of oxygen and metabolic substrates [14,15]. This drives an anaerobic shift [14] and crucially ATP depletion [16], which results in widespread deterioration of tissue structure [17,18,19,20,21,22]. The warm ischemic injury incurred also stimulates damaging inflammatory responses [23].
The warm ischaemic time (WIT) is associated with increased incidences of delayed graft function (DGF) [24] and therefore, the initial role of kidney preservation is to reduce ATP depletion, cell swelling and hypoxic injury. This is achieved by rapidly flushing the kidney at procurement with a cold preservation solution to slow metabolism and requirements for ATP.

4. Cold Ischaemia

Although effective in reducing metabolism, ongoing depletion of ATP leads to cold ischaemic damage. The cold ischaemic time (CIT) is an independent risk factor for the development of DGF [25]. The mechanisms of damage conferred under conditions of cold ischaemia have been described elsewhere [26].

5. Current Kidney Preservation Methods, Their Advantages and Limitations

5.1. Static Cold Storage

Static cold storage (SCS) is a simple and economical method of kidney preservation. Kidneys are placed in a bag of preservation solution and packed in wet ice, lowering the temperature to around 4 °C. At this temperature, enzymatic activity is reduced by approximately 58% [27]. Different preservations solutions are available, but University of Wisconsin (UW) solution is deemed the gold standard [27]. An overview of solutions used in SCS and the perfusion technologies described below is given in Table 1.

5.2. Hypothermic Machine Perfusion

Hypothermic machine perfusion (HMP) involves the circulation of cold preservation fluid through the kidney using a mechanical pump. With the exception of several recent clinical trials, clinical HMP does not utilise active oxygenation. The limited metabolic support provided in the perfusion fluid was thought to be sufficient to meet the residual aerobic requirements under hypothermia [18].
ROS are a primary driver of reperfusion injury [30], and commonly used preservation fluids contain potent antioxidants, such as glutathione, to combat ROS activity during preservation. However, extended CIT is associated with marked perfusate glutathione depletion [31].
Machine perfusion solutions provide low-level metabolic support and antioxidant protection throughout perfusion. A key difference between HMP and SCS is the provision of fluid flow, which facilitates nutrient supply, waste removal and a limited amount of tissue reoxygenation with the dissolved oxygen present in the perfusate.
There are several commercially available HMP devices. The Organ Assist Kidney Assist, Waters RM3 and Organ Recovery Systems Lifeport are pressure-controlled systems designed to limit mechanical damage to the kidney during perfusion. Several new devices have been trialled, such as the AirDrive system which includes an oxygenator [32]. A new two-pump perfusion device which circulates fluid through the kidney and also in the organ reservoir has been used to deliver clinical HMP [33,34].
A number of randomised controlled trials and a meta-analysis have shown superiority of HMP over SCS techniques in improving early and longer-term graft function; however, despite this evidence HMP has not gained wide acceptance in some countries.
The evidence base supporting the use of HMP for all deceased donor kidneys is growing, with benefits recently reported in the UK [35,36], France [37], Poland [38] and Brazil [39,40]. HMP can also improve renal function when the CIT is extended [35].
In extend criteria donor (ECD) kidneys, use of HMP enhances 1-year graft survival [41]. However, HMP has not shown a convincing benefit in prolonging longer-term graft survival [42]. The Netherlands is the first country to introduce HMP for all deceased donor kidneys as standard practice [43]. Other countries use HMP specifically for donation after circulatory death (DCD) kidneys, but this practice is not uniform.

5.3. Normothermic Machine Perfusion

Normothermic machine perfusion (NMP) is a relatively new technique of preservation in kidney transplantation. It is currently used in combination with hypothermic preservation strategies as a form of end graft reconditioning. During NMP, kidneys are perfused at near-physiological temperatures and pressures allowing cellular metabolism and function to be restored. In the 1980s, interest in NMP using oxygenated blood-based perfusion solutions started to emerge, and demonstrated that short intervals or an end period of NMP could replenish cellular ATP [44].
The first case of NMP in clinical practice was published in 2011 [45]. The recipient received a kidney from an ECD donor that had been rejected by five other transplant centres in the UK. The kidney underwent NMP for a short interval immediately before transplantation. The recipient did not require dialysis post-transplant and 10 years post-transplant has normal kidney function (personal communication). Subsequently a series of NMP in ECD kidneys demonstrated a remarkably low rate of DGF (11%) compared to SCS kidneys (37%).
More recently, NMP has been used to rescue kidneys that were deemed unsuitable for transplant due to inadequate in situ perfusion after retrieval. Both kidneys were transplanted successfully with immediate graft function following transplant [46]. Building on this work, the authors developed a scoring system which could be used to assess kidney quality prior to transplant [46,47]. A large multicentre clinical trial assessing the effects of 1 h NMP in DCD kidneys compared to SCS has been completed and is due to report this year [28].
NMP conditions are still being developed and have recently been used to counteract ‘rewarming injury’ which occurs during the warm reperfusion of cold stored grafts. In 2015, the Minor group demonstrated that gradual rewarming (controlled oxygenated rewarming (COR)) of cold stored kidney grafts using machine perfusion improve creatinine clearance and reduces apoptotic signalling when compared to cold stored controls [48].
Kidneys are rewarmed (8–35 °C) over a 1.5 h period to allow metabolic adaptation to the changing thermal environment. Building on this work, the same group trialled their method in the clinical setting, reporting immediate graft function and acceptable levels of creatinine clearance within 1 week of transplantation [29].
More recently, a porcine auto-transplantation model demonstrated that while 8h of NMP improves renal function compared to SCS, a similar improvement in renal function is observed when cold-stored kidneys are subjected to 2 h of COR [49]. The authors speculate that this may be a useful application given the current requirement for hypothermic storage in the logistics of organ transport.
The perfusates used in clinical NMP have been defined in Table 1. The Hosgood et al. protocol provides a more physiological environment, with multiple metabolic substrates and red cells as an oxygen carrier. This contrasts with the Minor protocol, which utilises an acellular perfusate based on Steen solution [29,50].
Adapted cardiac bypass technologies or other perfusion set-ups can be used for NMP. The Kidney Assist, a pressure-controlled system, is the only CE-marked device on the market.
There are some limitations of NMP compared to HMP. These include a more complicated, expensive perfusion circuit and the requirement of personnel for continuous monitoring of the kidney. In a recent publication, RNA sequencing of kidney tissue before and after NMP demonstrated the upregulation of genes associated with oxidative phosphorylation but also inflammatory pathways [51]. Modulation of NMP conditions by incorporating a cytokine filter into the circuit removed the inflammatory cytokines from the perfusate and reduced the inflammatory gene expression.
There is international interest in the development and clinical deployment of NMP [52] and it is the subject of other current clinical trials. The feasibility and safety of normothermic ex vivo kidney perfusion (NEVKP) trial will recruit 25 patients who receive a kidney after 1–10 h of NMP, and assess the device failure rate alongside standard measures of outcome such as DGF, graft failure and patient survival (Clinicaltrials.gov ID: NCT03136848). Perfusion at subnormothermic (20–32 °C) temperatures is also being explored [53]. A new clinical trial is due to start called ‘Oxygenated machine preservation in kidney transplantation’ (SNOPO; Clinicaltrials.gov ID: NCT04540640), which will address the safety of subnormothermic machine perfusion on transplant kidneys. This is an explorative trial that will also assess the rate of graft discard and assess graft function.
An overview of the active trials investigating both HMP and NMP is given in Table 2.
Alongside this, the more physiological environment generated by NMP permits the use of therapeutic agents to mediate cellular physiology ex vivo and in recent years there have been numerous developments in potential additives to the NMP perfusate which may confer clinical benefit. These are detailed below.

6. What Are the Advances in HMP?

6.1. Active Perfusate Oxygenation under HMP

A key focus in the optimisation of HMP over recent years has been the addition of oxygen. Aerobic metabolism, through facilitation of the electron transport chain, generates more ATP than anaerobic glycolysis, which, alongside compounding the injury caused by ATP depletion, also results in deleterious tissue acidification through the enhanced production of lactic acid [54].
Early studies using murine models demonstrated potential benefits of actively oxygenating the perfusate, with perfusate oxygen consumption during HMP appearing to be linked to improvements in post-transplant glomerular filtration rate (GFR) [55]. This was supported by a porcine kidney ex vivo reperfusion model, in which 21 h of HMP combined with provision of 100% oxygen resulted in improved blood flow and creatinine clearance, when compared with non-oxygenated HMP. In addition, non-oxygenated HMP resulted in evidence of greater tubular damage [56].
The Consortium for Organ Preservation in Europe (COPE) recently reported the COMPARE trial, a randomised controlled phase 3 trial comparing the effects of continuous HMP with oxygen, with HMP without oxygen in pairs of DCD kidneys using the Kidney Assist Device. A small improvement in estimated GFR (eGFR) and a lower incidence of graft failure was observed when compared to standard HMP (i.e., 3% vs. 10%, p = 0.028) at 1 year when sensitivity analysis was applied, but there was no significant difference in graft function, the primary outcome measure if both pairs from the same donor were functioning at 1 year [57]. Shorter periods of oxygenated HMP may also have some benefit. In a porcine model, active perfusate oxygenation was found to promote enhanced restoration of aerobic metabolism and also increase endpoint cortical ATP concentrations [58]. This latter finding was repeated by another group, who documented no effect of perfusate oxygenation on graft function, but noted increased endpoint ATP concentrations as a consequence of oxygenation [59].
There are concerns that such high-level oxygenation could be surplus to requirements and actually be deleterious to the preserved tissues through enhanced ROS activity [60].
Using an ex vivo ischaemia reperfusion model, it has been demonstrated that continuous oxygenation under HMP results in superior early graft recovery when compared to SCS, various transient oxygenation strategies and also end ischaemic NMP strategies [61].
Extending this into an auto-transplantation model, the same group compared different oxygenation strategies. The aim of these studies was to identify the minimal oxygenation strategy that provides a therapeutic effect, but avoids the potential for unnecessary oxidative stress. Oxygenation with a concentration of 30% or 90% resulted in no significant differences in functional outcomes; however, kidneys perfused with higher concentrations of oxygen had reduced perfusate lactate concentration [62].
The same group also examined methods of oxygenation. Kidneys received 22 h HMP with or without a 2 h oxygenation period at the beginning or end of perfusion [63]. In the first 4 days after transplant, kidneys that had received 22 h continuous oxygenation and 2 h oxygenation at the start of HMP had improved graft function compared to those without oxygenation or with HMP with 2 h of end oxygenation. While end ischaemic oxygenation may offer benefit, initial oxygen uploading appears the most promising transient oxygenation strategy. This may be delivered in the clinical setting using currently approved devices. The Waters and Kidney Assist Devices include the capacity for an oxygenator; the Organ Recovery Systems Lifeport is also capable of oxygenation through bubble and surface oxygenation methods [64].
In clinical kidney transplantation, the COPE consortium recently published the results of the HOPE trial. The effects of end ischaemic oxygenated HMP was trialled in ECD kidneys and compared to ECD kidneys undergoing SCS. No significant difference in DGF incidence was observed between group [65].
In contrast, a study published in letter format to the Journal of Clinical and Translational Research proposed end ischaemic oxygenation to be particularly therapeutic in renal HMP [66].
Other studies examining the effects of oxygenated HMP using these different strategies are also being carried out (Clinicaltrials.gov ID: NCT03837197).

6.2. Modification of Perfusate under HMP

M101 is an extracellular haemoglobin with a high oxygen-carrying capability. Porcine kidneys perfused with fluids supplemented with M101 had reduced vascular resistance during perfusion and less fibrosis after reperfusion [67].
M101 was assessed in a clinical study including 60 pairs of donation after brain death (DBD) kidneys. The OXYPOP study added M101 during HMP and SCS and found no adverse outcomes. There was some indication of improved early graft function [68].
Perfusate supplementation with Vectisol® is a recent advance that shows preclinical potential. Vectisol is a revesterol–cyclodextin conjugate which increases the solubility of the potent antioxidant resveratrol. When compared to porcine kidneys stored using standard HMP protocols, supplementing HMP perfusate kidney perfusion solution 1 (KPS-1) with 1.56 g Vectisol reduced the levels of oxidative stress and apoptosis in a porcine auto-transplantation model. In addition, Vectisol reduced total plasma levels of superoxide dismutase and lowered plasma creatinine levels at 3 months post-transplant compared to standard HMP [69].
Another recent development in perfusate optimisation includes perfusate supplementation with macromolecular heparin. Reperfusion results in rapid degradation of the endothelial glycocalyx. Using immunofluorescence analysis and confocal microscopy, it has been shown that heparin administered during HMP binds to the vascular endothelium lining the perfused vasculature and, simultaneously, perfusate concentrations of macromolecular heparin are depleted [70]. Using a paired porcine perfusion model with an ex vivo reperfusion model, the same group demonstrated that administration of 50 mg macromolecular heparin resulted in lower renal resistance and faster reductions in serum creatinine than kidneys that did not receive heparin administration. Additionally, heparinised kidneys had lower levels of the biomarker neutrophil gelatinase-associated lipocalin (NGAL) [71].
The ‘Renaparin® in kidney transplantation’ placebo-controlled trial is currently underway, in which kidney transplant recipients will receive a kidney stored using conventional HMP or HMP with the addition of Renaparin, a heparin analogue. This study will assess the incidence of DGF and adverse events within the first 30 post-operative days (Clinicaltrials.gov ID: NCT03773211).

6.3. Measures of Graft Quality during HMP

In recent years, there has been considerable focus on non-invasive methods through which kidney quality can be assessed during HMP. The most readily available indices on kidney quality come from macroscopic inspection: larger kidneys with poor perfusion at procurement are more likely to go on to experience impaired graft function post-transplant [72]. However, the perfusion period offers more ability to assess organ quality.
An assessment that can be performed in real time is the measurement of perfusion parameters. Renal resistance may have some value in kidney assessment. A renal resistance of <1 mmHg within the first hour of perfusion was associated with reduction in the incidence of DGF and primary non-function (PNF) and also improved rate of decrease in post-operative serum creatinine [73].
The value of using renal resistance to predict transplant outcomes was also demonstrated recently in an oxygenated HMP study. The percentage fall in renal resistance was found to be inversely correlated to serum creatinine on day 1 and 2 after transplant, indicating a functional link between an individual kidney’s response to perfusion during storage and graft function [65]. Patients who receive a kidney that exhibited high resistance during the first hour of HMP have higher post-operative serum creatinine at 1 year and a higher incidence of DGF immediately post-transplant [74].
Additionally, a high endpoint renal resistance was reported again to be a strong risk factor for the development of DGF [75] and a retrospective analysis has recently linked high renal resistance with graft failure [76]. Kidneys with resistance readings >0.19 mmHg/mL/min have a higher risk of acute rejection and inferior one-year graft survival compared to kidneys with an endpoint resistance of <0.19 mmHg/min [77].
Nonetheless, the utility of renal resistance in prediction of PNF or long-term kidney graft outcomes appears to be limited [78].
Measuring intracellular ATP using magnetic resonance imaging (MRI) is another strategy which could be used to assess kidney quality. MRI is a non-destructive technique which uses the unique magnetic profile of compounds containing MRI sensitive nuclei to produce images of the biological vessel they inhabit. With a protocol that specifically targets the 31P isotope of phosphorous, a Swiss study showed the deleterious effects of warm ischaemia on intracellular ATP concentrations and also enhanced ATP resynthesis during oxygenated perfusion. ATP levels were linked with the level of damage observed upon tissue examination [79].
Alternatively, the use of optical coherence tomography has been reported as another non-invasive technique which can be used to define macroscopic structural changes in the shallow cortex of pre-transplant human kidneys. Larger proximal tubule diameters had predictive potential for DGF development, and shorter distances between separate proximal tubule lumens was also indicative of DGF [80].
Monitoring of ongoing metabolism during organ preservation has been proposed as a useful method to assess kidney quality [81]. However, logistically, this requires rapid analysis and the use of perfusate samples.
Rapid sampling microdialysis has been applied during kidney perfusion, permitting a continual acquisition of tissue-derived analyte into a detection system [82]. While this approach does not offer comprehensive metabolic profiling, it does allow for continuous profiling of metabolites of interest, such as lactate, and may have a role in defining kidney quality before transplant. This technology will be applied in the upcoming REMO-HYMAP trial (Real-time monitoring of kidney grafts on hypothermic machine perfusion) (Clinicaltrials.gov ID: NCT04619732).
Another trial, ‘Metabolomics in assessing the quality of kidney transplants retained on a LifePort® perfusion machine’ (RENALIFE), will utilise nuclear magnetic resonance (NMR) spectroscopy to define metabolic changes in the perfusate and determine whether these are linked with graft function post-transplant (Clinicaltrials.gov ID: NCT03024229).
A similar study, the 13 Champion clinical study, will also utilise NMR to chart changing perfusate metabolic profiles prior to transplantation, with a perfusate modification enabling tracing of glucose metabolism (i.e., substitution of conventional naturally occurring glucose for glucose that has been carbon-13 enriched). This study will report the results in 2022 [83].
A complementary indicator of graft quality that may be used alongside measures of renal resistance is the detection of damage-associated biomarkers in the perfusate, of which many have been proposed and studied [84,85].
A recent meta-analysis assessed the published literature linking potential perfusate biomarkers with post-transplant outcomes. This study found that assessment of glutathione S transferase offered the best predictive potential for DGF and PNF [86].

7. What Are the Advances in NMP?

For logistical reasons, NMP is used in combination with hypothermic techniques of preservation. Therefore, kidneys are subject to periods of cold ischaemia. Furthermore, after NMP, it is necessary to flush the kidney with cold preservation solution to remove the red cell-based perfusate and cool the kidney in preparation for transplantation. Anastomosis of the renal vessels can take up to 60 min and cooling helps to prevent warm ischaemic injury.
One approach to avoid ischaemic injury is to continually perfuse the kidney using NMP conditions. A single-case report demonstrated use of the Kidney Assist apparatus to perfuse a kidney from a DBD donor from the time of retrieval until transplantation. Although logistically and technically difficult, the authors demonstrated proof of principle that it is possible to avoid ischaemic injury using this approach [87].
A more practical approach is normothermic regional perfusion (NRP), in which kidneys are perfused in situ within the donor’s body at the time of retrieval. NRP reduces the incidence of PNF in kidneys from DCD donors compared to in situ cooling [88]. Porcine models have demonstrated that 4 h of NRP is an optimal timeframe to limit kidney injury [89].

7.1. Modification of the Normothermic Perfusion Conditions

NMP may utilise red cells as oxygen carriers [50]; however, with the potential for haemolysis to occur, alternatives are desirable.
Haemoglobin-based oxygen carriers (HBOCs) may be a suitable alternative. Using 14 discarded human kidneys, a 2019 study demonstrated that the use of perfusate supplemented with HBOC (n = 7) exerted no significant differences in oxygen consumption or endpoint tissue ATP levels when compared to perfusate supplemented with packed red cells (n = 7) [90]. In addition, no differences in renal resistance or tissue histology were noted. This indicates that a cellular perfusate may not be required for optimal delivery of NMP.
NMP delivered with controlled rewarming and an acellular perfusate was found to improve creatinine clearance and reduce expression markers of innate immune activation when compared to SCS stored kidneys [91]. However, no benefit was gained by addition of red cells, indicating that they may not be necessary for NMP.
An alternative to supporting oxygen requirements is to suppress oxygen requirements by administering hydrogen sulphide (H2S) during NMP. Hydrogen sulphide inhibits the activity of complex IV of the mitochondrial electron transport chain. A study investigating the effects of H2S supplementation during perfusion demonstrated a 61% decrease in oxygen consumption; however, this suppression was quickly reversed after stopping H2S administration [92]. This suggests that continuous delivery of hydrogen sulphide may be required for longer preservation periods. Nonetheless, no differences in damage indicators were found between H2S and control kidneys.

7.2. Regenerative Therapies

NMP can also be used to administer regenerative therapies to the kidney. Mesenchymal stem cells (MSCs) have the capability to repair cellular damage and immunomodulation properties to reduce the immune response. To date, large animal models have demonstrated the safety and feasibility of introducing MSCs in this way [93].
Factors limiting the application of MSCs during NMP include observations that the infused cells localise within glomerular lumen and may not confer protection to post-glomerular structures such as the proximal tubules [94]. Additionally, high doses of MSCs (i.e., 107) appear to be required as cells can adhere to the filters and tubing of the NMP circuit.
Nonetheless, when trialled in pairs of non-transplanted human kidneys, the infusion of multipotent adult progenitor cells (MAPCs) resulted in decreased injury markers and improvements in both cortical microcirculation and urine output when compared to controls [95]. In a similar study, infusion of MSCs resulted in downregulation of inflammatory cytokines, improvements to tissue ATP concentrations and evidence of enhanced cellular repair [96].

7.3. Therapeutic Agents

A different approach may be to add compounds that target specific immune processes. An Australian study documented the use of a small animal model to determine the efficacy of three drugs added to the NMP perfusate. Of a CD47 receptor agonist (αCD47ab), soluble complement receptor 1 and recombinant thrombomodulin, the antibody therapy proved the most effective in ameliorating IRI. When this was taken forward to a porcine NMP auto-transplant model, the antibody was shown to significantly improve perfusion parameters when compared to controls [97].
Cold ischaemia causes fibrinogen to accumulate in the proximal tubule epithelium [98]. Upon reperfusion, this is released and causes red cell aggregates that plug the renal vasculature. Kidneys can be treated during NMP with the addition of plasminogen and plasminogen activator to clear the microcirculation and improve function [98].
An exciting development recently applied to the kidney is the use of NMP to deliver lentiviral vectors containing constructs that silence expression of major histocompatibility complex (MHC) I and II in rats [99]. This approach has the potential to mask human leucocyte antigen (HLA) differences between donor and recipient and reduce the risk of acute rejection. This study reported no evidence of damage to the kidney; however, there was a significant increase in the levels of inflammatory cytokines detected.
Restoring cellular processing during NMP after ischaemic injury inevitably leads to the upregulation of pro-inflammatory cytokines. The perfusion conditions can be modified by including a cytosorb filter in the NMP circuit to remove the circulating cytokines, which reduces the inflammatory gene expression within the kidney [51].

7.4. Modifying the Duration of NMP

At present, reports of NMP in clinical kidney transplantation use short durations of NMP (1–2 h). This is for logistical reasons and from evidence in experimental work demonstrating that cellular ATP is restored within this timeframe.
A recent study in the Netherlands reported the effect on recipients over the age of 65 of receiving a kidney from an ECD following 2 h of NMP [100]. The follow up included 11 patients who received a kidney stored by NMP and a historical cohort of 53 patients who received a kidney stored by HMP or SCS. This study found no differences in graft outcomes within the 1 year follow up, indicating that within this timeframe a 2 h preservation time is safe.
The PROlonged ex vivo normothermic PERfusion for kidney regeneration (PROPER) clinical trial will evaluate the effects of extending NMP duration on patient outcomes (Clinicaltrials.gov ID: NCT04693325). An initial cohort will examine the effects of 1 h NMP, followed by 3 h and then 6 h, with patient outcomes determining progression to each successive timepoint. The primary endpoint is eGFR at 6 months following transplantation
A recent study has reported 24 h NMP with discarded human kidneys [101]. The longer perfusion times were accredited to the use of a perfusion circuit which permits urine recirculation. Conventionally, the urine produced during perfusion is isolated and the volume replaced with a suitable fluid, such as Ringer’s lactate. In this study, the researchers found use of urine recirculation led to a more stable environment with normal electrolyte and acid–base balance compared to kidneys that did not have urine recirculation.

7.5. Assessment of Graft Quality during NMP

NMP offers enhanced capacity to monitor organ quality, with physiological temperatures allowing measures of blood flow, urine production and macroscopic changes detectable with visual inspection [102]. NMP technology has been used to assess kidneys rejected for clinical use and allowed successful transplantation [46].
Transcriptomic signatures measured in samples of kidney tissue after NMP may be able to predict graft outcome. A bioinformatic analysis performed by researchers at Wuhan University Hospital has identified a set of five genes, the level of transcription of which in post-implantation biopsies was predictive for DGF [103]. The authors postulate their genomic model could have value when used in conjunction with genomic data acquired from kidneys undergoing NMP.
This hypothesis has been proposed previously: an apparent DGF-linked gene signature was identified in NMP reconditioned kidneys. In this study, upregulation of components of inflammatory pathways, such as tumour necrosis factor α (TNF-α) and nuclear factor kappa B (NFκB), were linked with extended DGF durations, whereas enhancements of oxidative phosphorylation genes had the opposite effect [51]. This infers observation of the renal transcriptome during NMP may indicate post-transplant outcomes.
The use of final resistance values may also be useful for predicting poor outcomes after NMP [104]. This study reported that between 59% and 70% of transplanted kidneys that had a postoperative GFR of <30 ml/min at 6 months could be predicted by an endpoint resistance of 0.3 mmHg/ml/min during regional NMP.
However, there may be limitations to the use of short NMP durations to predict transplant outcomes [105] and extended durations may prove more prognostic.

8. Summary

Developments in hypothermic and normothermic machine perfusion technologies show potential in clinical kidney transplantation. HMP has been more widely adopted as the primary method of hypothermic preservation and modification of HMP protocols to include oxygen supplementation may or may not be beneficial. Other translational developments in the field of HMP include passive indications of graft quality through observation of perfusion parameters and more specific indications, such as ongoing metabolic processes or biomarker release.
NMP also has the potential to improve kidney transplant outcomes, with better measures of kidney assessment. Additionally, NMP shows great promise in the delivery of therapeutics which can modify the graft during perfusion. While these are currently in experimental stages, it is not inconceivable that advances similar to those discussed in this review could enhance graft compatibility, quality and survival. The advances described here are an indication that the full clinical potential of different machine perfusion strategies as a whole have not yet been realised.

Author Contributions

T.B.S. wrote the manuscript; S.A.H. and M.L.N. reviewed and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The research was funded by the National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Organ Donation and Transplantation at the University of Cambridge in collaboration with Newcastle University and in partnership with NHS Blood and Transplant (NHSBT). The views expressed are those of the author(s) and not necessarily those of the NIHR, the Department of Health and Social Care or NHSBT.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors have no conflict of interest to declare.

Abbreviations

ATPadenosine triphosphate
CITcold ischaemic time
CKDchronic kidney disease
CORcontrolled oxygenated rewarming
DBDdonation after brain death
DCDdonation after circulatory death
DGFdelayed graft function
ECDextended criteria donor
eGFRestimated glomerular filtration rate
ESRDend-stage renal disease
GFRglomerular filtration rate
HBOChaemoglobin-based oxygen carrier
HLAhuman leucocyte antigen
HMPhypothermic machine perfusion
IRIischaemia reperfusion injury
KPS-1kidney perfusion solution 1
MAPCsmultipotent adult progenitor cells
MHCmajor histocompatibility complex
MRImagnetic resonance imaging
MSCsmesenchymal stem cells
NEVKPnormothermic ex vivo kidney perfusion
NFκB nuclear factor kappa B
NGALneutrophil gelatinase-associated lipocalin
NMPnormothermic machine perfusion
NMRnuclear magnetic resonance
NRPnormothermic regional perfusion
PNFprimary non-function
ROSreactive oxygen species
SCSstatic cold storage
TNF-αtumour necrosis factor α
UKUnited Kingdom
USUnited States
WITwarm ischaemic time

References

  1. NHSBT. Fact Sheet 7: Cost-Effectiveness of Transplantation. 2009. Available online: https://nhsbtmediaservices.blob.core.windows.net/organ-donation-assets/pdfs/Organ_Donation_Registry_Fact_Sheet_7_21337.pdf (accessed on 26 September 2021).
  2. Kaballo, M.A.; Canney, M.; O’Kelly, P.; Williams, Y.; O’Seaghdha, C.M.; Conlon, P.J. A comparative analysis of survival of patients on dialysis and after kidney transplantation. Clin. Kidney J. 2018, 11, 389–393. [Google Scholar] [CrossRef]
  3. Santos, A.H., Jr.; Casey, M.J.; Wen, X.; Zendejas, I.; Rehman, S.; Womer, K.L.; Andreoni, K.A. Survival with Dialysis Versus Kidney Transplantation in Adult Hemolytic Uremic Syndrome Patients. Transplantation 2015, 99, 2608–2616. [Google Scholar] [CrossRef]
  4. Wolfe, R.A.; Ashby, V.B.; Milford, E.L.; Ojo, A.O.; Ettenger, R.E.; Agodoa, L.Y.; Held, P.J.; Port, F.K. Comparison of Mortality in All Patients on Dialysis, Patients on Dialysis Awaiting Transplantation, and Recipients of a First Cadaveric Transplant. N. Engl. J. Med. 1999, 341, 1725–1730. [Google Scholar] [CrossRef] [Green Version]
  5. Yoo, K.D.; Kim, C.T.; Kim, M.H.; Noh, J.; Kim, G.; Kim, H.; An, J.N.; Park, J.Y.; Cho, H.; Kim, K.H.; et al. Superior outcomes of kidney transplantation compared with dialysis: An optimal matched analysis of a national population-based cohort study between 2005 and 2008 in Korea. Medicine 2016, 95, e4352. [Google Scholar] [CrossRef]
  6. Caskey, F.; Dawnay, C.C.; Farrington, A.; Fogarty, K.; Kumwenda, F.S.; Macphee, M.; Md, S.; Steenkamp, R.; Aj, W. UK Renal Registry UK Renal Registry 18th Annual Report of the Renal Association. Nephron 2016, 132, 9–40. [Google Scholar]
  7. Zhong, Z.; Hu, Q.; Fu, Z.; Wang, R.; Xiong, Y.; Zhang, Y.; Liu, Z.; Wang, Y.; Ye, Q. Increased Expression of Aldehyde Dehydrogenase 2 Reduces Renal Cell Apoptosis During Ischaemia/Reperfusion Injury After Hypothermic Machine Perfusion. Artif. Organs 2016, 40, 596–603. [Google Scholar] [CrossRef]
  8. Yang, Z.; Zhong, Z.; Li, M.; Xiong, Y.; Wang, Y.; Peng, G.; Ye, Q. Hypothermic machine perfusion increases A20 expression which protects renal cells against Ischaemia/reperfusion injury by suppressing inflammation, apoptosis and necroptosis. Int. J. Mol. Med. 2016, 38, 161–171. [Google Scholar] [CrossRef] [Green Version]
  9. Chatauret, N.; Coudroy, R.; Delpech, P.O.; Vandebrouck, C.; Hosni, S.; Scepi, M.; Hauet, T. Mechanistic analysis of nonoxygenated hypothermic machine perfusion’s protection on warm ischemic kidney uncovers greater enos phosphorylation and vasodilation. Am. J. Transpl. 2014, 14, 2500–2514. [Google Scholar] [CrossRef] [PubMed]
  10. Moers, C.; Smits, J.M.; Maathuis, M.H.J.; Treckmann, J.; van Gelder, F.; Napieralski, B.P.; van Kasterop-Kutz, M.; van der Heide, H.; Squifflet, J.-P.; van Heurn, E.; et al. Machine perfusion or cold storage in deceased-donor kidney transplantation. N. Engl. J. Med. 2009, 360, 7–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Jochmans, I.; Moers, C.; Smits, J.M.; Leuvenink, H.G.; Treckmann, J.; Paul, A.; Rahmel, A.; Squifflet, J.-P.; van Heurn, E.; Monbaliu, D.; et al. Machine Perfusion Versus Cold Storage for the Preservation of Kidneys Donated After Cardiac Death. Ann. Surg. 2010, 252, 756–764. [Google Scholar] [CrossRef] [PubMed]
  12. Moers, C.; Pirenne, J.; Paul, A.; Ploeg, R.J. Machine Preservation Trial Study Group. Machine perfusion or cold storage in deceased-donor kidney transplantation. N. Engl. J. Med. 2012, 366, 770–771. [Google Scholar] [CrossRef] [Green Version]
  13. Bradley, J.A.; Pettigrew, G.J.; Watson, C.J. Time to death after withdrawal of treatment in donation after circulatory death (DCD) donors. Curr. Opin. Organ Transpl. 2013, 18, 133–139. [Google Scholar] [CrossRef] [PubMed]
  14. Van Erp, A.C.; Rebolledo, R.A.; Hoeksma, D.; Jespersen, N.R.; Ottens, P.J.; Nørregaard, R.; Pedersen, M.; Laustsen, C.; Burgerhof, J.G.M.; Wolters, J.C.; et al. Organ-specific responses during brain death: Increased aerobic metabolism in the liver and anaerobic metabolism with decreased perfusion in the kidneys. Sci. Rep. 2018, 8, 4405. [Google Scholar] [CrossRef] [Green Version]
  15. Schiffer, T.A.; Gustafsson, H.; Palm, F. Kidney outer medulla mitochondria are more efficient compared with cortex mitochondria as a strategy to sustain ATP production in a suboptimal environment. Am. J. Physiol. Physiol. 2018, 315, F677–F681. [Google Scholar] [CrossRef] [PubMed]
  16. Peris, A.; Fulceri, G.E.; Lazzeri, C.; Bonizzoli, M.; Li Marzi, V.; Serni, S.; Cirami, L.; Migliaccio, M.L. Delayed graft function and perfusion parameters of kidneys from uncontrolled donors after circulatory death. Perfusion 2021, 36, 299–304. [Google Scholar] [CrossRef] [PubMed]
  17. Kaminski, J.; Delpech, P.O.; Kaaki-Hosni, S.; Promeyrat, X.; Hauet, T.; Hannaert, P. Oxygen Consumption by Warm Ischaemia-Injured Porcine Kidneys in Hypothermic Static and Machine Preservation. J. Surg. Res. 2019, 242, 78–86. [Google Scholar] [CrossRef]
  18. Belzer, F.O.; Southard, J.H. Principles of Solid-Organ Preservation by Cold Storage. Transplantation 1988, 45, 673–676. [Google Scholar] [CrossRef]
  19. Hochachka, P. Defense strategies against hypoxia and hypothermia. Science 1986, 231, 234–241. [Google Scholar] [CrossRef]
  20. Bienholz, A.; Walter, B.; Pless-Petig, G.; Guberina, H.; Kribben, A.; Witzke, O.; Rauen, U. Characterization of injury in isolated rat proximal tubules during cold incubation and rewarming. PLoS ONE 2017, 12, e0180553. [Google Scholar] [CrossRef] [Green Version]
  21. Kellerman, P.S. Exogenous adenosine triphosphate (ATP) preserves proximal tubule microfilament structure and function in vivo in a maleic acid model of ATP depletion. J. Clin. Investig. 1993, 92, 1940–1949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Lieberthal, W.; Menza, S.A.; Levine, J.S. Graded ATP depletion can cause necrosis or apoptosis of cultured mouse proximal tubular cells. Am. J. Physiol. Physiol. 1998, 274, F315–F327. [Google Scholar] [CrossRef] [PubMed]
  23. Pefanis, A.; Ierino, F.L.; Murphy, J.M.; Cowan, P.J. Regulated necrosis in kidney Ischaemia-reperfusion injury. Kidney Int. 2019, 96, 291–301. [Google Scholar] [CrossRef]
  24. Kox, J.; Moers, C.; Monbaliu, D.; Strelniece, A.; Treckmann, J.; Jochmans, I.; Leuvenink, H.; Van Heurn, E.; Pirenne, J.; Paul, A.; et al. The Benefits of Hypothermic Machine Preservation and Short Cold Ischaemia Times in Deceased Donor Kidneys. Transplantation 2018, 102, 1344–1350. [Google Scholar] [CrossRef]
  25. Hendriks, K.D.W.; Brüggenwirth, I.M.A.; Maassen, H.; Gerding, A.; Bakker, B.; Porte, R.J.; Henning, R.H.; Leuvenink, H.G.D. Renal temperature reduction progressively favors mitochondrial ROS production over respiration in hypothermic kidney preservation. J. Transl. Med. 2019, 17, 1–10. [Google Scholar] [CrossRef]
  26. Stubenitsky, B.M.; Ametani, M.; Danielewicz, R.; Southard, J.H.; Belzer, F.O. Regeneration of ATP in kidney slices after warm Ischaemia and hypothermic preservation. Transpl. Int. 1995, 8, 293–297. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, Y.; Shi, J.; Xia, T.C.; Xu, R.; He, X.; Xia, Y. Preservation Solutions for Kidney Transplantation: History, Advances and Mechanisms. Cell Transpl. 2019, 28, 1472. [Google Scholar] [CrossRef] [PubMed]
  28. Hosgood, S.A.; Saeb-Parsy, K.; Wilson, C.; Callaghan, C.; Collett, D.; Nicholson, M.L. Protocol of a randomised controlled, open-label trial of ex vivo normothermic perfusion versus static cold storage in donation after circulatory death renal transplantation. BMJ Open 2017, 7, e012237. [Google Scholar] [CrossRef] [Green Version]
  29. Minor, T.; Horn, C.; Gallinat, A.; Kaths, M.; Kribben, A.; Treckmann, J.; Paul, A. First-in-man controlled rewarming and normothermic perfusion with cell-free solution of a kidney prior to transplantation. Am. J. Transpl. 2020, 20, 1192–1195. [Google Scholar] [CrossRef] [Green Version]
  30. Schlegel, A.; Muller, X.; Mueller, M.; Stepanova, A.; Kron, P.; de Rougemont, O.; Muiesan, P.; Clavien, P.A.; Galkin, A.; Meierhofer, D.; et al. Hypothermic oxygenated perfusion protects from mitochondrial injury before liver transplantation. EBioMedicine 2020, 60, 103014. [Google Scholar] [CrossRef]
  31. Nath, J.; Smith, T.B.; Patel, K.; Ebbs, S.R.; Hollis, A.; Tennant, D.A.; Ludwig, C.; Ready, A.R. Metabolic differences between cold stored and machine perfused porcine kidneys: A 1H NMR based study. Cryobiology 2017, 74, 115–120. [Google Scholar] [CrossRef] [PubMed]
  32. Res, E.S.; Houtzager, J.H.E.; David Hemelrijk, S.; Mirza, I.C.J.H.; Idu, M.; Bemelman, F.J.; Van Gulik, T.M.; Helene, J.; Surgery, E.H. The Use of the Oxygenated AirdriveTM Machine Perfusion System in Kidney Graft Preservation: A Clinical Pilot Study. Eur. Surg. Res. 2020, 61, 153–162. [Google Scholar] [CrossRef]
  33. Ravaioli, M.; Baldassarre, M.; Vasuri, F.; Pasquinelli, G.; Laggetta, M.; Valente, S.; De Pace, V.; Neri, F.; Siniscalchi, A.; Zanfi, C.; et al. Strategies to Restore Adenosine Triphosphate (ATP) Level After More than 20 Hours of Cold Ischaemia Time in Human Marginal Kidney Grafts. Ann. Transpl. 2018, 23, 34–44. [Google Scholar] [CrossRef] [PubMed]
  34. Ravaioli, M.; De Pace, V.; Angeletti, A.; Comai, G.; Vasuri, F.; Baldassarre, M.; Maroni, L.; Odaldi, F.; Fallani, G.; Caraceni, P.; et al. Hypothermic Oxygenated New Machine Perfusion System in Liver and Kidney Transplantation of Extended Criteria Donors:First Italian Clinical Trial. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef] [PubMed]
  35. Patel, K.; Nath, J.; Hodson, J.; Inston, N.; Ready, A. Outcomes of Donation after Circulatory Death kidneys undergoing Hypothermic Machine Perfusion following Static Cold Storage: A UK population-based cohort study. Am. J. Transpl. 2018, 18, 1408–1414. [Google Scholar] [CrossRef] [Green Version]
  36. Bellini, M.I.; Charalampidis, S.; Herbert, P.E.; Bonatsos, V.; Crane, J.; Muthusamy, A.; Dor, F.J.M.F.; Papalois, V. Cold pulsatile machine perfusion versus static cold storage in kidney transplantation: A single centre experience. BioMed Res. Int. 2019, 2019, 7435248. [Google Scholar] [CrossRef] [Green Version]
  37. Savoye, E.; Macher, M.A.; Videcoq, M.; Gatault, P.; Hazzan, M.; Abboud, I.; Thierry, A.; Bertrand, D.; Drouin, S.; Sayegh, J.; et al. Evaluation of outcomes in renal transplantation with hypothermic machine perfusion for the preservation of kidneys from expanded criteria donors. Clin. Transpl. 2019, 33, e13536. [Google Scholar] [CrossRef] [PubMed]
  38. Kruszyna, T.; Richter, P. Hypothermic Machine Perfusion of Kidneys Compensates for Extended Storage Time: A Single Intervention With a Significant Impact. Transpl. Proc. 2021, 53, 1085–1090. [Google Scholar] [CrossRef]
  39. Tedesco Silva, H.; Evans, R.W.; Gavaghan, M.B.; Vazquez, V.C. A Cost-Effectiveness Analysis of Organ Preservation Methods for Deceased Donor Kidneys at High Risk for Delayed Graft Function in Brazil. Transpl. Proc. 2018, 50, 3121–3127. [Google Scholar] [CrossRef]
  40. Martínez Arcos, L.; Fabuel Alcañiz, J.J.; Gómez Dos Santos, V.; Burgos Revilla, F.J. Functional Results of Renal Preservation in Hypothermic Pulsatile Machine Perfusion Versus Cold Preservation: Systematic Review and Meta-Analysis of Clinical Trials. Transpl. Proc. 2018, 50, 24–32. [Google Scholar] [CrossRef]
  41. Ruiz-Hernández, M.; Gómez-Dos Santos, V.; Díaz-Pérez, D.; Fernández-Alcalde, Á.; Hevia-Palacios, V.; Álvarez-Rodríguez, S.; Díez-Nicolás, V.; Elías-Triviño, S.; Burgos-Revilla, F.J. Experience with Hypothermic Machine Perfusion in Expanded Criteria Donors: Functional Outcomes. Transpl. Proc. 2019, 51, 303–306. [Google Scholar] [CrossRef] [PubMed]
  42. Sandal, S.; Luo, X.; Massie, A.B.; Paraskevas, S.; Cantarovich, M.; Segev, D.L. Machine perfusion and long-term kidney transplant recipient outcomes across allograft risk strata. Nephrol. Dial. Transpl. 2018, 33, 1251–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Rijkse, E.; IJzermans, J.N.; Minnee, R.C. Machine perfusion in abdominal organ transplantation: Current use in the Netherlands. World J. Transpl. 2020, 10, 15–28. [Google Scholar] [CrossRef]
  44. Wijk, J.; Slooff, M.J.; Rijkmans, B.G.; Kootstra, G. Successful 96- and 144-hour experimental kidney preservation: A combination of standard machine preservation and newly developed normothermic ex vivo perfusion. Cryobiology 1980, 17, 473–477. [Google Scholar] [CrossRef]
  45. Hosgood, S.A.; Nicholson, M.L. First in man renal transplantation after ex vivo normothermic perfusion. Transplantation 2011, 92, 735–738. [Google Scholar] [CrossRef] [PubMed]
  46. Hosgood, S.A.; Thompson, E.; Moore, T.; Wilson, C.H.; Nicholson, M.L. Normothermic machine perfusion for the assessment and transplantation of declined human kidneys from donation after circulatory death donors. Br. J. Surg. 2018, 105, 388–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Hosgood, S.A.; Saeb-Parsy, K.; Hamed, M.O.; Nicholson, M.L. Successful Transplantation of Human Kidneys Deemed Untransplantable but Resuscitated by Ex Vivo Normothermic Machine Perfusion. Am. J. Transpl. 2016, 16, 3282. [Google Scholar] [CrossRef] [Green Version]
  48. Schopp, I.; Reissberg, E.; Lüer, B.; Efferz, P.; Minor, T. Controlled Rewarming after Hypothermia: Adding a New Principle to Renal Preservation. Clin. Transl. Sci. 2015, 8, 475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. von Horn, C.; Zlatev, H.; Kaths, M.; Andreas, P.; Minor, T. Controlled oxygenated rewarming compensates for cold storage-induced dysfunction in kidney grafts. Transplantation 2021. published online ahead of print. [Google Scholar] [CrossRef]
  50. Elliott, T.R.; Nicholson, M.L.; Hosgood, S.A. Normothermic kidney perfusion: An overview of protocols and strategies. Am. J. Transpl. 2021, 21, 1382–1390. [Google Scholar] [CrossRef]
  51. Ferdinand, J.R.; Hosgood, S.A.; Moore, T.; Ferro, A.; Ward, C.J.; Castro-Dopico, T.; Nicholson, M.L.; Clatworthy, M.R. Cytokine absorption during human kidney perfusion reduces delayed graft function–associated inflammatory gene signature. Am. J. Transpl. 2021, 21, 2188–2199. [Google Scholar] [CrossRef]
  52. Hameed, A.M.; Miraziz, R.; Lu, D.B.; Warwick, N.; El-Ayoubi, A.; Burns, H.; Chew, Y.V.; Matthews, R.; O’Grady, G.; Yuen, L.; et al. Extra-corporeal normothermic machine perfusion of the porcine kidney: Working towards future utilization in Australasia. ANZ J. Surg. 2018, 88, E429–E434. [Google Scholar] [CrossRef] [PubMed]
  53. Bellini, M.I.; Yiu, J.; Nozdrin, M.; Papalois, V. The Effect of Preservation Temperature on Liver, Kidney, and Pancreas Tissue ATP in Animal and Preclinical Human Models. J. Clin. Med. 2019, 8, 1421. [Google Scholar] [CrossRef] [Green Version]
  54. Tejchman, K.; Sierocka, A.; Kotowski, M.; Zair, L.; Pilichowska, E.; Ostrowski, M.; Sieńko, J. Acid-Base Balance Disorders During Kidney Preservation in Cold Ischemia. Transpl. Proc. 2020, 52, 2036–2042. [Google Scholar] [CrossRef] [PubMed]
  55. Bunegin, L.; Tolstykh, G.P.; Gelineau, J.F.; Cosimi, A.B.; Anderson, L.M. Oxygen Consumption During Oxygenated Hypothermic Perfusion as a Measure of Donor Organ Viability. ASAIO J. 2013, 59, 427–432. [Google Scholar] [CrossRef] [PubMed]
  56. Hoyer, D.P.; Gallinat, A.; Swoboda, S.; Wohlschlaeger, J.; Rauen, U.; Paul, A.; Minor, T. Influence of oxygen concentration during hypothermic machine perfusion on porcine kidneys from donation after circulatory death. Transplantation 2014, 98, 944–950. [Google Scholar] [CrossRef]
  57. Jochmans, I.; Brat, A.; Davies, L.; Hofker, H.S.; van de Leemkolk, F.E.M.; Leuvenink, H.G.D.; Knight, S.R.; Pirenne, J.; Ploeg, R.J. Oxygenated versus standard cold perfusion preservation in kidney transplantation (COMPARE): A randomised, double-blind, paired, phase 3 trial. Lancet 2020, 396, 1653–1662. [Google Scholar] [CrossRef]
  58. Patel, K.; Smith, T.B.; Neil, D.A.H.; Thakker, A.; Tsuchiya, Y.; Higgs, E.B.; Hodges, N.J.; Ready, A.R.; Nath, J.; Ludwig, C. The Effects of Oxygenation on Ex Vivo Kidneys Undergoing Hypothermic Machine Perfusion. Transplantation 2019, 103, 314–322. [Google Scholar] [CrossRef]
  59. Venema, L.H.; Brat, A.; Moers, C.; Hart, N.A.; Ploeg, R.J.; Hannaert, P.; Minor, T.; Leuvenink, A.H.G.D. Effects of Oxygen During Long-term Hypothermic Machine Perfusion in a Porcine Model of Kidney Donation After Circulatory Death. Transplantation 2019, 103, 2057–2064. [Google Scholar] [CrossRef] [Green Version]
  60. Fuller, B.J.; Lee, C.Y. Hypothermic perfusion preservation: The future of organ preservation revisited? Cryobiology 2007, 54, 129–145. [Google Scholar] [CrossRef] [PubMed]
  61. Darius, T.; Gianello, P.; Vergauwen, M.; Mourad, N.; Buemi, A.; De Meyer, M.; Mourad, M. The effect on early renal function of various dynamic preservation strategies in a preclinical pig Ischaemia–reperfusion autotransplant model. Am. J. Transpl. 2019, 19, 752–762. [Google Scholar] [CrossRef]
  62. Darius, T.; Vergauwen, M.; Smith, T.B.; Patel, K.; Craps, J.; Joris, V.; Aydin, S.; Ury, B.; Buemi, A.; De Meyer, M.; et al. Influence of Different Partial Pressures of Oxygen during Continuous Hypothermic Machine Perfusion in a Pig Kidney Ischaemia-reperfusion Autotransplant Model. Transplantation 2020, 104, 731–743. [Google Scholar] [CrossRef] [PubMed]
  63. Darius, T.; Vergauwen, M.; Smith, T.; Gerin, I.; Joris, V.; Mueller, M.; Aydin, S.; Muller, X.; Schlegel, A.; Nath, J.; et al. Brief O2 uploading during continuous hypothermic machine perfusion is simple yet effective oxygenation method to improve initial kidney function in a porcine autotransplant model. Am. J. Transpl. 2020, 20, 2030–2043. [Google Scholar] [CrossRef]
  64. Darius, T.; Vergauwen, M.; Mueller, M.; Aydin, S.; Dutkowski, P.; Gianello, P.; Mourad, M. Brief Bubble and Intermittent Surface Oxygenation Is a Simple and Effective Alternative for Membrane Oxygenation during Hypothermic Machine Perfusion in Kidneys. Transpl. Direct 2020, 6, e571. [Google Scholar] [CrossRef]
  65. Meister, F.A.; Czigany, Z.; Rietzler, K.; Miller, H.; Reichelt, S.; Liu, W.J.; Boecker, J.; Moeller, M.J.; Tolba, R.H.; Hamesch, K.; et al. Decrease of renal resistance during hypothermic oxygenated machine perfusion is associated with early allograft function in extended criteria donation kidney transplantation. Sci. Rep. 2020, 10, 1–10. [Google Scholar] [CrossRef] [PubMed]
  66. Meister, F.A.; Czigany, Z.; Bednarsch, J.; Boecker, J.; Wiltberger, G.; Rohlfs, W.; Neumann, U.P.; Lurje, G. Hypothermic oxygenated machine perfusion—Preliminary experience with end-ischemic reconditioning of marginal kidney allografts. Clin. Transpl. 2019, 33, e13673. [Google Scholar] [CrossRef] [PubMed]
  67. Kasil, A.; Giraud, S.; Couturier, P.; Amiri, A.; Danion, J.; Donatini, G.; Matillon, X.; Hauet, T.; Badet, L. Individual and Combined Impact of Oxygen and Oxygen Transporter Supplementation during Kidney Machine Preservation in a Porcine Preclinical Kidney Transplantation Model. Int. J. Mol. Sci. 2019, 20, 1992. [Google Scholar] [CrossRef] [Green Version]
  68. Le Meur, Y.; Badet, L.; Essig, M.; Thierry, A.; Büchler, M.; Drouin, S.; Deruelle, C.; Morelon, E.; Pesteil, F.; Delpech, P.; et al. First-in-human use of a marine oxygen carrier (M101) for organ preservation: A safety and proof-of-principle study. Am. J. Transpl. 2020, 20, 1729–1738. [Google Scholar] [CrossRef]
  69. Soussi, D.; Danion, J.; Baulier, E.; Favreau, F.; Sauvageon, Y.; Bossard, V.; Matillon, X.; Turpin, F.; Belgsir, E.M.; Thuillier, R.; et al. Vectisol formulation enhances solubility of resveratrol and brings its benefits to kidney transplantation in a preclinical porcine model. Int. J. Mol. Sci. 2019, 20, 2268. [Google Scholar] [CrossRef] [Green Version]
  70. Sedigh, A.; Larsson, R.; Brännström, J.; Magnusson, P.; Larsson, E.; Tufveson, G.; Lorant, T. Modifying the vessel walls in porcine kidneys during machine perfusion. J. Surg. Res. 2014, 191, 455–462. [Google Scholar] [CrossRef] [PubMed]
  71. Sedigh, A.; Nordling, S.; Carlsson, F.; Larsson, E.; Norlin, B.; Lübenow, N.; Lennmyr, F.; Tufveson, G.; Magnusson, P.U.; Lorant, T. Perfusion of Porcine Kidneys with Macromolecular Heparin Reduces Early Ischaemia Reperfusion Injury. Transplantation 2019, 103, 420–427. [Google Scholar] [CrossRef]
  72. Tierie, E.L.; Roodnat, J.I.; Dor, F.J.M.F. Systematic Surgical Assessment of Deceased-Donor Kidneys as a Predictor of Short-Term Transplant Outcomes. Eur. Surg. Res. 2019, 60, 97–105. [Google Scholar] [CrossRef] [PubMed]
  73. Bissolati, M.; Gazzetta, P.G.; Caldara, R.; Guarneri, G.; Adamenko, O.; Giannone, F.; Mazza, M.; Maggi, G.; Tomanin, D.; Rosati, R.; et al. Renal Resistance Trend During Hypothermic Machine Perfusion Is More Predictive of Postoperative Outcome Than Biopsy Score: Preliminary Experience in 35 Consecutive Kidney Transplantations. Artif. Organs 2018, 42, 714–722. [Google Scholar] [CrossRef]
  74. Chen, G.; Wang, C.; Zhao, Y.; Qiu, L.; Yuan, X.; Qiu, J.; Wang, C.; He, X.; Chen, L. Evaluation of quality of kidneys from donation after circulatory death/expanded criteria donors by parameters of machine perfusion. Nephrology 2018, 23, 103–106. [Google Scholar] [CrossRef]
  75. Tai, Q.; Xue, W.; Ding, X.; Tian, P.; Xiang, H.; Feng, X.; Yan, H.; Hou, J. Perfusion Parameters of Donation After Cardiac Death Kidneys Predict Early Transplant Outcomes Based on Expanded Criteria Donor Designation. Transpl. Proc. 2018, 50, 79–84. [Google Scholar] [CrossRef]
  76. Sandal, S.; Paraskevas, S.; Cantarovich, M.; Baran, D.; Chaudhury, P.; Tchervenkov, J.I.; Sapir-Pichhadze, R. Renal resistance thresholds during hypothermic machine perfusion and transplantation outcomes—A retrospective cohort study. Transpl. Int. 2018, 31, 658–669. [Google Scholar] [CrossRef] [Green Version]
  77. Wszola, M.; Domagala, P.; Serwanska-Swietek, M.; Ostaszewska, A.; Perkowska-Ptasinska, A.; Piatek, T.; Gozdowska, J.; Durlik, M.; Chmura, A.; Kwiatkowski, A. Should Immunosuppression After Kidney Transplant Be Adjusted Based on Renal Resistance during Pretransplant Hypothermic Machine Perfusion? Transpl. Proc. 2019, 51, 2676–2682. [Google Scholar] [CrossRef]
  78. Jochmans, I.; Moers, C.; Smits, J.M.; Leuvenink, H.D.; Treckman, J.; Paul, A.; Rahmel, A.; Squifflet, J.P.; Heurn, E.; Monbaliu, D.; et al. The prognostic value of renal resistance during hypothermic machine perfusion of deceased donor kidneys. Am. J. Transpl. 2011, 11, 2214–2220. [Google Scholar] [CrossRef]
  79. Longchamp, A.; Klauser, A.; Songeon, J.; Agius, T.; Nastasi, A.; Ruttiman, R.; Moll, S.; Meier, R.P.H.; Buhler, L.; Corpataux, J.M.; et al. Ex Vivo Analysis of Kidney Graft Viability Using 31P Magnetic Resonance Imaging Spectroscopy. Transplantation 2020, 104, 1825–1831. [Google Scholar] [CrossRef] [PubMed]
  80. Konkel, B.; Lavin, C.; Wu, T.T.; Anderson, E.; Iwamoto, A.; Rashid, H.; Gaitian, B.; Boone, J.; Cooper, M.; Abrams, P.; et al. Fully automated analysis of OCT imaging of human kidneys for prediction of post-transplant function. Biomed. Opt. Express 2019, 10, 1794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Bon, D.; Claire, B.; Thuillier, R.; Hebrard, W.; Boildieu, N.; Celhay, O.; Irani, J.; Seguin, F.; Hauet, T. Analysis of Perfusates during Hypothermic Machine Perfusion by NMR Spectroscopy. Transplantation 2014, 97, 810–816. [Google Scholar] [CrossRef]
  82. Gowers, S.A.N.; Hamaoui, K.; Vallant, N.; Hanna, G.B.; Darzi, A.; Casanova, D.; Papalois, V.; Boutelle, M.G. An improved rapid sampling microdialysis system for human and porcine organ monitoring in a hospital setting. Anal. Methods 2018, 10, 5273–5281. [Google Scholar] [CrossRef] [Green Version]
  83. Patel, K.; Smith, T.; Thakker, A.; Inston, N.; Ready, A.; Ludwig, C.; Nath, J. Abstracts of the 19th Congress of the European Society for Organ Transplantation, 15–18 September 2019, Copenhagen, Denmark. Transpl. Int. 2019, 32, 5–433. [Google Scholar]
  84. Moers, C.; Varnav, O.C.; Van Heurn, E.; Jochmans, I.; Kirste, G.R.; Rahmel, A.; Leuvenink, H.G.D.; Squifflet, J.P.; Paul, A.; Pirenne, J.; et al. The value of machine perfusion perfusate biomarkers for predicting kidney transplant outcome. Transplantation 2010, 90, 966–973. [Google Scholar] [CrossRef] [Green Version]
  85. Parikh, C.R.; Hall, I.E.; Bhangoo, R.S.; Ficek, J.; Abt, P.L.; Thissen-Philbrook, H.; Lin, H.; Bimali, M.; Murray, P.T.; Rao, V.; et al. Associations of Perfusate Biomarkers and Pump Parameters with Delayed Graft Function and Deceased Donor Kidney Allograft Function. Am. J. Transpl. 2016, 16, 1526–1539. [Google Scholar] [CrossRef] [PubMed]
  86. Guzzi, F.; Knight, S.R.; Ploeg, R.J.; Hunter, J.P. A systematic review to identify whether perfusate biomarkers produced during hypothermic machine perfusion can predict graft outcomes in kidney transplantation. Transpl. Int. 2020, 33, 590–602. [Google Scholar] [CrossRef] [PubMed]
  87. He, X.; Chen, G.; Zhu, Z.; Zhang, Z.; Yuan, X.; Han, M.; Zhao, Q.; Zheng, Y.; Tang, Y.; Huang, S.; et al. The First Case of Ischaemia-Free Kidney Transplantation in Humans. Front. Med. 2019, 6, 276. [Google Scholar] [CrossRef] [Green Version]
  88. Antoine, C.; Savoye, E.; Gaudez, F.; Cheisson, G.; Badet, L.; Videcoq, M.; Legeai, C.; Bastien, O.; Barrou, B. Kidney transplant from uncontrolled donation after circulatory death: Contribution of normothermic regional perfusion. Transplantation 2019, 104, 130–136. [Google Scholar] [CrossRef] [PubMed]
  89. Kerforne, T.; Allain, G.; Giraud, S.; Bon, D.; Ameteau, V.; Couturier, P.; Hebrard, W.; Danion, J.; Goujon, J.M.; Thuillier, R.; et al. Defining the optimal duration for normothermic regional perfusion in the kidney donor: A porcine preclinical study. Am. J. Transpl. 2019, 19, 737–751. [Google Scholar] [CrossRef] [PubMed]
  90. Aburawi, M.M.; Fontan, F.M.; Karimian, N.; Eymard, C.; Cronin, S.; Pendexter, C.; Nagpal, S.; Banik, P.; Ozer, S.; Mahboub, P.; et al. Synthetic hemoglobin-based oxygen carriers are an acceptable alternative for packed red blood cells in normothermic kidney perfusion. Am. J. Transpl. 2019, 19, 2814–2824. [Google Scholar] [CrossRef]
  91. Minor, T.; von Horn, C.; Paul, A. Role of erythrocytes in short-term rewarming kidney perfusion after cold storage. Artif. Organs 2019, 43, 584–592. [Google Scholar] [CrossRef] [PubMed]
  92. Maassen, H.; Hendriks, K.D.W.; Venema, L.H.; Henning, R.H.; Hofker, S.H.; van Goor, H.; Leuvenink, H.G.D.; Coester, A.M. Hydrogen sulphide-induced hypometabolism in human-sized porcine kidneys. PLoS ONE 2019, 14, e0225152. [Google Scholar] [CrossRef]
  93. Lohmann, S.; Pool, M.; Rozenberg, K.; Keller, A.; Moers, C.; Møldrup, U.; Møller, B.; Lignell, S.; Krag, S.; Sierra-Parraga, J.; et al. Mesenchymal stromal cell treatment of donor kidneys during ex-vivo normothermic machine perfusion: A porcine renal autotransplantation study. Am. J. Transpl. 2021, 21, 2348–2359. [Google Scholar] [CrossRef]
  94. Pool, M.B.F.; Vos, J.; Eijken, M.; Van Pel, M.; Reinders, M.E.J.; Ploeg, R.J.; Hoogduijn, M.J.; Jespersen, B.; Leuvenink, H.G.D.; Moers, C. Treating Ischemically Damaged Porcine Kidneys with Human Bone Marrow—And Adipose Tissue-Derived Mesenchymal Stromal Cells during Ex Vivo Normothermic Machine Perfusion. Stem Cells Dev. 2020, 29, 1320–1330. [Google Scholar] [CrossRef]
  95. Thompson, E.R.; Bates, L.; Ibrahim, I.K.; Sewpaul, A.; Stenberg, B.; McNeill, A.; Figueiredo, R.; Girdlestone, T.; Wilkins, G.C.; Wang, L.; et al. Novel delivery of cellular therapy to reduce Ischaemia reperfusion injury in kidney transplantation. Am. J. Transpl. 2021, 21, 1402–1414. [Google Scholar] [CrossRef]
  96. Brasile, L.; Henry, N.; Orlando, G.; Stubenitsky, B. Potentiating Renal Regeneration using Mesenchymal Stem Cells. Transplantation 2019, 103, 307. [Google Scholar] [CrossRef] [PubMed]
  97. Hameed, A.M.; Lu, D.B.; Burns, H.; Byrne, N.; Chew, Y.V.; Julovi, S.; Ghimire, K.; Zanjani, N.T.; P’ng, C.H.; Meijles, D.; et al. Pharmacologic targeting of renal Ischaemia-reperfusion injury using a normothermic machine perfusion platform. Sci. Rep. 2020, 10, 1–17. [Google Scholar] [CrossRef]
  98. DiRito, J.R.; Hosgood, S.A.; Reschke, M.; Albert, C.; Bracaglia, L.G.; Ferdinand, J.R.; Stewart, B.J.; Edwards, C.M.; Vaish, A.G.; Thiru, S.; et al. Lysis of cold-storage-induced microvascular obstructions for ex vivo revitalization of marginal human kidneys. Am. J. Transpl. 2021, 21, 161–173. [Google Scholar] [CrossRef] [PubMed]
  99. Yuzefovych, Y.; Valdivia, E.; Rong, S.; Hack, F.; Rother, T.; Schmitz, J.; Bräsen, J.H.; Wedekind, D.; Moers, C.; Wenzel, N.; et al. Genetic Engineering of the Kidney to Permanently Silence MHC Transcripts During ex vivo Organ Perfusion. Front. Immunol. 2020, 11, 265. [Google Scholar] [CrossRef] [Green Version]
  100. Rijkse, E.; De Jonge, J.; Kimenai, H.J.A.N.; Hoogduijn, M.J.; De Bruin, R.W.F.; Van Den Hoogen, M.W.F.; Ijzermans, J.N.M.; Minnee, R.C. Safety and feasibility of 2 h of normothermic machine perfusion of donor kidneys in the Eurotransplant Senior Program. BJS Open 2021, 5, zraa024. [Google Scholar] [CrossRef] [PubMed]
  101. Weissenbacher, A.; Lo Faro, L.; Boubriak, O.; Soares, M.F.; Roberts, I.S.; Hunter, J.P.; Voyce, D.; Mikov, N.; Cook, A.; Ploeg, R.J.; et al. Twenty-four–hour normothermic perfusion of discarded human kidneys with urine recirculation. Am. J. Transpl. 2019, 19, 178–192. [Google Scholar] [CrossRef]
  102. Hosgood, S.A.; Barlow, A.D.; Hunter, J.P.; Nicholson, M.L. Ex vivo normothermic perfusion for quality assessment of marginal donor kidney transplants. Br. J. Surg. 2015, 102, 1433–1440. [Google Scholar] [CrossRef] [PubMed]
  103. Yu, B.; Liang, H.; Zhou, S.; Ye, Q.; Wang, Y. A novel genomic model for predicting the likelihood of delayed graft function in DCD kidney transplantation. Transl. Androl. Urol. 2021, 10, 1637–1646. [Google Scholar] [CrossRef] [PubMed]
  104. Gelpi, R.; Paredes, D.; Rodríguez-Villar, C.; Roque, R.; Ruiz, A.; Adalia, R.; Peri-Cusí, L.; Sole, M.; Oppenheimer, F.; Diekmann, F. The development of a predictive model of graft function in uncontrolled donors after circulatory death: Validity of a pulsatile renal preservation machine cut-off value for kidney acceptance. Nephrol. Dial. Transpl. 2019, 34, 531–538. [Google Scholar] [CrossRef]
  105. Hosgood, S.A.; Nicholson, M.L. A Short Period of Normothermic Machine Perfusion May Not Be Able to Predict Primary Nonfunction in Uncontrolled Circulatory Death Kidneys. Transplantation 2021, 105, E11–E12. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the changing graft tissue environment between organ donation and implantation. In health, homeostatic mechanisms ensure sufficient oxygen and nutrients are delivered to the renal tissue, resulting in balance between adenosine triphosphate (ATP) usage and regeneration. After donation, cessation of blood flow halts oxygen and nutrient supply (causing warm ischaemia). This causes an anaerobic switch that results in ATP depletion and accumulation of harmful metabolic by-products such as reactive oxygen species (ROS) and lactic acid. Cold ischaemia (chilling the organ) is deliberately implemented to slow the ATP depletion and damage that would occur under warm ischaemia. Restoration of blood flow drives ATP regeneration, but leads to another insult (ischaemia reperfusion injury) which occurs as a consequence of deleterious processes initiated by warm and cold ischaemia.
Figure 1. Overview of the changing graft tissue environment between organ donation and implantation. In health, homeostatic mechanisms ensure sufficient oxygen and nutrients are delivered to the renal tissue, resulting in balance between adenosine triphosphate (ATP) usage and regeneration. After donation, cessation of blood flow halts oxygen and nutrient supply (causing warm ischaemia). This causes an anaerobic switch that results in ATP depletion and accumulation of harmful metabolic by-products such as reactive oxygen species (ROS) and lactic acid. Cold ischaemia (chilling the organ) is deliberately implemented to slow the ATP depletion and damage that would occur under warm ischaemia. Restoration of blood flow drives ATP regeneration, but leads to another insult (ischaemia reperfusion injury) which occurs as a consequence of deleterious processes initiated by warm and cold ischaemia.
Transplantology 02 00044 g001
Table 1. Constituents of kidney preservation solutions in clinical use.
Table 1. Constituents of kidney preservation solutions in clinical use.
SCS FluidsHMP FluidsNMP Fluids
University of Wisconsin (UW) solutionCustodial-N solutionUW Machine perfusion solution (UWPS)Hosgood protocol [28]Minor protocol [29]
Base fluidWaterWaterWaterRinger’s solutionSteen solution
Ringer’s solution
Volume expanders/osmotic agentsHydroxyethyl starch
Raffinose pentahydrate
MannitolHydroxyethyl starch
Mannitol (USP)
Magnesium gluconate
Sodium gluconate
MannitolCalcium gluconate
Oxygen carriers---1 unit red blood cells (group O)-
DrugsAllopurinol
Magnesium sulphate heptahydrate
Lactobionic acid
Deferoxamine Dexamethasone
Heparin
Prostacyclin
Insulin
Ampicillin
AntioxidantsGlutathioneTryptophanGlutathione
Metabolic
support
AdenosinePotassium hydrogen 2-ketoglutarate
Sucrose
Aspartate
Arginine
Alanine
Glycine
Glucose, beta D (+)
Ribose
Glucose, beta D (+)
Synthamin 17
Cernevit multivitamins
-
Individual
electrolyte
additives
-Magnesium chloride
Calcium chloride
Potassium chloride
Sodium chloride
Calcium chloride--
Buffering agentsPotassium dihydrogen phosphateHistidine
Histidine · HCI
HEPES (free acid)
Potassium phosphate (monobasic)
Sodium bicarbonateSodium bicarbonate
pH adjustmentSodium hydroxide/hydrochloric acid
Potassium hydroxide
-Sodium hydroxide--
Table 2. Recent clinical trials optimising pretransplant kidney storage and machine perfusion protocols.
Table 2. Recent clinical trials optimising pretransplant kidney storage and machine perfusion protocols.
NMP Clinical Trials
NCT NumberTitlePrimary Outcome MeasureStart Date Completion Date
NCT05031052Normothermic machine perfusion (NMP) vs Static Cold Storage (SCS) in Human Kidney transplantationKidney function at 6 months post-transplant (eGFR)August 2021December 2025
NCT04882254Normothermic Machine Perfusion: An Additional Value for Kidney Transplant Outcomes?Number of patients with immediate graft function within three months post-transplantMay 2021February 2023
NCT03136848The Feasibility and Safety of Normothermic ex Vivo Kidney Perfusion
  • The ratio of actual/eligible kidney grafts subjected to study intervention at three months after enrolment or up to 4 years whichever is earlier
  • The rate of kidney discard or graft failure attributable to the study intervention from the date of first actual intervention to the date the last participant completes the study follow up period of 3 months post-intervention.
December 2016April 2019
NCT04693325PROlonged Ex-vivo Normothermic Machine PERfusion for Kidney RegenerationGlomerular filtration rate (GFR) at: 6 months post-transplantationFebruary 2021July 2022
NCT02525510Deceased Organ Donor Interventions to Protect Kidney Graft FunctionDelayed Graft Function incidence within 1 week of transplantationAugust 2017March 2022
ISRCTN15821205Ex Vivo Normothermic machine perfusion Trial Delayed Graft Function incidence within 1 week of transplantationJanuary 2017-
HMP Clinical trials
NCT NumberTitlePrimary outcome measureStart dateCompletion Date
NCT04619732Real-time Monitoring of Kidney Grafts on Hypothermic Machine PerfusionPost-operative recovery of kidney function within: 30 days of transplantJune 2021December 2021
NCT03378817Hypothermic Oxygenated Machine Perfusion of Extended Criteria Kidney Allografts from Brain Death DonorsDelayed Graft Function incidence within 1 week of transplantationDecember 2017March 2020
NCT03031067Hypothermic Oxygenated Perfusion Versus Static Cold Storage for Marginal GraftGraft function at 3 months post-transplantationOctober 2016 February 2018
NCT04359173Propensity Score Matched Comparison of HMP vs. SCS in Kidney TransplantationDelayed Graft Function incidence within 1 week of transplantationAugust 2015March 2020
NCT02055950Pulsed Perfusion for Marginal Kidneys
  • Glomerular filtration rate (GFR) at 6 months post-transplant
  • Renal resistance at 6 hours after pulsatile machine perfusion
July 2013August 2018
NCT03837197Clinical Trial of New Hypothermic Oxygenated Perfusion System Versus Static Cold StorageDelayed Graft Function incidence within 0–30 days of transplantationDecember 2018December 2021
NCT02876692Prediction and Management of Delayed Graft Function Based on Donor Criteria and LifePort Platform
  • Delayed Graft Function incidence within 1 week of transplantation
  • Transplant nephrectomy at 1 year
January 2016December 2019
NCT02652520Evaluation of a Marine OXYgen Carrier: HEMO2Life for hypOthermic Kidney Graft Preservation, Before Transplantation (OXYOP)Charting within three months of transplant:
  • HEMO2Life adverse effects
  • Graft safety
  • Recipient safety (any adverse event)
March 2016February 2018
NCT03773211Renaparin in Kidney TransplantationAdverse events within 30 daysFebruary 20191 April 2020
NCT03024229Metabolomics in Assessing the Quality of Kidney Transplants Retained on a LifePort Perfusion MachineImmediate graft function (IGF) ( i.e. the absence of a requirement for dialysis) within 7 days post-transplantMarch 2017January 2020
NCT01848249Deceased Donor Biomarkers and Recipient Outcomes
  • Delayed Graft Function incidence within 1 week of transplantation.
  • Death-Censored Graft Failure within 4 years post-transplant.
May 2010March 2020
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Smith, T.B.; Nicholson, M.L.; Hosgood, S.A. Advances in Hypothermic and Normothermic Perfusion in Kidney Transplantation. Transplantology 2021, 2, 460-477. https://doi.org/10.3390/transplantology2040044

AMA Style

Smith TB, Nicholson ML, Hosgood SA. Advances in Hypothermic and Normothermic Perfusion in Kidney Transplantation. Transplantology. 2021; 2(4):460-477. https://doi.org/10.3390/transplantology2040044

Chicago/Turabian Style

Smith, Thomas B., Michael L. Nicholson, and Sarah A. Hosgood. 2021. "Advances in Hypothermic and Normothermic Perfusion in Kidney Transplantation" Transplantology 2, no. 4: 460-477. https://doi.org/10.3390/transplantology2040044

Article Metrics

Back to TopTop