Next Article in Journal
Beyond the Gut: A Systematic Review of Oral Manifestations in Celiac Disease
Next Article in Special Issue
How to Preserve Steatotic Liver Grafts for Transplantation
Previous Article in Journal
Influence of Sociodemographic, Premorbid, and Injury-Related Factors on Post-Traumatic Stress, Anxiety, and Depression after Traumatic Brain Injury
Previous Article in Special Issue
Trends and Obstacles to Implement Dynamic Perfusion Concepts for Clinical Liver Transplantation: Results from a Global Web-Based Survey
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Outcomes of Kidney Perfusion Techniques in Transplantation from Deceased Donors: A Systematic Review and Meta-Analysis

by
Ahmed S. Ghoneima
1,†,
Richard X. Sousa Da Silva
2,†,
Martina A. Gosteli
3,
Adam D. Barlow
1 and
Philipp Kron
1,2,*
1
Department of HPB and Transplant Surgery, St. James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
2
Swiss HPB and Transplantation Center, Department of Surgery and Transplantation, University Hospital Zurich, 8091 Zurich, Switzerland
3
University Library, University of Zurich, 8006 Zurich, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2023, 12(12), 3871; https://doi.org/10.3390/jcm12123871
Submission received: 14 March 2023 / Revised: 3 May 2023 / Accepted: 9 May 2023 / Published: 6 June 2023

Abstract

:
The high demand for organs in kidney transplantation and the expansion of the donor pool have led to the widespread implementation of machine perfusion technologies. In this study, we aim to provide an up-to-date systematic review of the developments in this expanding field over the past 10 years, with the aim of answering the question: “which perfusion technique is the most promising technique in kidney transplantation?” A systematic review of the literature related to machine perfusion in kidney transplantation was performed. The primary outcome measure was delayed graft function (DGF), and secondary outcomes included rates of rejection, graft survival, and patient survival rates after 1 year. Based on the available data, a meta-analysis was performed. The results were compared with data from static cold storage, which is still the standard of care in many centers worldwide. A total of 56 studies conducted in humans were included, and 43 studies reported outcomes of hypothermic machine perfusion (HMP), with a DGF rate of 26.4%. A meta-analysis of 16 studies showed significantly lower DGF rates in the HMP group compared to those of static cold storage (SCS). Five studies reported outcomes of hypothermic machine perfusion + O2, with an overall DGF rate of 29.7%. Two studies explored normothermic machine perfusion (NMP). These were pilot studies, designed to assess the feasibility of this perfusion approach in the clinical setting. Six studies reported outcomes of normothermic regional perfusion (NRP). The overall incidence of DGF was 71.5%, as it was primarily used in uncontrolled DCD (Maastricht category I-II). Three studies comparing NRP to in situ cold perfusion showed a significantly lower rate of DGF with NRP. The systematic review and meta-analysis provide evidence that dynamic preservation strategies can improve outcomes following kidney transplantation. More recent approaches such as normothermic machine perfusion and hypothermic machine perfusion + O2 do show promising results but need further results from the clinical setting. This study shows that the implementation of perfusion strategies could play an important role in safely expanding the donor pool.

1. Introduction

Kidney transplantation remains the therapy of choice for patients with end-stage renal disease and is known to improve quality of life and life expectancy [1,2,3]. However, the number of patients waiting for a kidney graft continues to increase and far exceeds the availability of donor grafts. This has pushed transplant centers worldwide to expand their deceased donor pools by accepting grafts from less-ideal donors such as extended criteria donors (ECD), donors after circulatory death (DCD), and, in some countries, even uncontrolled DCD grafts (uDCD, Maastricht I–II) [4]. The results from these organs showed inferior outcomes compared to those of donation after brain death (DBD) [5,6,7]. This is due to the organ damage caused by hypoxia during a period of ischemia at body temperature (also known as warm ischemia time) between the time of asystole and cold perfusion. The effects of warm ischemia time are even more pronounced in uncontrolled DCD (uDCD). The utilization of DCD and marginal organs has created a need for new technologies of organ preservation, assessment, and optimization, moving from a static environment to dynamic perfusion strategies.
The standard method of organ preservation, since the 1970s, has been static cold storage (SCS), where organs are packed and placed on ice in an insulated container using the hypothermic environment to reduce the metabolic rate at a cellular level [8]. However, the need for acceptance of borderline grafts led to the development and introduction of dynamic perfusion strategies, with the aim of assessing, reconditioning, and predicting post-transplant outcomes. There is a variety of approaches to organ perfusion, mainly differing in temperature, the perfusion solution, the use of supplemental oxygen, and the timing of perfusion.
Hypothermic machine perfusion is a technique, developed in 1967 by Belzer, where the kidney is connected to a perfusion circuit in which cooled preservation solution flows through the organ with the aid of a pump [8,9]. This method benefits from the reduction in cellular metabolism, down to 5–10%, triggered by the low temperatures. Furthermore, hemodynamic stimulus is maintained, which improves renal cortical microcirculation during the preservation period [8]. Hypothermic machine perfusion without supplemental oxygen (HMP) has been based on this concept for decades. The most recent development was the addition of oxygen to hypothermic machine perfusion: HMP + O2. Oxygen is added to the preservation solution by methods such as membrane oxygenators or simply by inflating oxygen, thereby further reducing the anaerobic pathways within the remaining metabolic activity [10,11,12].
Normothermic machine perfusion (NMP) is also an ex situ perfusion approach that is applied after retrieval to circulate oxygenated perfusate/blood through the organ at temperatures of up to 35 to 38 °C. The setup includes an organ chamber, a perfusion pump, an oxygenator, a heat exchanger, and monitoring devices to measure flow, pressure, and temperature. This technique has the proposed benefit of providing a physiological environment for preservation and the potential of reconditioning the kidney as well as assessing viability and testing function during perfusion [13,14,15].
Normothermic regional perfusion (NRP) is applied in situ in DCD donors. An extra-corporeal membrane oxygenation (ECMO) machine is used to restore circulation with oxygenated blood to the abdominal organs in situ and has the benefit of reducing warm ischemia time and providing a controlled organ-retrieval process whilst protecting the organs from hypoxic injury [16,17,18]. NRP differs from other perfusion techniques because it is deployed within the donor during retrieval. The current standard of care for organ retrieval in DCD donors, against which NRP is being evaluated, is in situ cold perfusion (ICP), in which the cooling of organs is achieved by a single infusion of a cold preservative solution [19].
Due to the fact that more and more borderline grafts are being considered, the use of dynamic perfusion techniques is steadily increasing. The established approaches showed variable degrees of success in improving outcomes, as evidenced by several trials and meta-analyses, especially in organs from high-risk donors [15,17,20,21,22]. They have also introduced additional ways of assessing the viability and function of organs pre-transplant, the potential of optimizing organs by reconditioning, and ways of overcoming logistical challenges by prolonging the preservation period while maintaining better organ function [23,24]. Since a comparison of all clinical available perfusion approaches in kidney transplantation is lacking, this review was conducted to highlight the advantages and disadvantages of the different perfusion techniques.

2. Materials and Methods

2.1. Study Design

The study was registered with the PROSPERO database of systematic review protocols (PROSPERO ID: CRD42022379553). The systematic review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) [25](Figure 1).
Studies between 2010 to 2022 were identified from four electronic databases: MEDLINE (Ovid), Embase (embase.com), Scopus, and the Cochrane Trials Database. The literature search was completed by a professional information specialist from the University Library of the University of Zurich.

2.2. Search Strategy

  • Keywords for the search:
  • Kidney/renal perfusion
  • Kidney/renal preservation
  • Kidney/renal transplantation/transplant/graft/allograft
  • The specifics of the search strategy are outlined in Supplementary Table S9.
  • Last search was performed on 30 November 2022.

2.3. Outcomes

Primary outcome was the incidence of delayed graft function (DGF), defined as the need for dialysis within 7 days post-transplant.
Secondary outcomes were rates of rejection, graft survival, and patient survival after 1 year.

2.4. Data Extraction and Selection

Two reviewers independently reviewed the abstracts. Studies were included based on the following inclusion criteria; human grafts originated from deceased donors, with consecutive transplantation. and the primary outcome was reported.
Studies that fulfilled the following criteria were included in the meta-analyses:
  • A comparison between the outcomes (DGF) related to one of the machine perfusion techniques and SCS (i.e., randomized controlled trials, case control trials and observational studies) is included;
  • The results in the form of mean and standard deviation are published or can be obtained from median and range [26].
The following types of studies and publications were excluded:
  • Secondary studies such as systematic reviews and meta-analyses, as this would lead to duplication in data;
  • Pre-clinical studies such as animal and laboratory studies as well as letters to the editor and published protocols;
  • Case reports, abstracts, and publications that missed the minimum required data for reporting.
Studies involving combinations of more than one machine perfusion technique in the same patient/organ were excluded, because otherwise the effects of each perfusion approach could not be analyzed separately.
  • Suitable studies were identified, and full text analysis was performed. Discrepancies between reviewers were resolved after discussion between them and the senior authors.

2.5. Statistical Analysis

Case series and observational studies where the design included outcomes from only one technique of organ perfusion with no comparator were used as descriptive studies to demonstrate rates of incidence and frequencies of the outcomes with different types of machine perfusion in a variety of settings.
Results were presented as percentages for categorical data such as incidence of DGF, as well as graft and patient survival rates at 1 year. Numerical/quantitative data such as ages of donors and recipients, cold ischemia times (CIT), and serum creatinine levels at 1 year were presented as mean and standard deviation (SD) or median and interquartile range (IQR), depending on the mode in the publication.
Included studies were grouped according to technique, as in the aforementioned classification (i.e., HMP, HMP + O2, NMP, and NRP), and meta-analyses of the results of each group compared to SCS were performed using RevMan 5.4. A fixed-effect method was used for the analysis of more statistically homogeneous data (I2 < 50); furthermore, a random effects model was run for sensitivity analysis, which did not show any significant difference. For more heterogeneous results (I2 > 50), a random effects model was used to analyze and present data. Mantel–Haenszel approach was used for dichotomous data, and a standard inverse variance approach was used for continuous data. Outcomes were presented in separate forest plots to demonstrate effects and significance using Cochrane Review Manager [27].

3. Results

3.1. Literature Search and Systematic Review

In total, 5280 records were identified from the literature search based on the keywords, and then 2765 duplicates were removed. Of the 2515 remaining results, 2292 did not fulfill the inclusion criteria based on a title and abstract review. The full texts of 223 reports were assessed for eligibility, and 56 studies were included in the systematic review (Figure 1).

3.2. Hypothermic Machine Perfusion (without Oxygen) (HMP)

3.2.1. Descriptive Statistics

A total of 43 studies reported outcomes related to hypothermic machine perfusion (HMP).
The results from 34 studies revealed that 7543 patients received kidneys using HMP, 33.7% of which came from DCD. The mean age of donors was 45.4 years (SD +/−16), and the mean age of recipients was 50.34 years (SD +/−12.97). The overall incidence of DGF in kidneys preserved using HMP was 26.4%. Twenty studies exclusively reported outcomes for DCD kidneys or provided separate outcome results for DCD kidneys. The DGF rate in this subgroup was 45.6%. Ten studies exclusively provided results of ECD kidneys, and the incidence of DGF was 28%.
In nine studies, data were expressed as median with interquartile range (IQR) or 95% CI and, therefore, could not be included in the summary statistics but were maintained in the dataset for completeness.
Supplementary Table S10 contains a summary of the basic information available from all the studies reporting on HMP.

3.2.2. Meta-Analysis

Overall, 22 studies compared HMP to SCS, while 16 studies provided enough information for inclusion in the meta-analysis, representing a total of 7136 patients, 3546 (49.7%) HMP, and 3590 (50.3%) SCS. Supplementary Table S1 outlines the characteristics of the two groups and shows a higher donor age in the HMP group; otherwise, recipient age and the proportion of ECD and CIT were comparable among the two treatment groups (Supplementary Table S1).
DGF as the primary outcome was significantly lower in the HMP group vs. the SCS group (Figure 2), with an odds ratio (OR) of 0.63 (0.57, 0.71 confidence interval, CI). Further analysis also showed that, particularly within the group of DCD and ECD grafts, the DGF rates were lower in the HMP group (Figure 3 and Figure 4). A funnel plot to assess possible publication bias showed a fairly symmetrical distribution, indicating a low bias (Supplementary Figure S1). An assessment of the level of evidence and risk for bias for each study is shown in Supplementary Figures S2 and S3 as well as in Supplementary Table S12.
The rate of primary non-function (PNF) was also significantly lower in the group treated with HMP in comparison to that of SCS, with an OR of 0.44 (0.24, 0.82) (Figure 5).
Finally, where available, results of 1-year outcomes were analyzed and presented in the form of rates of rejection (Figure 6), 1-year graft survival (Figure 7), and 1-year patient survival (Figure 8). There was no statistically significant difference for any of these outcomes between HMP and SCS.

3.3. Hypothermic Oxygenated Machine Perfusion (HMP + O2)

3.3.1. Descriptive Statistics

Five studies assessed the effects of HMP + O2, including 263 grafts that were preserved using cold machine perfusion with oxygen. The mean age of donors was 62.5 years (SD +/−6), and the mean age of recipients was 60 years (SD +/−6.5). The overall rate of DGF was 29.7% (Supplementary Tables S2 and S3).

3.3.2. Comparative Studies

Three studies directly compared HMP + O2 to SCS. The characteristics in both groups were comparable with a longer CIT in the HMP + O2 treated grafts (Supplementary Table S4). All included grafts were from ECD.
Out of a total of 347 grafts, 152 (43.8%) HMP + O2 and 195 (56.2%) SCS preserved transplants were included. There was no significant difference between HMP + O2 and SCS in rates of DGF, with an OR of 0.86 (0.53, 1.40) for HMP + O2 (Figure 9). There was no significant difference between HMP + O2 and SCS in the rate of PNF or the 1-year rates of immune rejection, grafts and patient survival.

3.4. Normothermic Machine Perfusion (NMP)

Descriptive Statistics

Two studies assessing the effects of NMP fulfilled the inclusion criteria. In both studies, NMP was applied end-ischemically after SCS. In both studies, one of the main aims was to assess the safety of this technique and the potential of ex situ viability assessment.
Supplementary Table S5 summarizes the characteristics of the groups that were studied and the findings of both studies.
Due to the limited data that were provided, no further analysis could be performed.

3.5. Normothermic Regional Perfusion (NRP)

3.5.1. Descriptive Statistics

Six studies assessing the effects of NRP were within the inclusion criteria. In total, 340 participants received grafts treated with NRP, with a mean age of 42.9 years (SD +/−10.9) for recipients and 47.15 (SD +/−11.5) for donors (Supplementary Table S6). The overall rate of DGF was 71.5% (Supplementary Table S7). This technique is used exclusively in DCD grafts. Overall, 97% of the grafts included in our study were related to uncontrolled/Maastricht I–II DCD (uDCD), which is the highest risk group of donors for organ dysfunction (DGF and PNF) after transplantation.

3.5.2. Comparative Studies

Three studies covered in the systematic review provided enough data to evaluate the effect of NRP compared to that of ICP. In total, 343 grafts were included, of which 80 (23.3%) grafts were treated with NRP and 263 (76.7%) grafts were solely treated with ICP, as is common with the DCD retrieval procedure. There was no significant difference between the ages of the donors and recipients in both groups, and CIT was higher in the ICP group (Supplementary Table S8).
A meta-analysis of the results from these studies shows that NRP is associated with significantly lower rates of DGF compared to ICP, with an OR of 0.44 (0.23, 0.83) (Figure 10). The analysis that was conducted did not show any significance between the two groups for PNF, 1-year graft survival, or patient survival.

4. Discussion

This systematic review and meta-analysis highlights the importance of different dynamic perfusion techniques, especially new technologies such as HMP + O2, and the urgent need to further define their roles in the clinical setting of kidney transplantation. This is particularly relevant in an era of high-risk grafts, given the need to further expand the donor pool. The aim of dynamic preservation strategies is viability testing, reconditioning, and prediction of post-transplant outcomes.
Numerous studies showed worse short- to medium-term outcomes in DCD when compared to DBD, including high DGF rates, despite having comparable results in the long-term [48,49]. These data are supported by a recent meta-analysis that demonstrated higher rates of primary non-function (PNF) (RR 1.43, 1.26–1.62), DGF (RR 2.02, 1.88–2.16) and graft loss after 1 year (RR 1.10, 1.04–1.16) compared to DBD, with similar results for 10-year kidney function and graft loss [50]. Similarly, ECD grafts are known to have higher rates of DGF compared to DBD grafts [51,52].
Based on the findings of the included studies, DGF was used as the primary outcome for this study. There is evidence showing an association between DGF, episodes of rejection, and graft loss. A meta-analysis of 151,594 kidney transplants revealed a 41% increased risk of graft loss (RR 1.41, 1.27–1.56) at 3 years and a 38% relative increased risk of acute rejection (RR 1.38, 1.29–1.47) associated with DGF [53]. Furthermore, DGF has an impact on quality of life with the burden of dialysis sessions and transplant biopsies. DGF is also associated with considerable financial implications. Some studies from North America estimated an USD 18,000–USD 25,000 increase in costs for each episode of DGF due to the additional procedures and investigations [54,55]. Therefore, improvement in DGF rates would translate into improvements in clinical care, patient experience, and service costs. There is evidence showing the clear benefit of machine perfusion approaches in DCD kidney transplantation compared to SCS, but the results of newer machine perfusion approaches, e.g., HMP + O2 and normothermic machine perfusion, are limited to preclinical studies, feasibility studies, or studies that could not show any significant advantage of these perfusion strategies [15,20,28,43]. The basic research provided evidence on the mechanism of action and effects on a molecular level, and clinical trials are underway to prove that this is transferrable to practice [11,56,57].
Hypothermic machine perfusion is the most thoroughly studied method of machine perfusion in the literature and the gold standard of preservation for DCD kidneys in many countries worldwide. The review results show that HMP was associated with a significantly lower incidence of DGF compared to SCS (OR 0.63; 0.57, 0.71), despite having a longer CIT. These effects were even more marked in the subgroup analysis of kidneys from DCD, where there was an even lower incidence of DGF with HMP (OR 0.41, 0.81; p value < 0.001). The meta-analysis could also show a significantly lower rate of PNF when comparing HMP to SCS. No significant differences could be detected in the 1-year follow-ups comparing HMP to SCS. However, the short-term results justify the use of HMP to improve graft function and reduce the burden caused by DGF, such as additional dialysis sessions, ultrasound scans, and biopsies and a longer hospital stay. This would be even more valuable with marginal grafts at a higher risk of DGF, and there is growing evidence that supports that effect in DCD and ECD [28,29,30,31,32,33,42]. In addition to improving graft function, the clinical applications of HMP can be helpful in overcoming time and logistical challenges. This might help in bridging the lack of operating theater access or patient optimization, with less compromise in the outcomes. Thus, HMP can mitigate the effects of time and distance in the transplantation process, as it was shown to provide good outcomes despite a longer CIT [34,58,59,60,61]. One downside of this perfusion approach is the existing lack of objective parameters that can be assessed during perfusion to predict post-transplant outcomes. A recent Cochrane analysis showed the benefits of HMP compared to SCS, but newer perfusion approaches such as HMP + O2 and NMP could not be analyzed [62]. The studies included are heterogeneous regarding the time point when perfusion was started for upfront vs. end-ischemic graft perfusion. There is preclinical evidence showing the advantage of the upfront machine perfusion approach vs. the end-ischemic machine perfusion approach [63], but the damage in the end-ischemic perfusion group is much higher due to the additional cold ischemia time added.
Recent perfusion approaches such as HMP + O2 benefit from the same principles of hypothermia and the concepts of dynamic organ preservation such as HMP. Furthermore, the advantage of supplemental oxygen is demonstrated in pre-clinical studies and is already clinically established in liver transplantation [64]. Recently, this knowledge was assessed in the clinical setting of kidney transplantation in two randomized controlled trials (RCT) under the patronage of the COPE consortium. However, the most recent RCT failed to show additional benefits regarding the primary outcome measure, DGF. The study also did not show an advantage of HMP + O2 in patient survival or PNF. Looking deeper into the results and assessing the secondary outcomes, the addition of oxygen was able to decrease the number of severe complications (>Clavien Dindo IIIa) to 46 (11%) for 417 HMP + O2 (95% CI 8–14%) from 76 (16%) for 474 HMP (95% CI 13–20%) (p = 0.032), reduce the biopsy-proven episodes of acute rejections (a relative risk reduction of 44% (relative risk ratio 0.56, 95% CI 0.31–0.98)), and also show lower rates of graft failure with HMP + O2 (3 (3%) out of 106) compared with HMP (11 (10%) out of 106, hazard ratio 0.27, and 95% CI 0.07–0.95; p = 0.028) [12]. The other multicentric RCT conducted under the same patronage investigated the effects of short-term end-ischemic HMP + O2 vs. SCS in ECD grafts. The results of the study could not show any significant differences between the two treatment groups, but this study was probably underpowered. The very high overall graft survival rate contributed to that too [43]. Furthermore, with the limited number of patients included, the underlying graft injury in ECD grafts might be too small to show a significant difference in the included study population.
The results of our systematic review showed that DGF rates in the HMP + O2 vs. HMP-treated grafts were comparable (29.7% vs. 26.4%, respectively; ns). However, these results have to be interpreted with caution, as the grafts in the two groups were not comparable and were very heterogeneous. Mainly, all grafts in the HMP + O2 treatment group were either ECD or DCD grafts, whereas all donor types were represented in the HMP group. The meta-analysis comparing HMP + O2 vs. SCS did not show benefits for DGF, with an OR of 0.86 (0.53, 1.40). Again, the SCS group did not contain any DCD grafts, whereas the main indication for dynamic preservation strategies is grafts from high-risk donor groups. This analysis highlights the lack of clinical evidence assessing the effects of oxygenated perfusion in kidney transplantation. The strong, existing pre-clinical evidence supports intensifying further research to explore the effect of additional oxygen in kidney perfusion in the clinical setting. From a clinical point of view, HMP + O2 is easy to apply. Furthermore, HMP + O2 has the potential for organ assessment, reconditioning, immune modulation, and a reduction in rejection without many additional financial and time costs [56].
Normothermic machine perfusion (NMP) refers to ex situ perfusion of the organ at near physiological conditions, for organ preservation where the organ is oxygenated, and all metabolic pathways are maintained to keep the cellular functions intact. A deeper investigation of the underlying mechanism showed that normothermic preservation was associated with an increase in proteins mediating the key metabolic processes, including fatty acid ß-oxidation, the tricarboxylic acid cycle, and acid phosphorylation [65]. NMP upregulates certain cellular defense mechanisms, an effect that is similar to ischemic preconditioning, involving HIF proteins as well as pro-inflammatory cytokines such as TNF-a [11].
The effect and benefits of NMP are outlined in a number of pre-clinical studies, including animal models and laboratory experiments, which show the success of this method in preserving organs for prolonged periods of time [66]. This was also deemed successful after human kidneys were transplanted with a good outcome after being preserved at 34 °C using a blood-based solution. The results of our literature search revealed two small clinical studies in which NMP was used to preserve kidneys in a clinical setting for a cohort of patients, in the context of controlled trials that would yield clinically relevant data [67,68]. The rate of DGF in both studies was less for NMP when compared to SCS, but these differences were not significant. Despite the lack of significance, these trials could demonstrate the feasibility and safety of this technique. NMP might offer a solution to logistical issues, such as the lack of theater access, etc. Furthermore, NMP can be used for organ repair and reconditioning up to a certain degree [15]. In addition, this technique might represent a platform for pharmacological, gene, and stem cell therapy, as the intact metabolic pathways permits the administration of these treatments. This is still in the experimental phase for the time being but represents the potential for clinical implementation in the future [69,70]. The downside of this technique is the cost intensity as well as the rather complicated setup before organ perfusion.
Normothermic regional perfusion (NRP) is a technique exclusively used in a DCD setting, which was implemented in 1997 as a method for organ reconditioning before procurement [18,71]. This technique has to be assessed separately from the other techniques (HMP, HMP + O2, and NMP), as this perfusion approach is applied before and during retrieval. In our systematic review, six studies fulfilled the inclusion criteria, of which five presented data from uDCD. The inclusion of uDCD grafts might explain the higher DGF rate (71.5%) compared to that of other perfusion approaches. Due to its nature of being an in situ perfusion technique, it was directly compared with in situ cold perfusion (ICP). Here, organs are perfused with cold perfusion fluid in the donor after asystole during organ retrieval. Three studies were identified that compared these two perfusion approaches, including a mixture of DCD and uDCD grafts. The direct comparison of these two in situ perfusion approaches revealed a significant lower rate of DGF, with NRP (OR (95% CI) = 0.44 (0.23, 0.83)). These results have to be interpreted with caution because the number of studies and grafts is limited, and there is a degree of heterogeneity in the study population. In addition, ICP is performed after asystole, resulting in a relevant period of warm ischemia, whereas NRP is started earlier, before organ retrieval is initiated. Other studies showed the benefits of NRP in improving survival, so it is considered a useful method to enlarge the donor pool by allowing the utilization of marginal grafts while maintaining a reasonable safety profile [46]. From a clinical perspective, NRP is established in uDCD/Maastricht I–II, as it was successfully employed by Spanish organ retrieval teams in the “Donor from the street programme” and showed promising results [46,72]. It was recently adopted for DCD/Maastricht III organ donations as well. This is limited by the cost and labor-intensive nature of the technique as well as by the legislation in different countries.
A number of studies highlighted the possibility of using two different machine perfusion techniques in the same graft, combining the benefits of both approaches. Most studies highlighted the combination of NMP and HMP, while a further study suggested controlled oxygenated rewarming, which combines the use of HMP + O2 and NMP in a sequential manner [73,74,75,76]. The combination of NMP and HMP could show a higher urinary output, better flow, and higher oxygen consumption compared to NMP alone. Furthermore, preclinical studies combining HMP + O2 and NMP could show the benefits of lower renal resistance, better blood flow, and higher oxygen consumption compared to SCS grafts followed by NMP. In line with this, HMP + O2 significantly improved the initial perfusion hemodynamics [77]. However, these data are still preliminary and need further clinical validation.
There are a few limitations to our study including heterogeneity, risk of bias, and the effects of missing data.
Although I2 tests of the meta-analyses show reasonable homogeneity, there is a degree of heterogeneity among the study designs. Nevertheless, we felt that this mixture including observational studies and controlled trials allowed for coverage of a wide variety of contexts relevant to machine perfusion in kidney transplantation. Another source of heterogeneity within the data is the different types of donors; however, the inclusion of all types of donors, i.e., DBD, DCD, and ECD, allows for a better representation of all the types of grafts encountered in clinical practice; to overcome this issue, we performed subgroup analyses on the most important types of grafts.
Furthermore, there is a risk of bias as the meta-analysis is affected by the individual studies that make up the dataset, and, since some of the studies included are retrospective studies and not randomized trials, there is the potential for selection bias. Although there is no statistically significant difference between the characteristics of the groups included in the meta-analyses, the risk of selection bias cannot be completely ruled out.
There is also a limitation caused by missing data that are either not published and, therefore, inaccessible or reported in formats that did not allow for integration into the summary statistics and the meta-analyses (e.g., median and interquartile ranges). However, overall, a reasonable amount of information was reported and analyzed to provide the overall conclusion.

5. Conclusions

The systematic review and meta-analysis show the benefits of the different perfusion approaches in kidney transplantation. There is evidence directly comparing different perfusion strategies in pre-clinical models, but the transfer into the human setting is still pending. Real-time assessment, graft reconditioning, and the prediction of post-transplant outcomes are hot topics in the transplantation society worldwide, but, so far, objective parameters for the acceptance of grafts or for the prediction of post-transplant outcomes in kidney transplantation are missing. Whereas, the evidence for HMP in kidney transplantation is supported by several studies on newer machine perfusion approaches, such as HMP + O2 and NMP in kidney transplantation, which need further evaluation in humans before any clinically relevant conclusions can be drawn.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12123871/s1, Supplementary Table S1: Characteristics of HMP and SCS groups included in the meta-analysis; Supplementary Table S2: Study design, donor and recipient characteristics in HMP + O2; Supplementary Table S3: HMP + O2–outcomes; Supplementary Table S4: Characteristics of HMP + O2 and SCS groups included in the meta-analysis; Supplementary Table S5: Characteristics and outcomes of studies related to NMP; Supplementary Table S6: Study design, donor and recipient characteristics in NRP; Supplementary Table S7: NRP—Outcomes; Supplementary Table S8: Characteristics of NRP and ICP groups included in the meta-analysis; Supplementary Table S9: Search Strategy; Supplementary Table S10: Study design, donor and recipient characteristics in HMP; Supplementary Table S11: HMP—Outcomes; Supplementary Table S12: Level of evidence and quality level for bias risk (Newcas-tle-Ottawa score); Supplementary Figure S1: Funnel plot for DGF in studies comparing HMP vs. SCS; Supplementary Figure S2: Risk-of-bias graph and summary table. Authors’ judgments about each risk-of-bias item as percentages among all included HMP vs. SCS studies; Supplementary Figure S3: Risk-of-bias graph and summary table. Authors’ judgments about each risk-of-bias item as percentages among all included HMP + O2 vs. SCS studies [12,19,23,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,58,59,60,61,67,68,72,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102].

Author Contributions

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

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CITCold Ischemia Time
DBDDonation after Brain Death
DCDDonation after Circulatory Death
DGFDelayed Graft Function
ECDExtended Criteria Donors
ECMOExtra-Corporeal Membrane Oxygenation
HMPHypothermic Machine Perfusion
HOPEHypothermic Oxygenated Perfusion
ICPIn situ Cold Perfusion
IQRInterquartile Range
NMPNormothermic Machine Perfusion
NRPNormothermic Regional Perfusion
OROdds Ratio
PNFPrimary Non-Function
PRISMAPreferred Reporting Items for Systematic Reviews and Meta-Analyses
RRRelative Risk
SCDStandard Criteria Donors
SCSStatic Cold Storage
SDStandard Deviation
uDCDuncontrolled Donation after Cardiac Death

References

  1. Wolfe, R.A.; Ashby, V.B.; Milford, E.L.; Ojo, A.O.; Ettenger, R.E.; Agodoa, L.Y.C.; 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] [PubMed] [Green Version]
  2. Evans, R.W.; Manninen, D.L.; Garrison, L.P.; Hart, L.G.; Blagg, C.R.; Gutman, R.A.; Hull, A.R.; Lowrie, E.G. The Quality of Life of Patients with End-Stage Renal-Disease. N. Engl. J. Med. 1985, 312, 553–559. [Google Scholar] [CrossRef] [PubMed]
  3. Schaubel, D.; Desmeules, M.; Mao, Y.; Jeffery, J.; Fenton, S. Survival experience among elderly end-stage renal disease patients. Transplantation 1995, 60, 1389–1394. [Google Scholar] [CrossRef] [PubMed]
  4. Metzger, R.A.; Delmonico, F.L.; Feng, S.; Port, F.K.; Wynne, J.J.; Merion, R.M. Expanded criteria donors for kidney transplantation. Am. J. Transplant. 2003, 3, 114–125. [Google Scholar] [CrossRef] [Green Version]
  5. Messina, M.; Diena, D.; Dellepiane, S.; Guzzo, G.; Lo Sardo, L.; Fop, F.; Segoloni, G.P.; Amoroso, A.; Magistroni, P.; Biancone, L. Long-Term Outcomes and Discard Rate of Kidneys by Decade of Extended Criteria Donor Age. Clin. J. Am. Soc. Nephrol. 2017, 12, 323–331. [Google Scholar] [CrossRef] [Green Version]
  6. Rosengard, B.R.; Feng, S.; Alfrey, E.J.; Zaroff, J.G.; Emond, J.C.; Henry, M.L.; Garrity, E.R.; Roberts, J.P.; Wynn, J.J.; Metzger, R.A.; et al. Report of the crystal city meeting to maximize the use of organs recovered from the cadaver donor. Am. J. Transplant. 2002, 2, 701–711. [Google Scholar] [CrossRef] [Green Version]
  7. McDonald, S.; Clayton, P. DCD ECD Kidneys—Can You Make a Silk Purse from a Sow’s Ear? Am. J. Transplant. 2013, 13, 249–250. [Google Scholar] [CrossRef]
  8. Tatsis, V.; Dounousi, E.; Mitsis, M. Hypothermic Machine Perfusion of Kidney Transplant: A Mini-Review. Transpl. Proc. 2021, 53, 2793–2796. [Google Scholar] [CrossRef]
  9. Karangwa, S.A.; Dutkowski, P.; Fontes, P.; Friend, P.J.; Guarrera, J.V.; Markmann, J.F.; Mergental, H.; Minor, T.; Quintini, C.; Selzner, M.; et al. Machine Perfusion of Donor Livers for Transplantation: A Proposal for Standardized Nomenclature and Reporting Guidelines. Am. J. Transplant. 2016, 16, 2932–2942. [Google Scholar] [CrossRef]
  10. Hosgood, S.A.; Nicholson, H.F.L.; Nicholson, M.L. Oxygenated Kidney Preservation Techniques. Transplantation 2012, 93, 455–459. [Google Scholar] [CrossRef]
  11. Kron, P.; Schlegel, A.; de Rougemont, O.; Oberkofler, C.E.; Clavien, P.A.; Dutkowski, P. Short, Cool, and Well Oxygenated—HOPE for Kidney Transplantation in a Rodent Model. Ann. Surg. 2016, 264, 815–822. [Google Scholar] [CrossRef] [Green Version]
  12. 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.; Collaboration, C.T.; et al. 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]
  13. Hosgood, S.A.; Nicholson, M.L. Normothermic kidney preservation. Curr. Opin. Organ. Transplant. 2011, 16, 169–173. [Google Scholar] [CrossRef]
  14. 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. Brit. J. Surg. 2018, 105, 388–394. [Google Scholar] [CrossRef] [Green Version]
  15. Hosgood, S.A.; van Heurn, E.; Nicholson, M.L. Normothermic machine perfusion of the kidney: Better conditioning and repair? Transpl. Int. 2015, 28, 657–664. [Google Scholar] [CrossRef]
  16. Oniscu, G.C.; Randle, L.V.; Muiesan, P.; Butler, A.J.; Currie, I.S.; Perera, M.T.; Forsythe, J.L.; Watson, C.J. In situ normothermic regional perfusion for controlled donation after circulatory death—The United Kingdom experience. Am. J. Transplant. 2014, 14, 2846–2854. [Google Scholar] [CrossRef]
  17. Padilla, M.; Coll, E.; Fernandez-Perez, C.; Pont, T.; Ruiz, A.; Perez-Redondo, M.; Oliver, E.; Atutxa, L.; Mancino, J.M.; Daga, D.; et al. Improved short-term outcomes of kidney transplants in controlled donation after the circulatory determination of death with the use of normothermic regional perfusion. Am. J. Transplant. 2021, 21, 3618–3628. [Google Scholar] [CrossRef]
  18. Minambres, E.; Suberviola, B.; Dominguez-Gil, B.; Rodrigo, E.; Millan, J.C.R.S.; Juan, J.C.R.S.; Ballesteros, M.A. Improving the Outcomes of Organs Obtained from Controlled Donation After Circulatory Death Donors Using Abdominal Normothermic Regional Perfusion. Am. J. Transplant. 2017, 17, 2165–2172. [Google Scholar] [CrossRef] [Green Version]
  19. Delsuc, C.; Faure, A.; Berthiller, J.; Dorez, D.; Matillon, X.; Meas-Yedid, V.; Floccard, B.; Marcotte, G.; Labeye, V.; Rabeyrin, M.; et al. Uncontrolled donation after circulatory death: Comparison of two kidney preservation protocols on graft outcomes. BMC Nephrol. 2018, 19, 3. [Google Scholar] [CrossRef] [Green Version]
  20. Moers, C.; Smits, J.M.; Maathuis, M.H.; Treckmann, J.; van Gelder, F.; Napieralski, B.P.; van Kasterop-Kutz, M.; van der Heide, J.J.; 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] [Green Version]
  21. O’Callaghan, J.M.; Knight, S.R.; Morgan, R.D.; Morris, P.J. Preservation Solutions for Static Cold Storage of Kidney Allografts: A Systematic Review and Meta-Analysis. Am. J. Transplant. 2012, 12, 896–906. [Google Scholar] [CrossRef] [PubMed]
  22. O’Callaghan, J.M.; Pall, K.T.; Pengel, L.H.M. Cope. Supplemental oxygen during hypothermic kidney preservation: A systematic review. Transplant. Rev. 2017, 31, 172–179. [Google Scholar] [CrossRef] [PubMed]
  23. Jochmans, I.; Moers, C.; Smits, J.M.; Leuvenink, H.G.D.; Treckmann, J.; Paul, A.; Rahmel, A.; Squifflet, J.P.; van Heurn, E.; Monbaliu, D.; et al. The Prognostic Value of Renal Resistance During Hypothermic Machine Perfusion of Deceased Donor Kidneys. Am. J. Transplant. 2011, 11, 2214–2220. [Google Scholar] [CrossRef] [PubMed]
  24. Bissolati, M.; Pindozzi, F.; Guarneri, G.; Adamenko, O.; Giannone, F.; Mazza, M.; Maggi, G.; Rosati, R.; Secchi, A.; Socci, C. Hypothermic Machine Perfusion as an Alternative to Biopsy Assessment in Transplantation of Kidneys Donated After Cardiocirculatory Death: A Pilot Study. Transpl. Proc. 2019, 51, 2890–2898. [Google Scholar] [CrossRef]
  25. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ. 2021, 71, 732. [Google Scholar] [CrossRef]
  26. Hozo, S.P.; Djulbegovic, B.; Hozo, I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med. Res. Methodol. 2005, 5, 13. [Google Scholar] [CrossRef] [Green Version]
  27. The Cochrane Collaboration. Review Manager (RevMan). Available online: https://training.cochrane.org/online-learning/core-software/revman (accessed on 1 June 2022).
  28. 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: A multicenter, randomized, controlled trial. Ann. Surg. 2010, 252, 756–764. [Google Scholar] [CrossRef]
  29. Abboud, I.; Antoine, C.; Gaudez, F.; Fieux, F.; Lefaucheur, C.; Pillebout, E.; Viglietti, D.; Serrato, T.; Verine, J.; Flamant, M.; et al. Pulsatile perfusion preservation for expanded-criteria donors kidneys: Impact on delayed graft function rate. Int. J. Artif. Organs 2011, 34, 513–518. [Google Scholar] [CrossRef]
  30. Treckmann, J.; Moers, C.; Smits, J.M.; Gallinat, A.; Maathuis, M.H.; van Kasterop-Kutz, M.; Jochmans, I.; Homan van der Heide, J.J.; Squifflet, J.P.; van Heurn, E.; et al. Machine perfusion versus cold storage for preservation of kidneys from expanded criteria donors after brain death. Transpl. Int. 2011, 24, 548–554. [Google Scholar] [CrossRef] [Green Version]
  31. Gallinat, A.; Moers, C.; Treckmann, J.; Smits, J.M.; Leuvenink, H.G.; Lefering, R.; van Heurn, E.; Kirste, G.R.; Squifflet, J.P.; Rahmel, A.; et al. Machine perfusion versus cold storage for the preservation of kidneys from donors >/= 65 years allocated in the Eurotransplant Senior Programme. Nephrol. Dial. Transplant. 2012, 27, 4458–4463. [Google Scholar] [CrossRef] [Green Version]
  32. Yao, L.; Zhou, H.; Wang, Y.; Wang, G.; Wang, W.; Chen, M.; Zhang, K.; Fu, Y. Hypothermic Machine Perfusion in DCD Kidney Transplantation: A Single Center Experience. Urol. Int. 2016, 96, 148–151. [Google Scholar] [CrossRef]
  33. Wang, W.; Xie, D.W.; Hu, X.P.; Yin, H.; Liu, H.; Zhang, X.D. Effect of Hypothermic Machine Perfusion on the Preservation of Kidneys Donated After Cardiac Death: A Single-Center, Randomized, Controlled Trial. Artif. Organs 2017, 41, 753–758. [Google Scholar] [CrossRef]
  34. Basu, A.; Rosen, L.M.; Tan, H.P.; Fishbein, J.; Wu, C.M.; Donaldson, O.B.; Stuart, S.; Shah, N.A.; McCauley, J.; Humar, A.; et al. Outcomes of Deceased Donor Kidney Transplantation Using Expanded Criteria Donor Kidneys Following Pulsatile Preservation. Cureus 2019, 11, e5091. [Google Scholar] [CrossRef] [Green Version]
  35. Watson, C.J.E.; Wells, A.C.; Roberts, R.J.; Akoh, J.A.; Friend, P.J.; Akyol, M.; Calder, F.R.; Allen, J.E.; Jones, M.N.; Collett, D.; et al. Cold Machine Perfusion Versus Static Cold Storage of Kidneys Donated After Cardiac Death: A UK Multicenter Randomized Controlled Trial. Am. J. Transplant. 2010, 10, 1991–1999. [Google Scholar] [CrossRef]
  36. Cannon, R.M.; Brock, G.N.; Garrison, R.N.; Smith, J.W.; Marvin, M.R.; Franklin, G.A. To pump or not to pump: A comparison of machine perfusion vs cold storage for deceased donor kidney transplantation. J. Am. Coll. Surg. 2013, 216, 625–633. [Google Scholar] [CrossRef]
  37. Sedigh, A.; Tufveson, G.; Backman, L.; Biglarnia, A.R.; Lorant, T. Initial experience with hypothermic machine perfusion of kidneys from deceased donors in the Uppsala region in Sweden. Transplant. Proc. 2013, 45, 1168–1171. [Google Scholar] [CrossRef]
  38. Dion, M.S.; McGregor, T.B.; McAlister, V.C.; Luke, P.P.; Sener, A. Hypothermic machine perfusion improves Doppler ultrasonography resistive indices and long-term allograft function after renal transplantation: A single-centre analysis. Bju Int. 2015, 116, 932–937. [Google Scholar] [CrossRef]
  39. Forde, J.C.; Shields, W.P.; Azhar, M.; Daly, P.J.; Zimmermann, J.A.; Smyth, G.P.; Eng, M.P.; Power, R.E.; Mohan, P.; Hickey, D.P.; et al. Single centre experience of hypothermic machine perfusion of kidneys from extended criteria deceased heart-beating donors: A comparative study. Ir. J. Med. Sci. 2016, 185, 121–125. [Google Scholar] [CrossRef]
  40. Arlaban, M.; Barreda, P.; Ballesteros, M.A.; Rodrigo, E.; Suberviola, B.; Valero, R.; Minambres, E.; Ruiz-San Millan, J.C. Static Cold Storage vs Ex Vivo Machine Perfusion: Results from a Comparative Study on Renal Transplant Outcome in a Controlled Donation After Circulatory Death Program. Transpl. Proc. 2019, 51, 311–313. [Google Scholar] [CrossRef] [Green Version]
  41. 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]
  42. Zhong, Z.; Lan, J.; Ye, S.; Liu, Z.; Fan, L.; Zhang, Y.; Fu, Z.; Qiao, B.; Shiu-Chung Ko, D.; Wang, Y.; et al. Outcome Improvement for Hypothermic Machine Perfusion Versus Cold Storage for Kidneys from Cardiac Death Donors. Artif. Organs 2017, 41, 647–653. [Google Scholar] [CrossRef] [PubMed]
  43. Husen, P.; Boffa, C.; Jochmans, I.; Krikke, C.; Davies, L.; Mazilescu, L.; Brat, A.; Knight, S.; Wettstein, D.; Cseprekal, O.; et al. Oxygenated End-Hypothermic Machine Perfusion in Expanded Criteria Donor Kidney Transplant A Randomized Clinical Trial. Jama Surg. 2021, 156, 517–525. [Google Scholar] [CrossRef] [PubMed]
  44. 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, 17726. [Google Scholar] [CrossRef] [PubMed]
  45. 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, 6063. [Google Scholar] [CrossRef] [Green Version]
  46. Demiselle, J.; Augusto, J.F.; Videcoq, M.; Legeard, E.; Dube, L.; Templier, F.; Renaudin, K.; Sayegh, J.; Karam, G.; Blancho, G.; et al. Transplantation of kidneys from uncontrolled donation after circulatory determination of death: Comparison with brain death donors with or without extended criteria and impact of normothermic regional perfusion. Transpl. Int. 2016, 29, 432–442. [Google Scholar] [CrossRef] [Green Version]
  47. Pearson, R.; Geddes, C.; Mark, P.; Clancy, M.; Asher, J. Transplantation of kidneys after normothermic perfusion: A single center experience. Clin. Transplant. 2021, 35. [Google Scholar] [CrossRef]
  48. Summers, D.M.; Watson, C.J.E.; Pettigrew, G.J.; Johnson, R.J.; Collett, D.; Neuberger, J.M.; Bradley, J.A. Kidney donation after circulatory death (DCD): State of the art. Kidney Int. 2015, 88, 241–249. [Google Scholar] [CrossRef]
  49. Singh, R.P.; Farney, A.C.; Rogers, J.; Zuckerman, J.; Reeves-Daniel, A.; Hartmann, E.; Iskandar, S.; Adams, P.; Stratta, R.J. Kidney transplantation from donation after cardiac death donors: Lack of impact of delayed graft function on post-transplant outcomes. Clin. Transplant. 2011, 25, 255–264. [Google Scholar] [CrossRef]
  50. Rijkse, E.; Ceuppens, S.; Qi, H.C.; IJzermans, J.N.M.; Hesselink, D.A.; Minnee, R.C. Implementation of donation after circulatory death kidney transplantation can safely enlarge the donor pool: A systematic review and meta-analysis. Int. J. Surg. 2021, 92, 106021. [Google Scholar] [CrossRef]
  51. Johnston, T.D.; Thacker, L.R.; Jeon, H.; Lucas, B.A.; Ranjan, D. Sensitivity of expanded-criteria donor kidneys to cold ischaemia time. Clin. Transplant. 2004, 18, 28–32. [Google Scholar] [CrossRef]
  52. Pascual, J.; Zamora, J.; Pirsch, J.D. A systematic review of kidney transplantation from expanded criteria donors. Am. J. Kidney Dis. 2008, 52, 553–586. [Google Scholar] [CrossRef]
  53. Yarlagadda, S.G.; Coca, S.G.; Formica, R.N.; Poggio, E.D.; Parikh, C.R. Association between delayed graft function and allograft and patient survival: A systematic review and meta-analysis. Nephrol. Dial. Transpl. 2009, 24, 1039–1047. [Google Scholar] [CrossRef] [Green Version]
  54. Freedland, S.J.; Shoskes, D.A. Economic impact of delayed graft function and suboptimal kidneys. Transplant. Rev. 1999, 13, 23–30. [Google Scholar] [CrossRef]
  55. Kim, D.W.; Tsapepas, D.; King, K.L.; Husain, S.A.; Corvino, F.A.; Dillon, A.; Wang, W.; Mayne, T.J.; Mohan, S. Financial impact of delayed graft function in kidney transplantation. Clin. Transplant. 2020, 34, e14022. [Google Scholar] [CrossRef]
  56. Kron, P.; Schlegel, A.; Muller, X.; Gaspert, A.; Clavien, P.A.; Dutkowski, P. Hypothermic Oxygenated Perfusion: A Simple and Effective Method to Modulate the Immune Response in Kidney Transplantation. Transplantation 2019, 103, E128–E136. [Google Scholar] [CrossRef]
  57. Kaths, J.M.; Hamar, M.; Echeverri, J.; Linares, I.; Urbanellis, P.; Cen, J.Y.; Ganesh, S.; Dingwell, L.S.; Yip, P.; John, R.; et al. Normothermic ex vivo kidney perfusion for graft quality assessment prior to transplantation. Am. J. Transplant. 2018, 18, 580–589. [Google Scholar] [CrossRef] [Green Version]
  58. de Sandes-Freitas, T.V.; Costa, S.D.; de Andrade, L.G.M.; Girao, C.M.; Fernandes, P.; de Oliveira, C.M.C.; Esmeraldo, R.M. The Impact of Hypothermic Pulsatile Machine Perfusion Versus Static Cold Storage: A Donor-Matched Paired Analysis in a Scenario of High Incidence of Delayed Kidney Graft Function. Ann. Transplant. 2020, 25, e927010. [Google Scholar] [CrossRef]
  59. Matos, A.C.C.; Requiao Moura, L.R.; Borrelli, M.; Nogueira, M.; Clarizia, G.; Ongaro, P.; Durao, M.S.; Pacheco-Silva, A. Impact of machine perfusion after long static cold storage on delayed graft function incidence and duration and time to hospital discharge. Clin. Transplant. 2018, 32, e13130. [Google Scholar] [CrossRef]
  60. Guy, A.; McGrogan, D.; Inston, N.; Ready, A. Hypothermic machine perfusion permits extended cold ischemia times with improved early graft function. Exp. Clin. Transplant. 2015, 13, 130–137. [Google Scholar]
  61. Ciancio, G.; Gaynor, J.J.; Sageshima, J.; Chen, L.; Roth, D.; Kupin, W.; Guerra, G.; Tueros, L.; Zarak, A.; Hanson, L.; et al. Favorable outcomes with machine perfusion and longer pump times in kidney transplantation: A single-center, observational study. Transplantation 2010, 90, 882–890. [Google Scholar] [CrossRef]
  62. Tingle, S.J.; Figueiredo, R.S.; Moir, J.A.; Goodfellow, M.; Talbot, D.; Wilson, C.H. Machine perfusion preservation versus static cold storage for deceased donor kidney transplantation. Cochrane Database Syst. Rev. 2019, 3, CD011671. [Google Scholar] [CrossRef] [PubMed]
  63. 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 ischemia-reperfusion autotransplant model. Am. J. Transplant. 2019, 19, 752–762. [Google Scholar] [CrossRef] [PubMed]
  64. Dutkowski, P.; Polak, W.G.; Muiesan, P.; Schlegel, A.; Verhoeven, C.J.; Scalera, I.; DeOliveira, M.L.; Kron, P.; Clavien, P.A. First Comparison of Hypothermic Oxygenated PErfusion Versus Static Cold Storage of Human Donation After Cardiac Death Liver Transplants an International-matched Case Analysis. Ann. Surg. 2015, 262, 764–771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. McEvoy, C.M.; Clotet-Freixas, S.; Tokar, T.; Pastrello, C.; Reid, S.; Batruch, I.; RaoPeters, A.A.E.; Kaths, J.M.; Urbanellis, P.; Farkona, S.; et al. Normothermic Ex-vivo Kidney Perfusion in a Porcine Auto-Transplantation Model Preserves the Expression of Key Mitochondrial Proteins: An Unbiased Proteomics Analysis. Mol. Cell. Proteom. 2021, 20, 100101. [Google Scholar] [CrossRef] [PubMed]
  66. Brasile, L.; Stubenitsky, B.M.; Booster, M.H.; Arenada, D.; Haisch, C.; Kootstra, G. Hypothermia—A limiting factor in using warm ischemically damaged kidneys. Am. J. Transplant. 2001, 1, 316–320. [Google Scholar] [CrossRef]
  67. Nicholson, M.L.; Hosgood, S.A. Renal Transplantation After Ex Vivo Normothermic Perfusion: The First Clinical Study. Am. J. Transplant. 2013, 13, 1246–1252. [Google Scholar] [CrossRef]
  68. Chandak, P.; Phillips, B.L.; Uwechue, R.; Thompson, E.; Bates, L.; Ibrahim, I.; Sewpaul, A.; Figueiredo, R.; Olsburgh, J.; Hosgood, S.; et al. Dissemination of a novel organ perfusion technique: Ex vivo normothermic perfusion of deceased donor kidneys. Artif. Organs 2019, 43, E308–E319. [Google Scholar] [CrossRef]
  69. Steichen, C.; Erpicum, P. Combining cell-based therapy and normothermic machine perfusion for kidney graft conditioning has gone one step further. Am. J. Transplant. 2021, 21, 1359–1360. [Google Scholar] [CrossRef]
  70. Hosgood, S.A.; Bagul, A.; Kaushik, M.; Rimoldi, J.; Gadepalli, R.S.; Nicholson, M.L. Application of nitric oxide and carbon monoxide in a model of renal preservation. Brit. J. Surg. 2008, 95, 1060–1067. [Google Scholar] [CrossRef]
  71. Johnson, L.B.; Plotkin, J.S.; Howell, C.D.; Njoku, M.J.; Kuo, P.C.; Bartlett, S.T. Successful emergency transplantation of a liver allograft from a donor maintained on extracorporeal, membrane oxygenation. Transplantation 1997, 63, 910–911. [Google Scholar] [CrossRef]
  72. Reznik, O.; Bagnenko, S.; Scvortsov, A.; Loginov, I.; Ananyev, A.; Senchik, K.; Moysyuk, Y. The use of in-situ normothermic extracorporeal perfusion and leukocyte depletion for resuscitation of human donor kidneys. Perfusion 2010, 25, 343–348. [Google Scholar] [CrossRef]
  73. 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]
  74. Mori, G.; Cerami, C.; Facchini, F.; Fontana, F.; Alfano, G.; Giovanni, R.; Cappelli, G. Kidney Transplantation from Circulatory Death Donors: Monocentric Experience. Transplant. Proc. 2019, 51, 2865–2867. [Google Scholar] [CrossRef]
  75. Ravaioli, M.; De Pace, V.; Comai, G.; Capelli, I.; Baraldi, O.; D’Errico, A.; Bertuzzo, V.R.; Del Gaudio, M.; Zanfi, C.; D’Arcangelo, G.L.; et al. Preliminary experience of sequential use of normothermic and hypothermic oxygenated perfusion for donation after circulatory death kidney with warm ischemia time over the conventional criteria—A retrospective and observational study. Transpl. Int. 2018, 31, 1233–1244. [Google Scholar] [CrossRef] [Green Version]
  76. Matillon, X.; Danjou, F.; Petruzzo, P.; Thaunat, O.; Rimmele, T.; Delsuc, C.; Faure, A.; Rabeyrin, M.; Yedid, V.M.; Hanf, W.; et al. Hypothermic pulsatile preservation of kidneys from uncontrolled deceased donors after cardiac arrest—A retrospective study. Transpl. Int. 2017, 30, 1284–1291. [Google Scholar] [CrossRef] [Green Version]
  77. Zlatev, H.; von Horn, C.; Kaths, M.; Paul, A.; Minor, T. Clinical use of controlled oxygenated rewarming of kidney grafts prior to transplantation by ex vivo machine perfusion. A pilot study. Eur. J. Clin. Invest. 2022, 52, e13691. [Google Scholar] [CrossRef]
  78. Houtzager, J.H.E.; Hemelrijk, S.D.; Post, I.C.J.H.; Idu, M.M.; Bemelman, F.J.; van Gulik, T.M. The Use of the Oxygenated Airdrive(TM) Machine Perfusion System in Kidney Graft Preservation: A Clinical Pilot Study. Eur. Surg. Res. 2021, 61, 153–162. [Google Scholar] [CrossRef]
  79. Reznik, O.; Bagnenko, S.; Skvortsov, A.; Ananyev, A.; Senchik, K.; Loginov, I.; Moysyuk, Y. Rehabilitation of Ischemically Damaged Human Kidneys by Normothermic Extracorporal Hemoperfusion in situ with Oxygenation and Leukocyte Depletion. Transpl. Proc. 2010, 42, 1536–1538. [Google Scholar] [CrossRef]
  80. Miranda-Utrera, N.; Medina-Polo, J.; Pamplona-Casamayor, M.; Passas-Martinez, J.B.; Rodriguez-Antolin, A.; Kehrmann, F.D.; Duarte-Ojeda, J.M.; Tejido-Sanchez, A.; Auba, F.V.; Belmonte, A.A. Uncontrolled non-heartbeating donors (types I-II) with normothermic recirculation vs. heartbeating donors: Evaluation of functional results and survival. Actas Urol. Esp. 2015, 39, 429–434. [Google Scholar] [CrossRef]
  81. Catena, F.; Gazzotti, F.; Amaduzzi, A.; Fuga, G.; Montori, G.; Cucchetti, A.; Coccolini, F.; Vallicelli, C.; Pinna, A.D. Pulsatile Perfusion of Kidney Allografts With Celsior Solution. Transpl. Proc. 2010, 42, 3971–3972. [Google Scholar] [CrossRef]
  82. Guarrera, J.V.; Goldstein, M.J.; Samstein, B.; Henry, S.; Reverte, C.; Arrington, B.; Brown, T.; Coleman, T.K.; Mattei, G.; Mendez, N.; et al. ‘When good kidneys pump badly’: Outcomes of deceased donor renal allografts with poor pulsatile perfusion characteristics. Transpl. Int. 2010, 23, 444–446. [Google Scholar] [CrossRef] [PubMed]
  83. Matsuno, N.; Konno, Y.N.O.; Jyojima, Y.; Akashi, I.; Iwamoto, H.; Hama, K.; Hiirano, T.; Nagao, T. Machine Perfusion Preservation for Kidney Grafts with a High Creatinine from Uncontrolled Donation After Cardiac Death. Transpl. Proc. 2010, 42, 155–158. [Google Scholar] [CrossRef] [PubMed]
  84. Moers, C.; Varnav, O.C.; van Heurn, E.; Jochmans, I.; Kirste, G.R.; Rahmel, A.; Leuvenink, H.G.; 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] [PubMed] [Green Version]
  85. Ciancio, G.; Gaynor, J.J.; Sageshima, J.; Roth, D.; Kupin, W.; Guerra, G.; Tueros, L.; Zarak, A.; Hanson, L.; Ganz, S.; et al. Machine perfusion following static cold storage preservation in kidney transplantation: Donor-matched pair analysis of the prognostic impact of longer pump time. Transpl. Int. 2012, 25, 34–40. [Google Scholar] [CrossRef]
  86. Patel, S.K.; Pankewycz, O.G.; Weber-Shrikant, E.; Zachariah, M.; Kohli, R.; Nader, N.D.; Laftavi, M.R. Effect of Increased Pressure During Pulsatile Pump Perfusion of Deceased Donor Kidneys in Transplantation. Transpl. Proc. 2012, 44, 2202–2206. [Google Scholar] [CrossRef]
  87. Hoogland, E.R.; de Vries, E.E.; Christiaans, M.H.; Winkens, B.; Snoeijs, M.G.; van Heurn, L.W. The value of machine perfusion biomarker concentration in DCD kidney transplantations. Transplantation 2013, 95, 603–610. [Google Scholar] [CrossRef]
  88. Nagelschmidt, M.; Minor, T.; Gallinat, A.; Moers, C.; Jochmans, I.; Pirenne, J.; Ploeg, R.J.; Paul, A.; Treckmann, J. Lipid peroxidation products in machine perfusion of older donor kidneys. J. Surg. Res. 2013, 180, 337–342. [Google Scholar] [CrossRef]
  89. Wszola, M.; Kwiatkowski, A.; Diuwe, P.; Domagala, P.; Gorski, L.; Kieszek, R.; Berman, A.; Perkowska-Ptasinska, A.; Durlik, M.; Paczek, L.; et al. One-year results of a prospective, randomized trial comparing two machine perfusion devices used for kidney preservation. Transpl. Int. 2013, 26, 1088–1096. [Google Scholar] [CrossRef]
  90. Hall, I.; Bhangoo, R.; Reese, P.; Doshi, M.; Weng, F.; Hasz, R.; Goldstein, M.; Schroppel, B.; Parikh, C. Glutathione S-Transferase Iso-Enzymes in Perfusate From Pumped Kidneys Are Associated With Delayed Graft Function. Am. J. Transplant. 2014, 14, 882–883. [Google Scholar] [CrossRef] [Green Version]
  91. Guy, A.J.; Nath, J.; Cobbold, M.; Ludwig, C.; Tennant, D.A.; Inston, N.G.; Ready, A.R. Metabolomic Analysis of Perfusate During Hypothermic Machine Perfusion of Human Cadaveric Kidneys. Transplantation 2015, 99, 754–759. [Google Scholar] [CrossRef]
  92. Paloyo, S.; Sageshima, J.; Gaynor, J.J.; Chen, L.D.; Ciancio, G.; Burke, G.W. Negative impact of prolonged cold storage time before machine perfusion preservation in donation after circulatory death kidney transplantation. Transpl. Int. 2016, 29, 1117–1125. [Google Scholar] [CrossRef]
  93. Parikh, C.R.; Hall, I.E.; Bhangoo, R.S.; Ficek, J.; Abt, P.L.; Thiessen-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. Transplant. 2016, 16, 1526–1539. [Google Scholar] [CrossRef] [Green Version]
  94. Chen, G.D.; Wang, C.; Zhao, Y.; Qiu, L.H.; Yuan, X.P.; Qiu, J.; Wang, C.X.; He, X.S.; Chen, L.Z. 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]
  95. Ding, C.G.; Tian, P.X.; Ding, X.M.; Xiang, H.L.; Li, Y.; Tian, X.H.; Han, F.; Tai, Q.H.; Liu, Q.L.; Zheng, J.; et al. Beneficial Effect of Moderately Increasing Hypothermic Machine Perfusion Pressure on Donor after Cardiac Death Renal Transplantation. Chin. Med. J. 2018, 131, 2676–2682. [Google Scholar] [CrossRef]
  96. 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 Ischemia Times in Deceased Donor Kidneys. Transplantation 2018, 102, 1344–1350. [Google Scholar] [CrossRef]
  97. 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. Transplant. 2018, 18, 1408–1414. [Google Scholar] [CrossRef] [Green Version]
  98. Samoylova, M.L.; Nash, A.; Kuchibhatla, M.; Barbas, A.S.; Brennan, T.V. Machine perfusion of donor kidneys may reduce graft rejection. Clin. Transplant. 2019, 33, e13716. [Google Scholar] [CrossRef]
  99. Sevinc, M.; Stamp, S.; Ling, J.; Carter, N.; Talbot, D.; Sheerin, N.S. Comparison of the Outcome of Kidney Transplant After Pulsatile or Continuous Ex Vivo Hypothermic Machine Perfusion of Kidneys Donated After Cardiac Death: Analysis of Kidney Pairs. Transpl. Proc. 2019, 51, 1785–1790. [Google Scholar] [CrossRef]
  100. Wszola, M.; Domagala, P.; Ostaszewska, A.; Gorski, L.; Karpeta, E.; Berman, A.; Sobol, M.; Durlik, M.; Chmura, A.; Kwiatkowski, A. Time of Cold Storage Prior to Start of Hypothermic Machine Perfusion and Its Influence on Graft Survival. Transpl. Proc. 2019, 51, 2514–2519. [Google Scholar] [CrossRef]
  101. Krishnamoorthy, V.; Wunsch, S.; O’Connor, K.; Souter, M. Variation in Hypothermic Machine Perfusion Utilization and Clinical Outcomes for Deceased-Donor Kidneys. Am. J. Transplant. 2018, 18, 982. [Google Scholar]
  102. Singh, N.; Logan, A.; Schenk, A.; Bumgardner, G.; Brock, G.; El-Hinnawi, A.; Rajab, A.; Washburn, K. Machine perfusion of kidney allografts affects early but not late graft function. Am. J. Surg. 2022, 223, 804–811. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA.
Figure 1. PRISMA.
Jcm 12 03871 g001
Figure 2. Forest plot—overall rates of DGF in HMP compared to SCS [28,29,30,31,32,33,34,35,36,37,38,39,40,41].
Figure 2. Forest plot—overall rates of DGF in HMP compared to SCS [28,29,30,31,32,33,34,35,36,37,38,39,40,41].
Jcm 12 03871 g002
Figure 3. Forest plot—rates of DGF in DCD—HMP compared to SCS [28,32,33,35,40,42].
Figure 3. Forest plot—rates of DGF in DCD—HMP compared to SCS [28,32,33,35,40,42].
Jcm 12 03871 g003
Figure 4. Forest plot—rates of DGF in ECD—HMP compared to SCS [29,30,31,34,37,39].
Figure 4. Forest plot—rates of DGF in ECD—HMP compared to SCS [29,30,31,34,37,39].
Jcm 12 03871 g004
Figure 5. Forest plot—overall rates of PNF in HMP compared to SCS [28,29,30,31,32,33,34,35,41].
Figure 5. Forest plot—overall rates of PNF in HMP compared to SCS [28,29,30,31,32,33,34,35,41].
Jcm 12 03871 g005
Figure 6. Forest plot—rates of immune rejection at 1 year—HMP compared to SCS [31,35,37,41].
Figure 6. Forest plot—rates of immune rejection at 1 year—HMP compared to SCS [31,35,37,41].
Jcm 12 03871 g006
Figure 7. Forest plot—rates of graft survival at 1 year—HMP compared to SCS [28,29,30,31,35,36,37,39,41].
Figure 7. Forest plot—rates of graft survival at 1 year—HMP compared to SCS [28,29,30,31,35,36,37,39,41].
Jcm 12 03871 g007
Figure 8. Forest plot—rates of patient survival at 1 year—HMP compared to SCS [28,29,30,31,34,35].
Figure 8. Forest plot—rates of patient survival at 1 year—HMP compared to SCS [28,29,30,31,34,35].
Jcm 12 03871 g008
Figure 9. Forest plot—overall rates of DGF in HMP + O2 compared to SCS [43,44,45].
Figure 9. Forest plot—overall rates of DGF in HMP + O2 compared to SCS [43,44,45].
Jcm 12 03871 g009
Figure 10. Forest plot—overall rates of DGF in NRP compared to ICP [19,46,47].
Figure 10. Forest plot—overall rates of DGF in NRP compared to ICP [19,46,47].
Jcm 12 03871 g010
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ghoneima, A.S.; Sousa Da Silva, R.X.; Gosteli, M.A.; Barlow, A.D.; Kron, P. Outcomes of Kidney Perfusion Techniques in Transplantation from Deceased Donors: A Systematic Review and Meta-Analysis. J. Clin. Med. 2023, 12, 3871. https://doi.org/10.3390/jcm12123871

AMA Style

Ghoneima AS, Sousa Da Silva RX, Gosteli MA, Barlow AD, Kron P. Outcomes of Kidney Perfusion Techniques in Transplantation from Deceased Donors: A Systematic Review and Meta-Analysis. Journal of Clinical Medicine. 2023; 12(12):3871. https://doi.org/10.3390/jcm12123871

Chicago/Turabian Style

Ghoneima, Ahmed S., Richard X. Sousa Da Silva, Martina A. Gosteli, Adam D. Barlow, and Philipp Kron. 2023. "Outcomes of Kidney Perfusion Techniques in Transplantation from Deceased Donors: A Systematic Review and Meta-Analysis" Journal of Clinical Medicine 12, no. 12: 3871. https://doi.org/10.3390/jcm12123871

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

Article Metrics

Back to TopTop