This website uses only technical or equivalent cookies.
For more information click here.

Machine perfusion in organ transplantation

Special Issue 2 - August 2023 - Machine perfusion in organ transplantation

ABDOMINAL NORMOTHERMIC REGIONAL PERFUSION IN DONATION AFTER CIRCULATORY DEATH: ORGAN VIABILITY OR ORGAN PRESERVATION?

Authors

Key words: extracorporeal membrane oxygenation, kidney transplant, liver transplant, organ preservation, organ procurement
Publication Date: 2023-03-21

Abstract

Transplants from donation after circulatory death (DCD) provide a viable means to deal with the ever-growing shortage of donors but are burdened by a higher rate of complications and graft loss. Dismal results have prompted the application of stricter donor and graft selection criteria and the use of machine perfusion technologies, such as normothermic regional perfusion (NRP). In this review, we first describe the diffusion of NRP worldwide. Next, the role of NRP in liver transplantation is discussed, with a particular focus on graft selection during perfusion and posttransplant outcomes. Finally, we review the clinical studies reporting on NRP in kidney transplantation. The emerging use of NRP with complementary ex-situ machine perfusion is also described. NRP improves organ quality and maintenance before cold preservation, turns the DCD procedure into a more unhurried one, and allows the assessment of organ function following the warm ischemic injury. Moreover, it is beneficial for both the liver and the kidneys from the same donor.

Abbreviations

ALT : alanine transaminase

cDCD: controlled donation after circulatory death

DBD: donation after brain death

DCD: donation after circulatory death

DGF: delayed graft function

FMN: flavin mononucleotide

HOPE: hypothermic oxygenated perfusion

ITBL: Ischemic-type biliary lesions

KT: kidney transplant

LT: liver transplant

NMP: normothermic machine perfusion

NRP: normothermic regional perfusion

RCT: randomized controlled trial

SCS: static cold storage

SRR: super-rapid recovery

uDCD: uncontrolled donation after circulatory death

UK: United Kingdom

INTRODUCTION

Donation after circulatory death (DCD) provides a viable means to deal with the ever-growing shortage of donors. Nevertheless, transplants from DCD are traditionally burdened by a higher rate of complications and graft loss. Dismal results have prompted the application of stricter donor and graft selection criteria in some settings and the use of machine perfusion technologies in others 1.

DCD inevitably involves compromised hemodynamics in the agonal phase, followed by an obligatory additional circulatory standstill for the declaration of death. In super-rapid recovery (SRR), this is hurriedly followed by incision, aortic cannulation, and organ perfusion with hypothermic preservation solution. Normothermic regional perfusion (NRP) temporarily re-establishes blood flow following declaration of death, through arterial and venous cannulae placed in the femoral vessels or directly in the aorta and vena cava after rapid laparotomy (Fig. 1). Supraceliac aortic balloon occlusion prevents cerebral reperfusion during NRP and restricts perfusion to the abdomen 2. This way, NRP restores previously depleted energy substrates, clears by-products of anaerobic metabolism, and induces endogenous antioxidants, thus helping to improve organ quality and maintenance before cold preservation 3. Moreover, in contrast to SRR, NRP also allows the assessment of organ function following the warm ischemic injury 4.

Although some ethical concerns have been raised, especially with premortem cannulation, heparin administration, and potential brain reperfusion, NRP is currently spreading in many European countries5. To maintain the permanence principle for death, Manara et al. have recently suggested inserting a cannula in the ascending aorta to identify inadequate occlusion and divert any collateral flow away from the brain 6. Moreover, antemortem interventions in the potential donor should follow national legislation and are ethically acceptable if they do not add risk, harm, or discomfort to the patient 3.

In this review, we first describe the diffusion of NRP worldwide. Next, the role of NRP in liver transplantation (LT) is discussed, with a particular focus on graft selection during perfusion and posttransplant outcomes. Finally, we review the clinical studies reporting on NRP in kidney transplantation (KT). The emerging use of NRP with complementary ex-situ machine perfusion is also described.

DIFFUSION OF NRP

Currently, NRP is mandatorily applied in DCD organ recovery in 3 European countries (Italy, France, and Norway) and is permitted in 5 (Spain, United Kingdom, Belgium, the Netherlands, and Switzerland) 1,3,7,8. Moreover, a progressive increase in NRP use has been registered over time even in those countries where NRP is not mandatory. In Spain, NRP is currently far more frequent than SRR for liver recovery 9. Nevertheless, this practice is still less embraced in the United States, although good results have been reported recently in a series of 13 DCD donors maintained on NRP 2. A few cases have also been reported from Russia and Korea 7,10.

NRP IN LIVER TRANSPLANTS

Clinical studies on LTs with NRP are reported in Table I.

Results of LT with NRP

NRP has originally allowed recovery and utilization of uncontrolled DCD liver grafts (uDCD; Maastricht category II and IV) in Spain, France, and Italy. Then its use has been shifted to controlled DCD grafts (cDCD; Maastricht category III). NRP turns the cDCD procedure into a more unhurried one compared to SRR, thus enabling graft evaluation and even warm dissection. Ischemic-type biliary lesions (ITBL) are the leading cause of patient morbidity and early graft loss in LTs from DCD. Many retrospective studies have shown that NRP treatment is effective in preventing the occurrence of ITBL compared to SRR, but randomized trials comparing these two techniques have not been reported yet 9,11. Nevertheless, given the ever-increasing use of NRP instead of SRR, such clinical trials are unlikely to be carried out in the future.

Selection of livers during NRP

Different criteria are used for graft selection during NRP, and progressive evolution of both parameters and thresholds has been noticed over time (Tab. II). A combination of macroscopic and microscopic assessment, alanine transaminase (ALT) levels, and lactate in perfusate are used to assess the suitability of the liver for transplantation in most protocols. All of these parameters inform about both liver viability and quality of the perfusion and have contributed to the selection of LT series with excellent results. Nevertheless, no strong correlation has been found between each parameter and the transplantation outcome 12,13. According to a recent review, the most used acceptation criterion in cDCD is the macroscopic aspect, while in uDCD ALT level is considered the most reliable 4. This attitude has been partially reflected in a recent Italian survey, where macroscopic assessment was highly considered in the opinion of the participants, along with the stability of NRP perfusion conditions 14. Although transaminase release is a widely accepted marker of liver injury, its cut-off has, however, been modified from initially 3-4 times the normal values to upper thresholds reported in the most recent series 11,12. Lactate clearance has been proposed as a parameter to assess liver function, as in normothermic machine perfusion, with a downward lactate trend indicating a well-functioning liver 4. However, Watson et al. noted that lactate leaking back from non-perfused areas in the donor decreases the reliability of this parameter as an indicator of liver function 15. Wang et al. have recently analysed flavin mononucleotide (FMN) in the perfusate during NRP and found that FMN levels were significantly higher in those livers that were declined for transplantation 16. Unfortunately, no correlation was made with the LT outcomes in the livers that were accepted. Further insight on this topic will probably be given by a nonrandomized trial on viability assessment during NRP, which is currently ongoing in France (NCT05361044).

NRP vs machine perfusion

Various dynamic ex-situ preservation strategies have been explored to ameliorate the outcomes of DCD livers, including hypothermic oxygenated perfusion (HOPE) and normothermic machine perfusion (NMP). Preference for in-situ NRP or ex-situ techniques varies among centres and countries. NRP is most frequently undertaken in tertiary hospitals with extracorporeal membrane oxygenation devices and cardiothoracic intensive care units, although good results have also been reported with mobile NRP teams 17,18. A few studies have compared NRP with HOPE or NMP, but no randomized trials have been published. In a large-scale international multicentric study, the utilization rate was significantly lower in the NRP group, despite shorter warm ischemic times and lower donor age compared to the HOPE group. However, after propensity-score adjustment of donor-recipient combinations, both strategies achieved similar posttransplant outcomes 19. In a single-centre retrospective analysis from the United Kingdom (UK), the NRP group had a lower incidence of cholangiopathy than static cold storage, but the same benefit was not achieved with NMP (NMP vs NRP: hazard ratio 3.5, p = 0.02) 17. Conversely, a multicentric study comparing NRP cases from France with NMP cases from the UK, Germany, Spain, and Belgium, failed to show any significant difference in the incidence of non-anastomotic biliary strictures (1.5 vs 2.9%; p > 0.99) and 30-day graft loss (4.4 vs 8.8%; p = 0.40) between the two groups 20.

NRP and subsequent HOPE or NMP

Prolonged cold ischemia and indication for retransplantation were found to be independent risk factors for graft loss among 545 DCD livers treated with NRP in Spain 9. These data support a potential role for complimentary ex-situ perfusion preservation for those cases with prolonged cold ischemia and/or technically complex recipients. The combined use of NRP with subsequent HOPE was first proposed in 2016 by our group to face the detrimental effects of the 20-min stand-off period in Italy, thus providing safe prolonged preservation and further reconditioning to the DCD livers 21. Although a direct comparison between NRP with subsequent HOPE and NRP only is still lacking in the Italian population, some indirect evidence exists to support this approach 14. In a recent multicentric analysis, the combined protocol has yielded good results compared to a static-preserved comparator group from the UK, despite the higher risk profile in Italy 12. Moreover, the same Italian cohort had shown similar results to the benchmark outcomes in LT from DCD22. The combined use of NRP and NMP was first reported by Pavel et al. 23. More recently, Ghinolfi et al. have proposed a flow-chart, where machine perfusion is used following the initial DCD liver evaluation during NRP. While HOPE is suggested for cases partially fulfilling the criteria, the authors recommend NMP when the criteria are not fulfilled 24. This proposal sounds promising but is still mainly based on a small number of uDCD cases, and only a few centres currently use both HOPE and NMP in Italy 14. The results of a currently ongoing randomized trial in Italy between sequential HOPE and NMP after NRP (NCT04744389) will hopefully provide further insight into this matter.

NRP IN KIDNEY TRANSPLANTS

A few clinical series on KT after NRP have been published, mainly from cDCD donors (Tab. III).

Results of KTs with NRP

Foss et al. compared the outcome of 14 DCD kidneys recovered with NRP with 163 transplants from donation after brain death (DBD) and observed no differences in delayed graft function (DGF) and 1-year graft survival between the groups 25. Similarly, Miñambres et al. reported nonsignificant differences in DGF and short-term graft survival comparing DCD kidneys treated with NRP with DBD controls 26. However, these studies did not compare the use of NRP with the widespread SRR. Ramirez et al. found that DCD kidneys treated with NRP had a lower rate of DGF than those with SRR 27. In a recent Spanish nationwide propensity score analysis, Padilla et al. have found that NRP was associated with improved rates of DGF and 1-year graft loss compared to SRR 28.

Selection of kidneys during NRP

For the acceptance of the kidneys during NRP, published reports mention macroscopic aspect, microscopic findings, and urine production. However, the absence of urine output is frequent during NRP and should not per se lead to organ discard 3. Rodríguez-Villar et al. investigated the evolution of biochemical parameters during NRP between accepted and discarded kidneys in 38 uDCD donors. Neither creatinine nor lactate sequential values was a useful tool to predict kidney allocation. Nevertheless, the authors did not correlate any posttransplant outcome with these variables 29. Ravaioli et al. reported the preliminary experience with sequential NRP and HOPE in 10 KTs from cDCD in Italy. They reported a 30% incidence of DGF and did not find any correlation with creatinine or lactate values during NRP. However, they found that lactate levels in the HOPE perfusate were significantly higher in those cases developing DGF 30. Marginal kidneys are usually assessed histologically with the Karpinski score, which however does not take into account the ischemic insult 30. In this context, Zagni et al. have recently reported that ischemic alterations of the proximal tubule are correlated with functional recovery in DCD kidneys 31. Centres using ex-situ hypothermic perfusion report using renal resistance for further selection, though studies outside the NRP field have shown kidneys should not be discarded based upon renal resistance only 32,33.

CONCLUSIONS

NRP is beneficial for both the liver and the kidneys from the same donor. Therefore, NRP improves organ quality and maintenance before cold preservation, turns the DCD procedure into a more unhurried one, and allows the assessment of organ function following the warm ischemic injury. Different parameters inform about both graft viability and quality of the perfusion, but no strong correlation has been found between each parameter and the transplantation outcome. Nevertheless, a combination of macro-microscopic assessment, biochemical, and perfusion parameters has contributed to the selection of liver and kidney transplant series with excellent results.

Acknowledgments

The Authors want to thank Elisa Lodi for her support in figure editing.

Conflict of interest statement

The Authors declare no conflict of interest.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Authors’ contributions

RDC performed data collection, interpreted data, and wrote the paper; LC, MM, LP, RC, and IV performed data collection and reviewed the paper; AL and LDC critically reviewed the paper.

Ethical consideration

The present study did not imply any direct investigation on humans or animals. Formal consent was not required.

Figures and tables

Figure 1. NRP circuit and advantages. The abdominal NRP circuit relies on extracorporeal membrane oxygenation technology and includes a pump, an oxygenator, and a heater. The blood is pumped through arterial and venous cannulae in the femoral vessels. Supraceliac aortic balloon occlusion prevents cerebral reperfusion. aNRP: abdominal normothermic regional perfusion.

Author, year Country DCD type, n Utilization rate (from NRP to LT) EAD PNF AKI ITBL Graft survival (follow-up) Patient survival (follow-up)
uDCD
Fondevila, 2012 34 Spain uDCD: 34 34/290 (12%) N/A N/A N/A 8% 70% (1y) 82% (1y)
Savier, 2015 35 France uDCD: 13 13/183 (7%) 31% 23% N/A 8% 69% (1y) 85% (1y)
Justo, 2020 36 Spain uDCD: 75 N/A N/A 8% 20% 4% 78.3% (1y) excluding PNF 82% (1y) excluding PNF
cDCD (multicentre/national)
Watson, 2019 11 UK cDCD: 43 61% 12% 0% N/A 0% 97.7% (90d) 100% (90d)
Savier, 2020 37 France cDCD: 50 20% 18% N/A 26% 2% 88% (2y) 90% (2y)
De Carlis, 2021 12 Italy cDCD: 44 85% N/A 5% 36% 2% 91% (2y) 98% (2y)
Hessheimer, 20219 Spain cDCD: 545 70% 15% 3% N/A 1% 90% (1y) 92% (1y)
Sellers, 2022 2 US cDCD: 13 N/A 23% 0% N/A 0% 92% (439d) 92% (439d)
Mixed series (uDCD and cDCD)
De Carlis, 2018 38 Italy cDCD: 6 20/25 24% 10% 30% 10% 85% 95%
uDCD: 14 (80%) (1y) (1y)
Ghinolfi, 2020 24 Italy cDCD: 7 18/31 28% 0% 28% 6% 94% 94%
uDCD: 11 (58%) (15mo.) (15mo.)
Table I. Main series of LT from controlled and uncontrolled DCD donors maintained on NRP.
Author, year Type Pump flow Lactate Transaminase Macroscopical aspect Liver biopsy
Fondevila, 200739 uDCD >1.7 L/min - Initial < 3 ULN Before/after cold flush -
(>4h) Final > 4 ULN
Oniscu, 201413 cDCD 1.7-4 L/min/m2 - Initial ALT < 3 ULN - -
(60-120 min) Final ALT < 4 ULN
Savier, 201535 uDCD efficient flow for > 240 min - ALT at 2h < 200 IU/L - MaS < 20%
De Carlis, 201740 uDCD - Stable or downward ALT < 1000 IU/L Color, surface, margins, consistency MaS < 30%
cDCD Ishak 0-1
Watson, 201911 cDCD 2.5-3 L/min Fall is encouraging ALT < 200 IU/L Steatosis -
ALT < 500 IU/L
Savier, 202037 cDCD > 60 min - AST-ALT < 200 IU/L - MaS < 20%
Hesseimer, 20219 cDCD 2.2-2.4 L/min/m2 Ideally downward Ideally stable and < 200 IU/L Liver, gallbladder, bile duct and bowel -
(60-120 min)
De Carlis, 202112 cDCD 1.7-3 L/min/m2 (ideally 120 min, no upper limit if stable) Ideally stable or downward Ideally final ALT < 1000 IU/L Perfusion, congestion Ideally MaS < 30% Ishak 0-2
Table II. Main selection criteria of the liver during NRP.
Author, year Study type n Donor fWIT (min) MP PNF DGF Graft survival (follow-up) Patient survival (follow-up)
Foss, 2018 25 Single-centre, retrospective, observational NRP: 14 NRP: 26.5 (20-49) No NRP: 0% NRP:7.1% NRP: 93% N/A
DBD: 163 DBD: 0% DBD: 4.9% DBD: 95% (1y)
Miñambres, 2017 26 Single-centre, retrospective, observational NRP: 37 NRP: 12 (10-19) No NRP: 5% NRP: 27% NRP: 91.8% N/A
DBD: 36 DBD: 0% DBD: 33.3% DBD: 97.2% (18mo.)
Ravaioli, 2018 30 Single-centre, retrospective, observational NRP: 5 NRP: 151.2 (40-325) HOPE NRP: 0% NRP: 30% NRP: 100% (6mo.) NRP: 100% (6mo)
Padilla, 2020 28 Nation-wide, retrospective, observational NRP: 865 NRP: 13 (10-17) NRP: 15.9% NRP: 4.8% NRP: 30.3% NRP: 93.1% NRP: 97.6%
SRR: 1437 SRR: 18 (13-3) SRR: 7.3% SRR: 4.4% SRR: 48.4% SRR: 91.5% (1y) SRR: 95.6% (1y)
Ramirez, 2021 27 Single-centre, retrospective, observational NRP: 22 NRP: 10 (10-35) No NRP: 4,55% NRP: 36.36% NRP: 91% NRP: 77.27%
SRR: 62 SRR: 15 (11-28) SRR: 6,45% SRR: 46.77% SRR: 87% SRR: 88.71%
DBD: 98 DBD: 10,20% DBD: 20.41% DBD: 84,4% (15mo.) DBD: 85.71% (1y)
Table III. Clinical series of KTs from cDCD with NRP.

References

  1. Hessheimer A, Polak W, Antoine C. Regulations and procurement surgery in DCD liver transplantation: expert consensus guidance from the International Liver Transplantation Society. Transplantation. 2021;105:945-951. doi:https://doi.org/10.1097/TP.0000000000003729
  2. Sellers M, Nassar A, Alebrahim M. Early United States experience with liver donation after circulatory determination of death using thoraco-abdominal normothermic regional perfusion: a multi-institutional observational study. Clin Transplant. 2022;36. doi:https://doi.org/10.1111/ctr.14659
  3. Jochmans I, Hessheimer A, Neyrinck A. Consensus statement on normothermic regional perfusion in donation after circulatory death: report from the European Society for Organ Transplantation’s Transplant Learning Journey. Transpl Int. 2021;34:2019-2030. doi:https://doi.org/10.1111/tri.13951
  4. Schurink I, van de Leemkolk F, Fondevila C. Donor eligibility criteria and liver graft acceptance criteria during normothermic regional perfusion: a systematic review [published online ahead of print, 2022 May 23]. Liver Transpl. Published online 2022. doi:https://doi.org/10.1002/lt.26512
  5. Dalle Ave A, Shaw D, Bernat J. Ethical issues in the use of extracorporeal membrane oxygenation in controlled donation after circulatory determination of death. Am J Transplant. 2016;16:2293-2299. doi:https://doi.org/10.1111/ajt.13792
  6. Manara A, Shemie S, Large S. Maintaining the permanence principle for death during in situ normothermic regional perfusion for donation after circulatory death organ recovery: a United Kingdom and Canadian proposal. Am J Transplant. 2020;20:2017-2025. doi:https://doi.org/10.1111/ajt.15775
  7. Lomero M, Gardiner D, Coll E. Donation after circulatory death today: an updated overview of the European landscape. Transpl Int. 2020;33:76-88. doi:https://doi.org/10.1111/tri.13506
  8. Melandro F, Basta G, Torri F. Normothermic regional perfusion in liver transplantation from donation after cardiocirculatory death: technical, biochemical, and regulatory aspects and review of literature. Artif Organs. 2022;46:1727-1740. doi:https://doi.org/10.1111/aor.14330
  9. Hessheimer A, de la Rosa G, Gastaca M. Abdominal normothermic regional perfusion in controlled donation after circulatory determination of death liver transplantation: outcomes and risk factors for graft loss. Am J Transplant. 2022;22:1169-1181. doi:https://doi.org/10.1111/ajt.16899
  10. Lee J, Hong S, Oh C. Kidney transplantation from a donor following cardiac death supported with extracorporeal membrane oxygenation. J Korean Med Sci. 2012;27:115-119. doi:https://doi.org/10.3346/jkms.2012.27.2.115
  11. Watson C, Hunt F, Messer S. In-situ normothermic perfusion of livers in controlled circulatory death donation may prevent ischemic cholangiopathy and improve graft survival. Am J Transplant. 2019;19:1745-1758. doi:https://doi.org/10.1111/ajt.15241
  12. De Carlis R, Schlegel A, Frassoni S. How to preserve liver grafts from circulatory death with long warm ischemia? A retrospective italian cohort study with normothermic regional perfusion and hypothermic oxygenated perfusion. Transplantation. 2021;105:2385-2396. doi:https://doi.org/10.1097/TP.0000000000003595
  13. Oniscu G, Randle L, Muiesan P. In-situ normothermic regional perfusion for controlled donation after circulatory death – the United Kingdom experience. Am J Transplant. 2014;14:2846-2854. doi:https://doi.org/10.1111/ajt.12927
  14. De Carlis R, Lauterio A, Centonze L. Current practice of normothermic regional perfusion and machine perfusion in donation after circulatory death liver transplants in Italy. Updates Surg. 2022;74:501-510. doi:https://doi.org/10.1007/s13304-022-01259-9
  15. Watson C, Jochmans I. From “Gut Feeling” to objectivity: machine preservation of the liver as a tool to assess organ viability. Curr Transplant Rep. 2018;5:72-81. doi:https://doi.org/10.1007/s40472-018-0178-9
  16. Wang L, Thompson E, Bates L. Flavin mononucleotide as a biomarker of organ quality-a pilot study. Transplant Direct. 2020;6. doi:https://doi.org/10.1097/TXD.0000000000001046
  17. Gaurav R, Butler A, Kosmoliaptsis V. Liver Transplantation outcomes from controlled circulatory death donors: SCS vs in-situ NRP vs ex-situ NMP. Ann Surg. 2022;275:1156-1164. doi:https://doi.org/10.1097/SLA.0000000000005428
  18. Pérez Redondo M, Alcántara Carmona S, Fernández Simón I. Implementation of a mobile team to provide normothermic regional perfusion in controlled donation after circulatory death: pilot study and first results. Clin Transplant. 2020;34. doi:https://doi.org/10.1111/ctr.13899
  19. Muller X, Mohkam K, Mueller M. Hypothermic oxygenated perfusion versus normothermic regional perfusion in liver transplantation from controlled donation after circulatory death: first international comparative study. Ann Surg. 2020;272:751-758. doi:https://doi.org/10.1097/SLA.0000000000004268
  20. Mohkam K, Nasralla D, Mergental H. In situ normothermic regional perfusion versus ex situ normothermic machine perfusion in liver transplantation from donation after circulatory death. Liver Transpl. Published online 2022. doi:https://doi.org/10.1002/lt.26522
  21. De Carlis L, De Carlis R, Lauterio A. Sequential use of normothermic regional perfusion and hypothermic machine perfusion in donation after cardiac death liver transplantation with extended warm ischemia time. Transplantation. 2016;100:e101-e102. doi:https://doi.org/10.1097/TP.0000000000001419
  22. Schlegel A, van Reeven M, Croome K. A multicentre outcome analysis to define global benchmarks for donation after circulatory death liver transplantation. J Hepatol. 2022;76:371-382. doi:https://doi.org/10.1016/j.jhep.2021.10.004
  23. Pavel M, Reyner E, Fuster J. Liver transplantation from type II donation after cardiac death donor with normothermic regional perfusion and normothermic machine perfusion. Trasplante hepático con injerto de donante en asistolia tipo 2 con perfusión regional normotérmica y máquina de perfusión normotérmica. Cir Esp (Engl Ed). 2018;96:508-513. doi:https://doi.org/10.1016/j.ciresp.2018.06.016
  24. Ghinolfi D, Dondossola D, Rreka E. Sequential use of normothermic regional and ex-situ machine perfusion in donation after circulatory death liver transplant. Liver Transpl. 2021;27:385-402. doi:https://doi.org/10.1002/lt.25899
  25. Foss S, Nordheim E, Sørensen D. First Scandinavian protocol for controlled donation after circulatory death using normothermic regional perfusion. Transplant Direct. 2018;4. doi:https://doi.org/10.1097/TXD.0000000000000802
  26. Miñambres E, Suberviola B, Dominguez-Gil B. 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. doi:https://doi.org/10.1111/ajt.14214
  27. Ramirez P, Vázquez D, Rodríguez G. Kidney transplants in controlled donation following circulatory death, or maastricht type III donors, with abdominal normothermic regional perfusion, optimizing functional outcomes. Transplant Direct. 2021;7. doi:https://doi.org/10.1097/TXD.0000000000001174
  28. Padilla M, Coll E, Fernández-Pérez C. 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. doi:https://doi.org/10.1111/ajt.16622
  29. Rodríguez-Villar C, Paredes D, Roque R. Clinical utility and evolution of donor serum lactate during normothermic regional perfusion in uncontrolled donation after circulatory death. Transplant Proc. 2021;53:2650-2654. doi:https://doi.org/10.1016/j.transproceed.2021.05.013
  30. Ravaioli M, De Pace V, Comai G. 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(11):1233-1244. doi:https://doi.org/10.1111/tri.13311
  31. Zagni M, Croci G, Cannavò A. Histological evaluation of ischemic alterations in donors after cardiac death: a useful tool to predict post-transplant renal function. Clin Transplant. 2022;36. doi:https://doi.org/10.1111/ctr.14622
  32. Parikh C, Hall I, Bhangoo R. Associations of perfusate biomarkers and pump parameters with delayed graft function and deceased donor kidney allograft function. Am J Transplant. 2016;16:1526-1539. doi:https://doi.org/10.1111/ajt.13655
  33. de Vries E, Hoogland E, Winkens B. Renovascular resistance of machine-perfused DCD kidneys is associated with primary nonfunction. Am J Transplant. 2011;11:2685-2691. doi:https://doi.org/10.1111/j.1600-6143.2011.03755.x
  34. Fondevila C, Hessheimer A, Flores E. Applicability and results of Maastricht type 2 donation after cardiac death liver transplantation. Am J Transplant. 2012;12:162-170. doi:https://doi.org/10.1111/j.1600-6143.2011.03834.x
  35. Savier E, Dondero F, Vibert E. First experience of liver transplantation with type 2 donation after cardiac death in France. Liver Transpl. 2015;21:631-643. doi:https://doi.org/10.1002/lt.24107
  36. Justo I, Nutu A, García-Conde M. Incidence and risk factors of primary non-function after liver transplantation using grafts from uncontrolled donors after circulatory death. Clin Transplant. 2021;35. doi:https://doi.org/10.1111/ctr.14134
  37. Savier E, Lim C, Rayar M. Favorable outcomes of liver transplantation from controlled circulatory death donors using normothermic regional perfusion compared to brain death donors. Transplantation. 2020;104:1943-1951. doi:https://doi.org/10.1097/TP.0000000000003372
  38. De Carlis R, Di Sandro S, Lauterio A. Liver grafts from donors after circulatory death on regional perfusion with extended warm ischemia compared with donors after brain death. Liver Transpl. 2018;24:1523-1535. doi:https://doi.org/10.1002/lt.25312
  39. Fondevila C, Hessheimer A, Ruiz A. Liver transplant using donors after unexpected cardiac death: novel preservation protocol and acceptance criteria. Am J Transplant. 2007;7:1849-1855. doi:https://doi.org/10.1111/j.1600-6143.2007.01846.x
  40. De Carlis R, Di Sandro S, Lauterio A. Successful donation after cardiac death liver transplants with prolonged warm ischemia time using normothermic regional perfusion. Liver Transpl. 2017;23:166-173. doi:https://doi.org/10.1002/lt.24666

Downloads

Authors

Riccardo De Carlis - Department of General Surgery and Transplantation, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy

Leonardo Centonze - Department of General Surgery and Transplantation, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy

Michele Migliorini - Department of General Surgery and Transplantation, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Surgical Sciences, University of Pavia, Pavia, Italy

Ludovica Pitoni - Department of General Surgery and Transplantation, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Medicine and Surgery, University of Milano- Bicocca, Milan, Italy

Raffaele Cerchione - Department of General Surgery and Transplantation, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of General Surgery, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy

Andrea Lauterio - Department of General Surgery and Transplantation, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Medicine and Surgery, University of Milano- Bicocca, Milan, Italy

Luciano De Carlis - Department of General Surgery and Transplantation, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Medicine and Surgery, University of Milano- Bicocca, Milan, Italy

How to Cite
[1]
De Carlis, R., Centonze, L., Migliorini, M., Pitoni, L., Cerchione, R., Lauterio, A. and De Carlis, L. 2023. ABDOMINAL NORMOTHERMIC REGIONAL PERFUSION IN DONATION AFTER CIRCULATORY DEATH: ORGAN VIABILITY OR ORGAN PRESERVATION?. European Journal of Transplantation. 1, 2 (Mar. 2023), 113–120. DOI:https://doi.org/10.57603/EJT-013.
  • Abstract viewed - 1347 times
  • PDF downloaded - 274 times