Semin Liver Dis 2020; 40(03): 264-281
DOI: 10.1055/s-0040-1713118
Review Article

Machine Perfusion: Cold versus Warm, versus Neither. Update on Clinical Trials

E. Bonaccorsi-Riani*
1   Abdominal Transplant Unit, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
2   Pôle de Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Brussels, Belgium
,
I.M.A. Brüggenwirth*
3   Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
,
J.E. Buchwald
4   Division of Transplant, Department of Surgery, UMass Memorial Medical Center, University of Massachusetts, Worcester, Massachusetts
,
S. Iesari
2   Pôle de Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Brussels, Belgium
5   Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
,
P.N. Martins
4   Division of Transplant, Department of Surgery, UMass Memorial Medical Center, University of Massachusetts, Worcester, Massachusetts
› Author Affiliations
 

Abstract

Machine perfusion (MP) preservation is potentially one of the most significant improvements in the field of liver transplantation in the last 20 years, and it has been considered a promising strategy for improved preservation and ex situ evaluation of extended criteria donor (ECD) organs. However, MP preservation adds significant cost and logistical considerations to liver transplantation. MP protocols are mainly classified according to the perfusion temperature with hypothermic machine perfusion (HMP) and normothermic machine perfusion (NMP) being the two categories most studied so far. After extensive preclinical work, MP entered the clinical setting, and there are now several studies that demonstrated feasibility and safety. However, because of the limited quality of clinical trials, there is no compelling evidence of superiority in preservation quality, and liver MP is still considered experimental in most countries. MP preservation is moving to a more mature phase, where ongoing and future studies will bring new evidence in order to confirm their superiority in terms of clinical outcomes, organ utilization, and cost-effectiveness. Here, we present an overview of all preclinical MP studies using discarded human livers and liver MP clinical trials, and discuss their results. We describe the different perfusion protocols, pitfalls in MP study design, and provide future perspectives. Recent trials in liver MP have revealed unique challenges beyond those seen in most clinical studies. Randomized trials, correct trial design, and interpretation of data are essential to generate the data necessary to prove if MP will be the new gold standard method of liver preservation.


#

Over the past five decades, liver transplantation (LT) has become the standard of care for end-stage liver diseases. This success led the liver transplant community to broaden indications for LT.[1] [2] The direct consequence of this progress is the increasing number of patients on the waiting list, which worsens the chronic problem of organ shortage. In fact, the amount of LTs performed is directly affected by allograft availability, as reflected in the 10 to 20% mortality rate on the waiting list.[3] [4] To overcome this problem, transplant surgeons are required to accept allografts from donors with extended criteria (ECD), such as elderly donors, obese donors who regularly provide steatotic grafts, and donors after cardiac death (DCD). It is well known that these organs do not tolerate long periods of static cold storage (SCS), the current standard of liver graft preservation, as they are more susceptible to ischemia and reperfusion injury (IRI), have a higher incidence of primary nonfunction (PNF), and are more prone to develop biliary complications, which leads to a greater number of graft failures with a higher rate of retransplantation compared with non-ECD organs.[5] Ex situ organ machine perfusion (MP) can be an alternative preservation modality to improve the quality and function of ECD allografts. The concept of ex situ organ perfusion, idealized by Charles Lindbergh and Alexis Carrel, who built the first MP prototype in 1935,[6] [7] was abandoned after few experiments in the late 1960s performed in the United States and Europe[8] [9] due to improved static cold preservation solutions.[10] [11] The growing demand for ECD organs refueled the interest of the transplant community in MP preservation of ex situ allografts. In the past decade, MP has become a hot topic in organ transplantation and research groups worldwide have investigated different MP modalities to optimize preservation.[12] [13] [14] In this current review, we present an overview of all clinical liver MP trials and MP studies using discarded human liver grafts. We analyzed recent clinical experience with liver MP, taking into account the different protocols with respect to temperature, type of perfusion, duration of perfusion, and post-LT outcome.

Types of Perfusion

Ex situ liver perfusion protocols can be mainly defined according to the perfusion temperature used ([Fig. 1]). In hypothermic machine perfusion (HMP) protocols, the temperature is maintained at 4 to 10°C, while it is around 20 to 30°C in subnormothermic MP studies. In normothermic machine perfusion (NMP), the temperature is kept at physiological levels (37°C).[15] Innovative modalities of organ preservation and perfusion include super cooling organ preservation (−4°C) and hyperthermic perfusion (> 37°C). However, such strategies are still in the experimental stage.[16] [17] [18] While HMP can be performed with or without active oxygenation, NMP always requires oxygenation. Other protocol variations include the perfusion route (through the hepatic artery, portal vein, or both vessels) and the number and kind of pumps (pulsatile vs. nonpulsatile) or via gravitational force like the portal vein perfusion in the OrganOx metra (OrganOx Limited) device.

Zoom Image
Fig. 1 Graph representing the dynamics of the metabolism rate according to the machine perfusion temperature used. Based on Van't Hoff's principle (expressed as Q10 = (k2/k1) 10/(t2-t1)) the metabolism rate is much lower in the cold (0–12°C). Types of organ preservation and machine perfusion proposed in accordance with temperature intervals: Hyper MP, hyperthermic machine perfusion (38–42°C); NMP, normothermic machine perfusion (35–38°C); SNMP, subnormothermic machine perfusion (24–34°C); MMP, midthermic machine perfusion (13–24°C); HMP, hypothermic machine perfusion (0–12°C); SCS, static cold storage (0–8°C), super-cooling preservation (−4 to 0°C). (Adapted from Karangwa et al.[15])

#

Hypothermic Machine Perfusion

For years, cooling has been the cornerstone in organ preservation. The concept of hypothermic preservation is largely based on the idea that the graft is maintained in a hypometabolic state, thereby decreasing energy utilization during ischemia and preserving essential mechanisms to regenerate ATP upon reperfusion. However, due to persistent low-grade cellular metabolism at 4°C, SCS preservation on ice leads to anaerobic metabolism within short time due to the lack of oxygen and substrates. In contrast, the concept of HMP is to maintain viability and aerobic metabolism prior to implantation of the graft by providing oxygen while the organ’s metabolism is limited.[19] The underlying mechanisms in HMP mainly involve resuscitation of mitochondria. Even a short period of 2 hours of oxygenated HMP can switch mitochondria from ischemic to fully recharged cellular energy levels.[19] [20]

MP was traditionally designed as a continuous approach, starting directly after organ procurement. However, in the last decade, it became clear that end-ischemic (after SCS) MP is an attractive and simpler approach, avoiding transportation of the perfusion devices and extra perfusion personnel to the donor hospital. Livers are preserved by SCS after procurement and are then subjected to end-ischemic MP upon arrival at the recipient center. Even after relatively longer periods of SCS, end-ischemic HMP can protect liver grafts.[21] An additional advantage of HMP is that in case of pump failure, the liver can easily be restored to traditional SCS. Also, since the organ is in a hypometabolic state, it produces lesser waste products and requires less intensive labor by perfusionists when compared with NMP. A disadvantage of HMP used to be the lack of reliable prediction methods of organ function. However, a recent study by Muller et al shows that real-time fluorometric analysis of perfusate flavin, a marker of mitochondrial injury, can be predictive of graft function and loss during cold MP.[22] This achievement has high clinical relevance, and real-time estimation of liver function during cold preservation could substantially increase safe utilization of liver grafts.

Prospective Clinical Studies Using Hypothermic Machine Perfusion

In 2010, Guarrera et al were the first to report successful transplantation of 20 ECD donation after brain death (DBD) human livers after HMP via the portal vein and hepatic artery without active oxygenation ([Table 1]).[23] Because this was a nonrandomized trial comparing a series of machine-perfused livers with historical controls, they were only able to demonstrate safety of end-ischemic HMP of human liver grafts during 3 to 7 hours. This was also the first study to describe that the HMP system is user-friendly, such as that MP pressures and temperature remained stable, and little intervention from the perfusionist was needed. They found diminished peak aspartate aminotransferase (AST) and alanine aminotransferase (ALT), shorter hospital length of stay, and fewer biliary complications in the HMP-treated grafts compared with the matched SCS group. In the years following, HMP was increasingly used in kidney transplantation worldwide, but clinical implantation in LT remained investigational.[24] Five years after publishing their first clinical series, Guarrera et al reported successful transplantation of 31 “orphan” ECD livers after preservation by HMP in a nonrandomized fashion.[25] The term orphan was denoted to any liver turned down by all centers within the donor's originating United Network for Organ Sharing region, requiring more complex allocation management and expected longer cold ischemia time (CIT). Their study shows good outcomes with 84% graft survival 1 year after transplantation. In addition, biliary complications, often described as the “Achilles heel” of LT, occurred significantly less in the HMP group compared with the SCS historical control group. Better preservation of the arterial vasculature lining the biliary tree by providing a continuous supply of oxygen, better distribution of the perfusion fluid, and washout of waste products is suggested to play a role.[26] In the same year, Dutkowski et al reported on a nonrandomized study of 25 DCD livers treated by oxygenated HMP (HOPE) through the portal vein only.[27] Lower peak ALT, lesser cases of intrahepatic cholangiopathy and biliary complications, and 1-year graft survival of 90% versus 69% were observed in the HOPE group compared with the SCS historical control group. Although conclusive evidence of a beneficial effect of HMP on reducing biliary injury after transplantation could not yet be provided, these first results seemed promising. In 2017, van Rijn and colleagues published a nonrandomized clinical study of end-ischemic dual HOPE (DHOPE) in 10 DCD liver grafts. Compared with SCS alone, DHOPE seemed to provide better preservation of DCD liver grafts, resulting in reduced graft injury and improved early graft function. Noteworthy, none of the 10 DHOPE-preserved livers required retransplantation for nonanastomotic biliary strictures (NAS), compared with 5 out of 20 in the control group.[28] The potential benefit of DHOPE to better preserve the biliary tree is investigated in a randomized clinical trial (RCT), initiated by Groningen et al in 2015 (clinicaltrials.gov; NCT02584283) and of which the results are expected mid-2020. In collaboration between liver transplant centers in the Netherlands, the United Kingdom, and Belgium, this trial investigates the impact of DHOPE preservation versus SCS in DCD liver grafts. The primary outcome of the DHOPE-DCD trial is the development of NAS within 6 months after LT. Single HOPE is investigated by the prospective HOPE-ECD-DBD clinical trial started in 2017 by the University of Aachen. In this RCT using DBD livers only, 1 to 2 hours of single HOPE preservation is compared with the traditional SCS (clinicaltrials.gov; NCT03124641).

Table 1

Clinical trials on ex situ liver hypothermic machine perfusion

Author or organization

Year

Donor type (DCD/DBD)

N total (HMP/SCS)

Perfusion characteristics

Perfusate

Total time of preservation (min) (range)

Endpoints

Outcome (HMP vs. SCS)

PI: Stefania Camagni, Bergamo, Italy[a]

Estimated completion date: 2022

Not reported

20 (20/0)

Device not reported.

HA < 30 mm Hg/PV < 5 mm Hg.

Time on machine: 4 h

UW-MPS

Results awaited

Primary: incidence of PRS

Secondary: entity of IRI, incidence of EAD

Results awaited

PI: Mickael Lesurtel, Lyon, France[b]

Estimated completion date: 2022

DBD

266 (133/133)

Device: Liver Assist.

PV only.

Time on machine: 1–4 h

UW-MPS

Results awaited

Primary: incidence of EAD

Secondary: MEAF score, L-GrAFT, metabolic profiling, PRS. 90-d morbidity/mortality, length of hospital stay, MCRP within 1 y, 3-mo/1-y graft/patient survival, hospital costs

Results awaited

PI: Matteo Ravaioli, Bologna, Italy[c]

Estimated completion date: 2021

DBD

110

Device not reported.

Oxygenated (500–600 mm Hg)

UW-MPS

Results awaited

Primary: incidence of EAD

Secondary: surgical complications, liver function at 6/12 mo, patient survival at 6/12 mo

Results awaited

PI: Stefania Camagni, Bergamo, Italy[d]

Estimated completion date: 2021

DCD/DBD

20

Device not reported.

HA 25–30 mm Hg/PV < 5 mm Hg.

Time on machine: 1 h.

Oxygenated (50–70 kPa)

UW-MPS

Results awaited

Primary: incidence of EAD

Secondary: Dindo-Clavien complications, ischemic cholangiopathy, length of hospital stay, 30-d/1-y graft/patient survival

Results awaited

Organ Recovery Systems[e]

Estimated completion date: 2021

Not reported

140

Device: LifePort Liver Transporter

Vasosol

Results awaited

Primary: incidence of EAD

Results awaited

PI: Robert Porte, Groningen, The Netherlands[f]

Completion date: 2019

DCD

156 (78/78)

Device: Liver Assist

HA 25 mm Hg/PV 5 mm Hg.

0.5 mL/min 100% O2

Time on machine: 2 h

UW-MPS

Results awaited

Primary: incidence of NAS at 6 mo.

Secondary: graft/patient survival, PNF, IPF, recipient hemodynamics during LT, hospital length of stay, postoperative complications, liver function and injury markers, costs of treatment, quality of life

Results awaited

PI: Georg Lurje (Aachen, Germany)[g]

Completion date: 2019

DBD

46 (23/23)

Device: Liver Assist

PV < 3 mm Hg.

Oxygenated (150–200 mm Hg)

Time on machine: 1 h

IGL-1

Results awaited

Primary: postoperative peak ALT in the first postoperative week.

Secondary: Dindo/Clavien classification, hospital and ICU stay, IRI, 1-y patient/graft survival

Results awaited

Renes University Hospital[h]

Completion date: 2019

DBD

25 (25/0)

Device not reported.

PV < 3 mm Hg.

Oxygenated (40 kPa)

Time on machine: 2 h

UW-MPS

Results awaited

Primary: incidence of PNF/EAD.

Secondary: nr of intraoperative transfusions, PRS, morbidity on day 7, graft survival at 3 mo, hospital length of stay, cost of initial stay, cost of the hospitalization stay

Results awaited

PI: Matteo Ravaioli, Bologna, Italy[i]

Completion date: 2018

DBD

10 (10/0)

Device: Exiper, Bologna Machine Perfusion

Oxygenated (80–100 kPa)

Time on machine: 2 h

Not reported

Results awaited

Primary: graft function at 3 mo

Secondary: graft/patient survival at 3 mo

Results awaited

van Rijn et al[28]

2017

HMP 10/0

SCS 20/0

30 (10/20)

Device: Liver Assist

HA 20–30 mm Hg/PV 5 mm HG.

500 mL/min 100% O2

Time on machine: 126 min (123–135)

UW-MPS

HMP: 521 (469–592)

SCS: 503 (476–526)

Primary: graft survival at 6 mo.

Secondary: 1-y graft/patient survival, technical safety, perfusate microbiology, postoperative complications

100 vs. 80% 6-mo graft survival, 100 vs. 67% 1-y graft survival. 100 vs. 85% 1-y patient survival.

No technical problems.

Peak ALT (IU/L): 966 vs. 1,858.

NAS: 0/10 vs. 5/20

Dutkowski et al[27]

2015

HMP 25/0

SCS 50/0

75 (25/50)

Device: ECOPS device

PV 120–180 mL/min

Oxygenated

Time on machine: 118 min (101–149)

KPS-1

HMP: 317 (280–391)

SCS: 395 (349–447)

Primary: incidence and severity of biliary complications within 1 y after LT.

Secondary: liver IRI and function, graft survival

Ischemic cholangiopathy: 0 vs. 22%.

Biliary complications: 20 vs. 46%.

Peak ALT (IU/L): 1,239 vs. 2,065.

90 vs. 69% 1-y graft survival

Guarrera et al[25]

2015

HMP 0/31

SCS 0/30

61 (31/30)

Device: Medtronic PBS

0.667 mL/g/liver/min

No active oxygenation

Time on machine: 258 ± 54 min

Vasosol

HMP: 564 ± 96

SCS: 534 ± 144

Primary: incidence of PNF, EAD, and vascular complications, 1-y graft/patient survival.

Secondary: incidence of biliary complications, AKI, hospital length of stay, liver/kidney function markers

PNF: 3 vs. 7%. EAD: 19 vs. 30%. Vascular complications: 9 vs. 7%.

84 vs. 80% 1-y patient survival.

Biliary complications: 4/31 vs. 13/30

AKI: 10 vs. 27%.

Hospital stay: 13.6 vs. 20.1 d

Guarrera et al[23]

2010

HMP 0/20

SCS 0/20

40 (20/20)

Device: Medtronic PBS

0.667 mL/g/liver/min

No active oxygenation

Time on machine: 228 ± 54 min

Vasosol

HMP: 558 ± 126

SCS: 516 ± 168

Primary: incidence of PNF, EAD, and vascular complications, 1-y graft/patient survival.

Secondary: incidence of biliary and vascular complications, AKI, hospital length of stay, liver/kidney function markers

No PNF in either group.

EAD: 5 vs. 25%.

No vascular complications in either group.

90 vs. 90% 1-y graft/patient survival.

Biliary complications: 2/20 vs. 5/20.

Hospital stay: 10.9 vs. 15.3 d.

Peak ALT (IU/L): 560 vs. 1,358

Abbreviations: AKI, acute kidney injury; ALT, alanine aminotransferase; D, duration; DBD, donation after brain death; DCD, donation after circulatory death; DHOPE, dual hypothermic oxygenated machine perfusion solution; EAD, early allograft dysfunction; ECD, extended criteria donor; h, hours; HA, hepatic artery; HMP, hypothermic machine perfusion; IGL-1, Institute George Lopez solution; HOPE, hypothermic oxygenated machine perfusion; KPS-1, kidney perfusion solution; IRI, ischemia-reperfusion injury; L-GrAFT, liver graft assessment following transplantation risk factor; LT, liver transplantation; MEAF, model of early allograft function; MP, machine perfusion; NAS, nonanastomotic biliary strictures; PNF, primary nonfunction; PRS, postreperfusion syndrome; PV, portal vein; SCS, static cold storage; UW-MPS, University of Wisconsin machine perfusion solution.


a Trial name: HOPE with Cytokine Filtration in Liver Transplantation (Cyto-HOPE); NCT04203004.


b Trial name: HOPE for Extended Criteria Donors in Liver Transplantation (HOPEext); NCT03929523.


c Trial name: Clinical Trial of New HOPE System Versus SCS; NCT03837197.


d Trial name: Post-SCS HOPE in Bergamo Liver Transplant Program; NCT03098043.


e Trial name: Study to Evaluate Performance of LifePort Liver Transporter System, a Machine Perfusion System, for Liver Transplant (PILOT); NCT03484455.


f Trial name: DHOPE of DCD Liver Grafts in Preventing Biliary Complications after Transplantation (DHOPE-DCD); NCT02584283.


g Trial name: HOPE for Human ECD and DBD Liver Allografts (HOPE-ECD-DBD); NCT03124641.


h Trial name: Interest of Oxygenated Hypothermix Perfusion in Preservation of Hepatic Grafts from ECD (PERPHO); NCT03376074.


i Trial name: HOPE versus SCS for Margina Graft (PIO); NCT03031067.


The results of two single-center trials from France and Italy may be expected this year. The PIO trial from Italy compares 10 ECD livers treated with HOPE with 30 matched historical controls preserved by SCS (clinicaltrials.gov; NCT03031067). The PERHO trial from France compares 25 DBD livers preserved by HOPE with 75 historical controls preserved by SCS (NCT03376074). The results of several other clinical trials may be expected in the coming years. As such, a large RCT in the United States is investigating HMP versus SCS in 140 transplant recipients with the incidence of early allograft dysfunction (EAD) as the primary outcome (NCT03484455). The multicenter HOPExt trial from France randomizes 266 ECD liver grafts between HOPE and SCS, also studying EAD as its primary outcome (NCT03929523). Similarly, an RCT in France compares HOPE versus SCS in 110 ECD livers with EAD as the primary outcome (NCT03837197). A smaller single-center study from the Papa Giovanni XXIII Hospital in Italy aims to study the feasibility and safety of HOPE in their liver transplant program in 20 patients (NCT03098043). In 2020, the same hospital plans to start a RCT to study the effects of HOPE using a cytokine filter (cyto-HOPE) versus traditional HOPE in 20 ECD livers (NCT04203004). The primary outcome of the trial is the incidence of post-reperfusion syndrome in the recipient, hypothesizing that cytokine filtration further reduces IRI during HOPE.


#

Hypothermic Machine Perfusion in Experimental Studies Using Discarded Human Livers

The majority of studies published on HMP over the last decades involve mainly preclinical studies without implantation of the graft. Instead, reperfusion with whole blood and reoxygenation is used to simulate transplantation. Most experimental studies have used animal models to investigate different HMP modalities, and to date, only a handful of studies using declined human livers have been published ([Table 3]).

There is a wide variance in the reported HMP perfusion times. In general, HMP perfusion protocols are shorter than for NMP. Perfusion durations up to 7 hours have been reported in clinical studies, but a period of 1 to 2 hours of end-ischemic HMP is most commonly used.[23] [25] [27] [28] [29] Cold oxygenation restores cellular energy levels already within the first 1 to 2 hours of perfusion.[19] [29] [30] In addition, a short period of 2 hours of HMP seems to improve endothelial cell function as evidenced by increased nitric oxide production upon reperfusion of declined DCD livers, while this increase was not observed in SCS-preserved grafts.[31] Vekemans et al found reduced levels of endothelin-1 (a vasoconstrictive factor) and intercellular adhesion molecule-1 (an inflammatory factor) after HMP preservation compared with SCS, suggesting a potential benefit of HMP on vascular hemostasis and endothelial cell stability. However, the effect did not persist during reperfusion, making it unlikely to have an effect in a real transplant setting.[29] Besides short-term preservation, HMP holds potential to extend liver preservation times, which could facilitate logistics for allocation and transplantation. De Carlis et al recently reported successful transplantation of two livers with extended CIT for up to 20 hours including 8-hour end-ischemic HMP.[21] In addition, Brüggenwirth et al showed successful preservation of porcine and discarded human livers after 24-hour DHOPE with similar hepatocellular function and injury compared with shorter preservation times. It would be of interest to further investigate the boundaries of extended cold MP.[32]

Several studies have been undertaken to elucidate the beneficial effects of oxygenated versus nonoxygenated HMP. In 2005, the first study describing HMP in discarded human livers was published by Guarrera et al, which showed safe preservation of livers without active oxygenation of the MP solution.[33] The work of Schlegel et al has shown that complete absence of oxygen during HMP triggers hepatocyte cell death via mitochondria, and fails to prevent reperfusion injury.[19] The liver is a metabolically high-demanding organ, even under hypothermic conditions. Therefore, the dissolved oxygen in the perfusate may be sufficient for standard grafts, but active oxygenation is desired in grafts with more anticipated preservation injury, such as DCD livers.[34] The two clinical trials performed by Guarrera et al showed good results after HMP without active oxygenation in DBD livers.[23] [25] Both clinical trials using DCD livers were performed using an oxygenated MP solution.[27] [28] A recent preclinical study by Abudhaise et al failed to observe differences between oxygen-supplemented versus non–oxygen-supplemented portal venous perfusion during HMP in DCD and DBD livers.[35] However, alterations between the groups are most likely to manifest after rewarming and reperfusion with whole blood, which was not performed in their study. Monbaliu et al performed HMP without active oxygenation in DBD and DCD liver grafts and they reported signs of hypoxic injury as reflected by the presence of anoxic vacuoles on liver electron microscopy images, suggesting that supplemental oxygenation might have been beneficial.[36] On the other hand, some researchers fear the effects of oxidative stress when the liver is exposed to oxygen under cold conditions. However, several studies demonstrate only minimal oxidative stress during HOPE, even when high oxygen concentrations are used, or when HOPE is extended for longer hours.[19] [32] [37]

Caution should be given to the perfusion devices' pressure settings during HMP, as higher vascular resistances in the cold pose a risk of undesired endothelial shear stress to the liver sinusoids.[38] Some of the currently used perfusion devices are flow-controlled, such as the Medtronic PBS used by Guarrera et al, whereas others are pressure-controlled, like the Liver Assist from Organ Assist used by Porte et al.[23] [25] [28] Based on recent studies, adjusting MP settings to portal pressures of ≤ 3 mm Hg and arterial pressures of ≤ 25 mm Hg can avoid shear stress.[19] [39]

Regarding the perfusion route, two different approaches have been advocated. Single portal vein perfusion adds simplicity and has been shown to mitigate IRI of the liver parenchyma and biliary tree.[27] Complete perfusion of the liver and the biliary system during single portal vein perfusion was shown by fluorescence in rat, pig, and discarded human livers.[40] However, researchers opting for a dual perfusion route suggest improved oxygen supply to the peribiliary vascular plexus of the bile ducts by additional arterial perfusion.[26] [41] In a feasibility study using discarded human livers, Jomaa et al found no effect of using a dual versus single MP approach on sinusoidal endothelial injury.[42] On another note, arterial perfusion only does not seem to be a favorable approach.[35] In clinical trials, Dutkowski et al have repeatedly used single HOPE, whereas Guarrera et al and van Rijn et al used dual HOPE in their series.[23] [25] [27] [28] Mitigating biliary IRI and reducing ischemic cholangiopathy remain a lofty goal for DCD LT in particular. It remains unknown whether dual perfusion is superior to prevent ischemic cholangiopathy after transplantation, and only randomized trials comparing a single versus dual approach could answer this question. Currently, the only clinical trial studying the effects of DHOPE uses DCD livers, whereas single HOPE is used in a prospective trial in DBD grafts. A recent report by van Leeuwen et al describes feasibility of HMP through the umbilical vein instead of the portal vein in discarded human livers.[43] Portal venous flows were similar to those obtained after cannulation of the portal vein main stem. MP through the umbilical vein could enable continuous oxygenated perfusion of liver grafts during procurement, splitting, and implantation.

A variety of MP solutions have been explored during HMP, with Belzer MP solution, histidine–tryptophan–ketoglutarate preservation solution, Polysol, Celsior, Institut Georges Lopez preservation solution, and UW gluconate being the most routinely used.[29] The majority of clinical perfusions are based on the original or modified UW solution. Solutions with low potassium concentrations and without the presence of starch appear particularly advantageous. A low potassium concentration decreases vascular resistance under hypothermic conditions and the addition of starch has been shown to increases perfusate viscosity.[44] Under HMP, there is no need to add an oxygen carrier and the transport of oxygen is done passively though dilution.


#
#

Normothermic Machine Perfusion

In this MP preservation protocol, as the name implies, the temperature of the perfusate is maintained at “normal,” physiological levels at 37°C. The objective of mimicking the physiological environment is to keep the metabolic activity of the organ as close as possible to normal levels to minimize the impact of IRI, thereby increasing viability of the organ. To do this, NMP requires the use of oxygenators and the presence of oxygen transporters in the perfusate, with human red blood cells being the most common oxygen transporter used in current protocols.[45] [46] [47] [48] [49] [50] Unlike the HMP configuration, in NMP protocols, liver grafts are normally dually perfused through the portal vein and the hepatic artery. The maintenance of the organ metabolic activity during NMP offers the possibility to assess organ viability by measuring many metabolic parameters, such as lactate clearance, pH and glycemic levels, as well as bile production and its constitution.[51] Finally, normothermic conditions seem to offer a more adequate setting for organ modulation, such as the use of defatting cocktails or gene therapies during ex situ perfusion.

Prospective Clinical Studies Using Normothermic Machine Perfusion

While two case reports were initially published online before 2016 describing LTs performed with ECD livers after preservation by NMP, the first multicenter clinical trial comparing NMP versus SCS was conducted by the Oxford group being published by Ravikumar et al in 2016 ([Table 2]).[45] [52] [53] In this nonrandomized study, which was designed to assess the viability and safety of NMP by comparing 20 LTs after NMP preservation with 40 LTs using SCS preservation (20% of DCD grafts in each group), no differences were observed in 30-day graft and patient survival rates and in 6-month patient survival rates between the two groups.[45] Later, two other non-RCTs conducted independently at two Canadian transplant centers, using the same Oxford perfusion device, confirmed the feasibility and safety of NMP.[46] [47] Selzner et al of the Toronto group compared 10 NMP-preserved livers with a matched historical series of 30 transplanted livers using SCS. The proportion of DCD livers was 20% in the NMP group and 27% in the control group, and both groups presented similar graft and patient survival rates at 1 month, without differences in graft function in the early stages.[46] The second clinical trial conducted at the University of Alberta, with a similar study design, confirmed the absence of differences in graft survival rates at 1 and 6 months after transplantation. It is worth noting that one DCD liver was discarded shortly after connection to the machine due to a hidden portal venous twist that prevented organ perfusion.[47] In July 2014, the Oxford team initiated the first European multicenter RCT on NMP to extensively assess graft function and survival at 6 months and 1 year, as well as patient morbidity and mortality. This study, published in 2018, compared 121 patients who received an NMP-preserved liver with 101 patients transplanted with SCS-preserved livers. They showed lower rates of EAD and reperfusion syndrome. However, no differences were observed in the overall complication rates, hospital and intensive care unit length of stay, and in 1-year graft and patient survival rates. Noteworthy, the NMP group had 28% of DCD grafts versus 21% in the SCS group, and with longer functional warm ischemia time and total preservation time compared with the SCS group. Furthermore, there was no significant difference in the rate of biliary complications (anastomotic and nonanastomotic strictures) between the two groups, neither for DCD nor DBD grafts at the MRCP performed at 6 months post-LT.[48] Recently, the Pisa group conducted the first RCT to investigate the impact of NMP on LT with older donors. Twenty grafts from donors aged ≥ 70 years were randomized in a 1:1 ratio to NMP or SCS preservation. There was no statistical difference in the patient and graft survival rates at 6 months. A patient in the NMP group received a second graft due to hepatic artery thrombosis 10 days after LT. On the other hand, one patient in the SCS group died 31 days after LT due to an episode of intestinal occlusion that evolved into sepsis shock.[49] A single-center study from the Cleveland group tested the use of fresh-frozen plasma (FFP) as a constituent of the perfusate in association with RBC units. Using a noncommercial custom-made perfusion device, Liu et al compared the outcome of 21 LT performed with NMP-preserved livers to a historical series of LT performed with livers preserved by SCS in a 1:4 ratio. The NMP group presented less EAD, no cases of biliary complications, and the only death in this group was due to a cerebral vascular accident 8 months after LT.[50] Recently, Jassem et al reanalyzed liver tissue samples and hepatic mononuclear cells collected from DBD livers transplanted after NMP and initially published by Ravikumar et al. They observed that NMP changed the gene expression profile from proinflammation to prohealing and regeneration compared with SCS liver samples. In addition, they observed a reduction in the population of T cells that produce inflammatory factors and an increase in the pool of regulatory T cells in NMP livers. These livers presented less necrosis and apoptosis and less neutrophil infiltration compared with SCS livers. Together, these results provide mechanistic insights for the understanding of the reduction of IRI provided by perfusion of liver grafts.[45] [54]

Table 2

Clinical trials on ex situ liver normothermic machine perfusion

Author or organization

Year

Donor type DCD/DBD

N total (NMP/SCS)

Perfusion characteristics

Perfusate

Total time of preservation (min) (range)

Endpoints

Outcome (NMP vs. SCS)

PI: Cristiano Quintini, The Cleveland Clinic[a]

Ongoing

Completion date: 2023

Results awaited

15

Device:

Institutional Liver MP Device

Not reported

Results awaited

Primary: 30-d posttransplantation rate of survival and PNF

Secondary: EAD, 6-mo graft survival, liver function, and injury markers

Results awaited

Instituto de Investigacion Sanitaria La Fe[b]

Ongoing

Completion date: 2023

Results awaited

50

Device:

Not reported

Not reported

Results awaited

Primary: peak of AST and ALT at 1, 3, 5, 7 d post-LT

Secondary: PNF, graft/patient survival at 30 d, 6/12 mo, PRS, EAD, liver function and injury markers, hospital/ICU stay, RRT, intra-op thromboelastogram result, biliary stenosis in MRS evidence

Results awaited

PI: Cristiano Quintini, The Cleveland Clinic[c]

Ongoing

Completion date: 2022

ECD

15

Device:

Institutional Liver MP device

Not reported

Results awaited

Primary: patient/graft survival at 1 mo

Secondary: EAD, patient/graft survival at 6 mo, blood loss, liver function, and injury markers, hospital/ICU length of stay

Results awaited

PI: David Grant, University Health Network, Toronto[d]

Ongoing

Completion date: 2021

Results awaited

40

Device:

OrganOx metra

Not reported

Results awaited

Primary: incidence of PNF, re-LT, survival at 3 mo

Secondary: rate of device failures resulting in organ discard, recruitment rates to study, IRI, graft function, ability of perfusion parameters to predict clinical outcomes following LT

Results awaited

PI: James Shapiro, University of Alberta[e]

Ongoing

Completion date: 2021

Results awaited

50

Device:

OrganOx metra

Not reported

Results awaited

Primary: 30-d graft survival

Secondary: 30-d patient survival, EAD

Results awaited

PI: Cristiano Quintini, The Cleveland Clinic[f]

Ongoing

Completion date: 2020

Results awaited

25

Device:

Institutional Liver MP device

Not reported

Results awaited

Primary: incidence of EAD

Secondary: PNF, 6 mo graft/patient survival, 7-d peak liver function tests, intra-op flow measurement, PRS, intra-op surgical outcomes, kidney failure, biliary/vascular complications at 6 mo, hospital/ICU stay, rejection rate at 6 mo, opportunistic viral infection rate

Results awaited

PI: Stuart Knechtle, Duke University[g]

Ongoing

Completion date: 2020

Results awaited

266

Device:

OrganOx metra

Not reported

Results awaited

Primary: incidence of EAD

Secondary: PNF, graft/patient survival, PRS, liver function and injury markers, biliary complications, incidence of livers randomized but not transplanted, organ utilization, healthcare costs, quality-of-life measures

Results awaited

PI: Darius Mirza, University Hospital Birmingham[h]

Ongoing

Completion date: 2020

Results awaited

22

Device:

OrganOx metra

Not reported

Results awaited

Primary: 90-d patient survival, use of NMP to identify the proportion of transplantable liver grafts from currently rejected donor organ pool

Secondary: 12-mo liver graft function, 90-d morbidity associated with receipt of extended criteria graft, physiological response to reperfusion of perfused grafts

Results awaited

TransMedics[i]

Ongoing

Completion date: 2020

Results awaited

300

Device:

TransMedics OCS

Not reported

Results awaited

Primary: Incidence of EAD, SAEs in first 30 d

Results awaited

de Vries et al[69]

2019

DHOPE-COR-NMP 7/0

7 (7/0)

Device: Liver Assist

Pressure DHOPE:

HA: 11 mm Hg

PV: 5 mm HG

Pressure NMP:

HA: 70 mm Hg

PV: 11 mm Hg

Flow NMP:

HA: 0.55 L/min (0.24–0.73(

PV: 1.7 L/min (01.46–1.74)

HBOC-201

Total MP time: 427 (283–517)

Primary: graft survival at 3 mo

100% 3 mo graft survival

Ghinolfi et al[49]

2019

NMP 0/10

SCS 0/10

20

(10/10)

Device: Liver Assist

Flow:

HA: 0.205–0.420 L/min

PV: 1.1–1.7 L/min

Time on machine:

4.2 h (3.25–4.7)

Gelofusine (B Braun) + ABO-compatible RBC concentrate

NMP: 246

(206–267)

SCS: 394

(366–465)

Primary: graft/patient survival at 6 mo

Secondary: peak transaminases within 7 d, biliary complications at 6 mo

No PNF in either group.

EAD: 2/10 vs. 1/10.

Peak ALT (IU/L): 332 (263–610) vs. 428 (303–1,162).

Graft survival: 90 vs. 100%.

Patient survival: 100 vs. 90%.

Biliary complication: 1/10 vs. 0/10

Liu et al[50]

2019

NMP 8/13

SCS 17/68

105

(21/84)

Device:

Noncommercial, institutional apparatus

Flow:

HA: 0.5 L/min

(0.2–0.7)

PV: 1.6 L/min

(1.1–2.1)

Time on machine:

3.35–7.89 h

4 units of blood bank–obtained FFP + 4 units of PRBC

NMP: 528

(462–594)

SCS: 498

(408–588)

Primary: safety, feasibility, and impact on intrahepatic hemodynamics of FFP

Secondary: prove safety and feasibility of a noncommercial, institutional perfusion apparatus

No PNF in either group.

EAD: 19 vs. 46.4%, p = 0.02.

Peak ALT (IU/L): 363 ± 318 vs. 1,021 ± 999.

No cases of ischemic. cholangiopathy.

Patient survival: NMP: 95.2% (One patient died of intracranial hemorrhage on postoperative month 8 with normal liver function). Mortality in the historical control group not reported

Nasralla et al[48]

2018

NMP 34/87

SCS 21/80

221 (121/101)

Device:

OrganOx metra

Flow:

HA ≈ 0.28 L/min

PV ≈ 1.1 L/min

Time on machine:

9.13 h (1.42–24)

Gelofusine (B Braun) + 3-unit donor-matched PRBC

NMP: 714 (258–1527)

SCS: 465 (223–967)

Primary: peak level of serum AST within 7 d after LT

Secondary: organ discard rate, PRS, PNF, EAD, length of hospital/ICU stay, RRT, cholangiopathy on MRCP at 6 mo, graft/patient survival at 1 y

PNF: 0.8 vs. 0%.

EAD: 10 vs. 30%.

Peak AST (IU/L): 488 (408.9–582.8) vs. 964 (794.5–1,172.0).

Patient survival at year: 95.8 vs. 97%.

Graft survival at year: 95 vs. 96%

Watson et al[65]

2018

NMP 35/12

47 (47/0)

Device: Liver Assist

Pressure:

HA: 60 mm Hg

PV: 8–10 mm Hg

Flow: not reported

Time on machine: 4 h

Leukocyte depleted red cells + Gelofusine (B Braun) or Steen solution

NMP: 460–1,388

Primary: observation of biochemistry and perfusion characteristics

22 livers were transplanted. 1 recipient died following PNF, 1 developed EAD, 4 developed ITBL (3 required re-LT)

Watson et al[64]

2017

NMP 9/3

12 (12/0)

Device: Liver Assist

Pressure:

HA: 60 mm Hg

PV: 8–10 mm Hg

Flow: not reported

Time on machine: 284 (122–530)

Leukocyte depleted red cells + Gelofusine (B Braun) or Steen solution

NMP: 778 (564–1,561)

Primary: assessment of viability in declined marginal livers and research livers

5/6 developed PRS, 4 sustained vasoplegia, 1 PNF, 3 DCD livers developed cholangiopathy

Bral et al[47]

2017

NMP 4/6

SCS 8/22

39

(10/30)

Device:

OrganOx metra

Pressure:

Not reported.

Flow: not reported.

Time on machine:

11.5 h (3.3–22.5)

Gelofusine (B Braun) + 3-unit type “O” PRBC

NMP: 786 (304–1631)

SCS: 235

(64–890)

Primary: 30-d graft survival

Secondary: patient survival at day 30, peak AST within 7 d, EAD, liver function and injury markers, Clavien-Dindo score, graft/patient survival at 6 mo, biliary complications at 6 mo

No PNF in either group.

EAD: 55.5 vs. 29.6%.

Peak AST (IU/L): 1,252 (383–2,600) vs. 839 (153–2,600).

6 mo graft survival: 80 vs. 100%

6 mo patient survival: 89 vs. 100%

6 mo biliary complications 0% (0/8) vs. 14.8% (4/27) p = 0.55

Mergental et al[63]

2016

NMP 4/2

6 (6/0)

Device: Liver Assist, OrganOx

Pressure: not reported.

Flow:

HA: 0.53 L/min (0.36–0.62)

PV: 1.1 L/min (0.7–1.5)

Time on machine: 3 h

Blood-based

NMP: 798 (724–951)

Primary: demonstrate feasibility of rejected allografts transplanted following assessment and resuscitation by NMP

Uneventful transplant procedure in all 5 transplanted patients and immediate function recovery in all grafts. Normalized liver tests at median follow-up of 7 mo. One graft did not met viability criteria after 3 h of MP

Selzner et al[46]

2016

NMP 2/8

SCS 8/24

40 (10/30)

Device:

OrganOx metra

Pressure: not reported.

Flow: HA: 0.3 L/min (0.2–0.4)

PV: 1.25 L/min (1.2–1.3)

Time on machine: 8 h (5.7–9.7)

3 units of PRBC + Steen solution

NMP: 586 (221–731)

SCS: 634 (523–783)

Primary: assess safety and feasibility of NMP

Peak ALT (IU/L): 619 (55–2,858) vs. 949 (233–3,073).

3 mo graft/patient survival 100% in both groups

Ravikumar et al[45]

2016

NMP 4/16

SCS 8/32

60

(20/40)

Device: OrganOx metra

Pressure:

HA 60–75 mm Hg

PV not reported.

Flow: HA ≈0.2 L/min

PV ≈0.8 L/min

Time on machine: 9.3 h (3.5–18.5)

3 units of cross-matched PRBC + 1 unit of Gelofusine (B Braun)

Not reported

Primary: 30-d graft survival

Secondary: liver function and injury markers within 7 d after LT, patient/graft survival at 6 mo

No PNF in either group.

EAD: 15 vs. 22.5%.

Peak AST (IU/L): 417 (84–4,681) vs. 902 (218–8,786)/

30-d graft survival: 100 vs. 97.5%.

6-mo patient survival: 100 vs. 97.5%

Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; D, duration; DBD, donor after brain death; DCD, donor after circulatory death; EAD, early allograft dysfunction; ECD, extended criteria donors; FFP, fresh frozen plasma; GGT, gamma-glutamyl transferase; h, hours; HA, hepatic artery; HAT, hepatic artery thrombosis; ICU, intensive care unit; INR, international normalized ratio; IRI, ischemia–reperfusion injury; LT, liver transplantation; MAP, mean arterial pressure; MP, machine perfusion; MRCP, magnetic resonance imaging scan of biliary tree; MRS, magnetic resonance spectroscopy; POD, postoperative day; NCT, national clinical trial identifier; NMP, normothermic machine perfusion; PI, principal investigator; PNF, primary nonfunction; PRBC, packed red blood cells; PRS, postreperfusion syndrome; PV, portal vein; RRT, renal replacement therapy; SAEs, serious adverse events; SCS, static cold storage.


a Trial name: Safety and Feasibility of NMP to Preserve and Evaluate Orphan Livers; NCT03456284.


b Trial name: Efficacy of Ex-situ NMP versus Cold Storage in the Transplant with Steatotic Liver Graft (ORGANOXLAFE); NCT03930459.


c Trial name: Sequential Hypo- and Normo-thermic Perfusion to Preserve Extended Criteria Donor Livers for Transplantation; NCT04023773.


d Trial name: Using Ex-Vivo NMP With the OrganOx Metra Device to Store Human Livers for Transplantation; NCT02478151.


e Trial name: Normothermic Liver Preservation Trial; NCT03089840.


f Trial name: Pilot Study to Assess Safety and Feasibility of NMP in Human Liver Transplantation; NCT02515708.


g Trial name: WP01—Normothermic Liver Preservation; NCT02775162.


h Trial name: Viability Testing and Transplantation of Marginal Livers (VITTAL); NCT02740608.


i Trial name: TransMedics (OCS) Liver PROTECT; NCT02522871.


In all the aforementioned studies, dual perfusion was the perfusion route of choice. Hepatic artery and portal vein flows were around 0.2 to 0.3 L/min and 0.8 to 1.25 L/min, respectively. The duration of NMP varied between the studies ranging from 1.42 to 24 hours with a median time (when described) varying from 8 to 11.5 hours. In two studies, there was a period of initial SCS approximately 4 hours prior to NMP.[49] [50] Red blood cells were the oxygen transporter in all the mentioned studies in association with either Gelofusine or FFP or Steen solution.

Finally, there are eight ongoing clinical trials registered in Clinical Trials.gov that assess the impact of NMP on LT ([Table 2]). The Cleveland group is conducting three of them, which use an institutional liver MP device. The number of patients to be recruited varies from 15 to 25 in the studies. In general, the objective is to evaluate the safety and viability of MP to preserve orphaned livers and ECD organs (NCT02515708, NCT04023773, and NCT03456284). Similarly, the Valencia group in Spain is conducting a clinical trial dedicated to evaluate the efficacy of MP of steatotic livers compared with the SCS. The goal is to enroll up to 50 patients, and the final results are expected by 2023 (NCT03930459). Two Canadian groups, both using the OrganOx metra MP device are conducting two other clinical trials. The Alberta group aims to enroll 50 patients and the completion date is estimated in 2021 (NCT03089840). The Toronto group will enroll 40 patients (NCT02478151). Currently, two multicenter clinical trials are being conducted in the United States. Twenty transplant centers participated in one of these studies that uses the TransMedics MP device (TransMedics, Inc.) and initially aimed to enroll 300 participants. Recruitment is already closed and the results are expected by the end of 2020 (NCT02522871). The Duke University group leads the second multicenter trial on a group of 15 transplant centers. This study uses the Metra OrganOx device and aims to enroll 266 participants until December 2020 (NCT02775162). The results of these two large multicenter clinical trials will probably be critical for the approval of NMP devices by the U.S. Food and Drug Administration agency in the United States.


#

Normothermic Machine Perfusion in Experimental Studies Using Discarded Human Livers

Similar to what happened in the HMP scenario, experimental animal studies, mainly using porcine models, anticipated studies of NMP with discarded human livers.[55] Generally, preclinical studies of NMP using discarded livers aimed to verify the feasibility of NMP as a tool to resuscitate graft function, with the objective of identifying biomarkers and/or parameters of organ viability, as well as to investigate the time limit of NMP ([Table 3]). In this way, op den Dries et al demonstrated the viability of NMP in a group of four discarded DCD livers showing a decrease in lactate levels and bile production during the NMP period of 6 hours.[56] In addition, Sutton et al used a 6-hour NMP protocol after 6.5 hours of SCS, to show that livers with low bile production have more signs of necrosis and hepatic congestion compared with livers with high bile output.[57] However, more recent studies underline the importance of bile composition instead of mere bile production.[58] Karimian et al established a detailed technical protocol for NMP with dual perfusion in 12 ECD livers and Vogel et al showed that within 24 hours of NMP, there is a positive correlation between perfusion parameters and accepted predictors of post-LT graft survival.[59] [60] The Cleveland group evaluated the effect of 24 hours of NMP on lipid metabolism in steatotic livers. They showed active lipid metabolism over time with an increase in triglyceride levels and a decrease in total cholesterol, high-density lipoprotein, and low-density lipoprotein cholesterol, suggesting that MP can be used for lipid modulation.[61] After a preclinical study that evaluated the viability criteria in 12 ECD livers, Mergental et al reported a series of five transplanted livers with an uneventful postoperative period in all cases.[62] [63] The Cambridge group published about their NMP experiences in two articles where they initially assessed the impact of different O2 tensions in the perfusate during NMP of 12 discarded ECD livers. A high O2 tension was associated with an increased risk of post-reperfusion syndrome and sustained vasoplegia. In total, 47 livers were perfused using NMP, and 20 livers were transplanted from a group of 28 livers accepted for possible transplantation. In addition, 2 livers were transplanted from a group of 19 livers initially declined for transplantation. One recipient died due to PNF and one presented EAD, and three out of four recipients who developed intrahepatic biliary lesions required a retransplantation.[64] [65] Although MP offers the possibility of increasing the group of transplantable livers due to all the advantages mentioned earlier, these results remind us that care must be taken not to push the limits too far when deciding to implant an initially declined liver. Van Leeuwen et al recently proposed a definition of viability criteria based on both hepatocellular (liver parenchyma) and cholangiocyte (bile duct) function (see section “Hypothermic followed by Normothermic Machine Perfusion”).[66] In addition, we hope that the results of the Viability Testing and Transplantation of marginal Livers (VITTAL study), an ongoing prospective clinical trial, can provide more information on the algorithm to decide which initially discarded liver should be transplanted (NCT02740608; [Table 2]).[67]

Table 3

Studies on ex situ hypothermic and normothermic machine perfusion in discarded or declined human livers

Author

Year

Donor type

MP (n)

Perfusion characteristics

Perfusate

D (h)

Aim/Endpoints

Main outcomes

Hypothermic machine perfusion

Abudhaise et al[35]

2018

DCD/DBD

38

4–8°C

HA 30 mm Hg/PV 7 mm Hg

0.5 L/min 100% O2

KPS-1

4

Dynamic, biochemical, and histological markers during HMP

Single arterial perfusion had higher resistance, lower flows, and higher perfusate markers compared with single portal venous perfusion. No differences between oxygen-supplemented vs. non–oxygen-supplemented perfusion

Burlage et al[31]

2017

DCD/DBD

6

10°C

HA 25 ± 5 mm Hg/PV 5 mm Hg

1 L/min 100% O2

UW-MPS

2

Endothelial cell function

NO levels increased during reperfusion in HMP livers, but not in controls. Cumulative TM release was lower in HMP livers

Westerkamp et al[30]

2016

DCD/DBD

6

10°C

HA 25 ± 5 mm Hg/PV 5 mm Hg

1 L/min 100% O2

UW-MPS

2

Hepatobiliary function and injury

ATP levels, bile production, biliary bilirubin, and biliary HCO3 were higher, and lactate and glucose levels were lower in HMP livers. No differences in hepatobiliary injury markers

Schlegel et al[40]

2016

DCD

3

10°C

PV 2.5–3 mm Hg

Oxygenated (40–60 kPa)

KPS-1

n/a

Complete perfusion of liver grafts

Fluorescence under dark light confirmed complete perfusion of liver grafts by single arterial perfusion

van Leeuwen et al[43]

2019

DCD

3

10–12°C

UV 5 mm Hg

Oxygenated

UW-MPS

n/a

Assess feasibility of machine perfusion via the umbilical vein

HMP through the umbilical vein is feasible. Portal venous flows were similar to those obtained after cannulation of the portal main stem

Jomaa et al[42]

2013

DBD/DCD

12

4–8°C

HA 30 mm Hg/PV 7 mm Hg

KPS-1

1

Feasibility study

Pressures and temperature were maintained throughout the perfusions. No differences in sinusoidal endothelial ultrastructure between the groups. Sterility was maintained. Single or dual perfusion did not affect vascular resistance or flow

Monbaliu et al[36]

2012

DBD/DCD

17

4–6°C

HA 20–30 mm Hg/PV 7 mm Hg

No active oxygenation

UW-MPS

24

Explore whether quality of livers could be assessed during HMP

Nontransplantable livers released more AST and LDH than transplantable livers. Vascular resistances and metabolic profiles did not differ between transplantable vs. nontransplantable livers

Vekemans et al[29]

2011

DBD/DCD

13

5–8°C

HA 20 mm Hg/PV 3 mm Hg

Oxygenated

KPS-1

4

Test the clinical validity of end-ischemic HMP

Lower AST and LDH in HMP livers. Morphological scores were similar. MAPK was downregulated by HMP

Guarrera et al[33]

2005

Not reported

10

3–6°C

HA 3–5 mm Hg/PV 12–18 mm Hg

No active oxygenation

Vasosol

5–10

Develop a reproducible technique for liver HMP

The described method for HMP appeared to be safe and reliable to preserve livers

Normothermic machine perfusion

Liu et al[61]

2018

DCD/Steatosis

10

37°C

HA 60–100 mm Hg

PV 5–10 mm Hg

Oxygenated

Matched RBC + FFP from blood bank

24

Determine effect of NMP on steatosis and markers of hepatic quality during NMP

Active lipid metabolism present in all livers as measured by perfusate triglyceride, total cholesterol, and lipoprotein concentrations. Lactate levels undergo 3 phases during NMP: priming-to-peak phase, clearance phase, and steady-state phase

Mergental et al[62]

2018

DCD/DBD

12

37°C

HA 50 mm Hg

PV 10 mm Hg

Non-lactate-clearing (n = 6)

Lactate-clearing (n = 6)

Oxygenated

3 units of donor-specific blood group + Rhesus-negative + PRBC

6

Develop a standardized protocol for NMP with a functional assessment of rejected donor livers and to propose liver viability criteria

Viability criteria composed of lactate clearance, pH maintenance, bile production, vascular flow patterns, and liver macroscopic appearance

Boteon et al[68]

2018

DCD/DBD

5

37°C

HA 256–760 mL/min

HA 30–50 mm Hg

PV 926–1,500 mL/min

PV 8–10 mm Hg

Oxygenated

4 units of hemoglobin-based oxygen carrier Hemopure

6

Comparative study of 2 perfusion strategies to restore ECD livers

NMP + HOPE livers achieved lower expression of markers of oxidative injury and inflammation compared with NMP alone. All 5 livers in NMP + HOPE group achieved viability criteria, but 40% in NMP-only group failed

Vogel et al[60]

2017

DCD/ECD

13

37°C

HA 17.2–18.6 mm Hg

IVC 1.22–1.66 mm Hg

Oxygenated

3–4 units of compatible PRBC + Sterofundin

24

Analysis of discarded liver grafts that underwent NMP for extended 24 h

NMP preservation for 24 h demonstrated to be technically feasible even in suboptimal donor organs. Positive correlation between perfusion parameters and accepted predictors of posttransplant graft survival

Karimian et al[59]

2015

DCD/DBD

12

37°C

MAP 70 mm Hg

HA 240–272 mL/min

PV 714–782 mL/min

PV 11 mm Hg

4 L/min at 100% O2

PRBC + FFP + albumin

6

Demonstrate feasible protocol for ex situ NMP using dual-perfusion device

Initial increase in HA and PV flow. Increase in concentration of total bilirubin and bicarbonate in bile. ATP content increased during NMP. Stable concentrations of hepatic injury markers demonstrated minimal graft injury during NMP. Histological examination revealed no additional injury to grafts

Sutton et al[57]

2014

DCD/DBD

12

37°C

HA 60 mm Hg

PV 11 mm Hg

High bile output (n = 6)

Low bile output (n = 6)

Oxygenated

Heparinized plasma + PRBC

6

Assess whether bile production is a suitable biomarker of graft viability

Two patterns of bile flow identified: steadily increasing bile production and cumulative bile production. Livers in low bile output group demonstrated signs of hepatic necrosis and venous congestion vs. high bile output group

op den Dries et al[56]

2013

DCD

4

37°C

HA 50 mm Hg

PV 11 mm Hg

100% O2

PRBC + FFP; blood group and Rhesus factor identical to donor liver

6

Demonstrate feasibility of NMP in discarded livers

Biochemical markers in perfusion fluid demonstrated minimal hepatic injury and improving function. Lactate levels decreased to normal values. Bile production throughout perfusion period. Histological examination demonstrated preserved morphology, without signs of ischemia or injury

Abbreviations: AST, aspartate aminotransferase; ATP, adenosine triphosphate; D, duration; DBD, donor after brain death; DCD, donor after circulatory death; ECD, extended criteria donor; FFP, fresh frozen plasma; h, hour; HA, hepatic artery; HMP, hypothermic machine perfusion; HOPE, hypothermic oxygenated perfusion; IVC, inferior vena cava; KPS-1, kidney perfusion solution; LDH, lactate dehydrogenase; MAP, mean arterial pressure; MP, machine perfusion; NMP, normothermic machine perfusion; NO, nitric oxide; PRBC, packed red blood cells; PV, portal vein; RBC, red blood cells; TM, thrombomodulin; UW-MPS, University of Wisconsin Machine Perfusion Solution; UV, umbilical vein.



#
#

Hypothermic followed by Normothermic Machine Perfusion

In a proof-of-concept preclinical work, Boteon et al tested a sequential protocol of 2 hours of HOPE and subsequent 6 hours of NMP, which was compared with NMP alone. The combination of HOPE and NMP leads to a lower expression of oxidative stress markers, with all livers achieving viability criteria against only 40% of livers in the NMP group.[68] This strategy, which aims to combine the advantages of HMP to resuscitate the energy levels of the organ and reduce cell injury with the functional evaluation of the organ provided by the NMP, was subsequently explored in the clinical setting by the Groningen group. De Vries et al enrolled 20 discarded livers in a single-arm prospective clinical trial of initial DHOPE followed by an intermediate phase of controlled oxygenated rewarming before the final NMP period (DHOPE-COR-NMP). Seven of the 20 livers were subjected to the DHOPE-COR-NMP protocol, 5 met the LT viability criteria during 2.5 hours of NMP, while 2 livers were declined for transplantation during the NMP evaluation. All transplanted livers reached the primary endpoint of 3-month graft survival.[69] These results motivated the Groningen group to conduct a national prospective clinical trial where initially declined livers underwent the same DHOPE-COR-NMP protocol using an acellular perfusion solution containing a hemoglobin-based oxygen carrier, HBOC-201 (Hemopure; HbO2 Therapeutics LLC). Sixteen livers were enrolled and 69% (11/16) of them met all the viability criteria, including lactate < 1.7 mmol/L, perfusate pH 7.35 to 7.45, bile production > 10 mL, and bile pH > 7.45 within the first 2.5 hours of NMP. All 11 livers were successfully implanted and the patient and graft survival rates were 100% at 3 and 6 months.[66]


#

Logistical Aspects of Machine Perfusion

Based on the studies reviewed here, we believe that MP is a promising technology for organ rescue and function assessment and improvement. However, its implementation in clinical practice will require caution from the transplant community. Due to logistical and operational reasons, end-ischemic MP will probably be used in the clinical situation, instead of immediate MP at the donor hospital as happened in some initial clinical trials. Because of its high costs, MP is expected to be reserved for organs at high risk of developing complications of PNF and EAD post-LT, such as ECD organs (e.g., DCD, steatosis, advanced age, expected prolonged CIT, and hemodynamically unstable donors). In addition, MP is time-consuming and requires experienced personnel to set up the machine, place the organ, and monitor the perfusion session. It will probably increase the already overloaded work time of many transplant teams. In general, we believe that the accepting surgical team should be responsible for placement of the organ on the device and its initial monitoring. Special trained organ perfusionists may be responsible for monitoring the organ during MP. Centers might need to establish either a specialized perfusion room or use one of the operating rooms, as cannulation and perfusion of the organ must be performed in a location that meets the current standards for performing sterile procedures. Currently, the cost of MP devices available on the market is around US$ 100,000 ([Fig. 2]). Disposable sets cost from US$ 5,000 to 30,000 per unit. In general, the additional cost of MP per transplant is estimated between US$ 25,000 and 50,000 considering the device, disposable, personnel, blood products, increased donor operation room time, etc. It raises the question of who will be responsible for these extra costs: the transplant center that pumps the organ, or in case that the organ is perfused by the allocation agency (e.g., OPO in the United States and Eurotransplant in Europe) should the extra costs be shared by all centers under its jurisdiction? Would this additional cost be covered by all medical insurances? These questions and many others raised by the future implementation of MP in clinical practice were highlighted by a committee of experts on organ perfusion and published by Quintini et al in 2018 on behalf of the American Society of Transplant Surgeons. As indicated, the answers to all questions will require the participation of the entire liver transplant community, including transplant centers, allocation agencies, medical insurance organizations, and scientific associations.[70]

Zoom Image
Fig. 2 Panel showing four of the currently available devices available in the market for ex situ liver perfusion. (A) Left: an overview of the Liver Assist machine perfusion device (Organ Assist Products, B.V., Groningen, NL); top right: single hypothermic machine perfusion with a commercial perfusion solution; bottom right: dual normothermic machine perfusion using a perfusion solution based on a hemoglobin-based oxygen carrier. (B) Left: an overview of the OrganOx metra machine perfusion device (OrganOx Limited, Oxford, UK), with a close-up on the perfusion parameters panel; right: a closed container with a liver being perfused using red blood cells as perfusate. (C) Left: an overview on the TransMedics Organ Care System (OCS) Liver (Transmedics Inc, Andover, MA) machine perfusion device; right: close-up of a liver being perfused with red blood cells as a perfusate. (D) Left: an overview of the LiverPort machine perfusion device (Organ Recovery Systems, Itasca, IL), right: dual hypothermic machine perfusion with a commercial perfusion solution.

#

Pitfalls in Machine Preservation Studies

As with any clinical trial, it is extremely important to avoid selection biases. This is particularly important for organ transplantation, as both characteristics of recipient and the graft can have a tremendous impact on outcomes. It is very important to randomize the groups and the randomization process needs to be done after the organ has been accepted and not at the time of the donor offer. Although attempts are made, there are currently no widely accepted criteria for transplantability of a liver graft. Unconscious biases may influence the decision to accept or decline an organ at the time of the procurement. To transplant grafts that were initially declined by all centers in the region but later successfully transplanted after MP does not necessarily mean that those organs were nonviable prior to MP—pointing to several studies showing good outcomes for “livers-that-nobody-wants.”[25] [71] [72] [73] [74] [75] Another limitation for clinical trials of MP is that it requires large sample sizes (and increased costs) for variables that are infrequent like PNF and ischemic cholangiopathy. Therefore, it seems more reasonable to perform clinical trials using ECD organs, like DCD and severely steatotic organs, until more data are available.

It is also very important to choose the right endpoints when designing clinical trials.[76] [77] [78] The primary endpoint needs to be of significant clinical relevance. A surrogate endpoint has been defined as “a biomarker that is intended to substitute for a clinical endpoint” and generally is considered valid given a more rapid and frequent incidence and strong association with traditional endpoints.[77] Obtaining statistical significance for irrelevant endpoints does not mean superiority for MP. For example, peak transaminase posttransplant has been used in several studies and even as the primary endpoint in a RCT.[48] We believe peak transaminases/EAD should not be used as primary endpoint in machine preservation trials for two reasons.[20] First, transaminases have not been shown to have an important clinical significance in both the liver donor and post-LT settings, unless extremely high.[79] [80] [81] [82] [83] Second, since the grafts in the MP trials are flushed with several extra liters of preservation solution, part of the transaminases accumulated in the liver before transplantation are eliminated, and they have a long half-life (17 ± 5 hours for AST, 47 ± 10 hours for ALT).[84] Therefore, if transaminases are going to be used as secondary endpoints, ideally the grafts from the control arm (standard cold preservation) should be flushed with the same amount of fluid used in the perfusion circuit just before implantation to minimize the differences related to the wash-out phenomenon.


#

Conclusion

Over the last decade, MP of donor livers has gained considerable interest as an approach to resuscitate and assess ECD livers prior to transplantation. After extensive preclinical work, prospective clinical trials increased the evidence of the beneficial effects of MP over SCS preservation. It is still early to definitively confirm that MP is superior to SCS preservation in regard to graft and patient survival. We need to wait for more RCTs comparing MP with SCS preservation, as well as comparing different modalities of machine preservation. Likely, all modalities of MP could have a role in the clinical setting with each having pros and cons in different situations. There are grounds for optimism that MP leads to better preservation of the biliary tree with less expected ischemic cholangiopathy. Besides, there has been a lot of progress toward optimizing MP in terms of duration, perfusion solution, and temperatures used. Within the area of liver MP, we are now progressing toward a more individualized approach based on donor organ characteristics. In the coming years, it should become clear which organ would benefit most from MP and under which preservation conditions (e.g., hypothermic, normothermic, or both). The sequential strategy of HMP followed by NMP seems to combine the advantages of both protocols, the restoration of cellular energy with the possibility to evaluate organ function, and providing conditions for organ modulation and recondition. As such, genetic modulation therapies can be administered during MP to improve post-LT outcome.[85] [86] [87] [88] In the future, the development of nanoparticles may be translated into clinical use with the potential to mitigate organ damage through a variety of mechanisms.[89] More studies are needed to address these strategies. On the other hand, important logistical issues related to costs, MP reimbursement, personnel in charge, time, and place of the MP session must be addressed to move MP from the research environment to a reality in the clinical practice of LT. Altogether, we can expect that, in the coming years, the field of liver preservation will become even more interesting with new technologies and more evidence through clinical trials.


#

Main Concepts and Learning Points

  • Machine perfusion is a feasible and safe procedure for organ preservation.

  • Hypothermic oxygenated machine perfusion effectively restores cellular energetic capital within few hours of perfusion.

  • Normothermic machine perfusion may be more adequate to assess organ viability during the perfusion session.

  • Further clinical trials are required to define in which clinical scenario MP will be superior to SCS and cost-effective.


#
#

Conflicts of Interest

None.

Authors' Contributions

E.B-R., I.M.A.B., J.B., S.I., and P.N.M. contributed to conception and design, acquisition of data, and analysis and interpretation of data. E.B.-R., I.M.A.B., and P.N.M. drafted the article and revised it critically for important intellectual content. P.N.M. approved the final version to be published.


* These authors contributed equally to this work.


  • References

  • 1 Mazzaferro V, Battiston C, Sposito C. Pro (with caution): extended oncologic indications in liver transplantation. Liver Transpl 2018; 24 (01) 98-103
  • 2 Schaefer B, Zoller H, Schneeberger S. Con: liver transplantation for expanded criteria malignant diseases. Liver Transpl 2018; 24 (01) 104-111
  • 3 Kim WR, Lake JR, Smith JM. , et al. OPTN/SRTR 2017 annual data report: liver. Am J Transplant 2019; 19 (Suppl. 02) 184-283
  • 4 Neuberger J. Liver transplantation in the United Kingdom. Liver Transpl 2016; 22 (08) 1129-1135
  • 5 Saidi RF. Utilization of expanded criteria donors in liver transplantation. Int J Organ Transplant Med 2013; 4 (02) 46-59
  • 6 Lindbergh CA. An apparatus for the culture of whole organs. J Exp Med 1935; 62 (03) 409-431
  • 7 Dutkowski P, de Rougemont O, Clavien PA. Alexis Carrel: genius, innovator and ideologist. Am J Transplant 2008; 8 (10) 1998-2003
  • 8 Brettschneider L, Daloze PM, Huguet C. , et al. Successful orthotopic transplantation of liver homografts after eight to twenty-five hours preservation. Surg Forum 1967; 18: 376-378
  • 9 Kestens PJ, Otte JB, Alexandre GP. , et al. [Human orthotopic liver graft]. Acta Chir Belg 1970; 69 (05) 285-303
  • 10 Collins GM, Bravo-Shugarman M, Terasaki PI. Kidney preservation for transportation. Initial perfusion and 30 hours' ice storage. Lancet 1969; 2 (7632): 1219-1222
  • 11 Belzer FO, D'Alessandro AM, Hoffmann RM. , et al. The use of UW solution in clinical transplantation. A 4-year experience. Ann Surg 1992; 215 (06) 579-583 , discussion 584–585
  • 12 Laing RW, Mergental H, Mirza DF. Normothermic ex-situ liver preservation: the new gold standard. Curr Opin Organ Transplant 2017; 22 (03) 274-280
  • 13 Martins PN, Selzner M, Dayangac M. , et al. What is hot and new in basic science in liver transplantation in 2018? report of the Basic Science Committee of the International Liver Transplantation Society. Transplantation 2019; 103 (04) 654-659
  • 14 Taner T, Martins PN, Ling Q. , et al. What is hot and new in basic and translational science in liver transplantation in 2019? Report of the Basic and Translational Research Committee of the International Liver Transplantation Society. Transplantation 2019; 103 (04) 654-659
  • 15 Karangwa SA, Dutkowski P, Fontes P. , et al. Machine perfusion of donor livers for transplantation: a proposal for standardized nomenclature and reporting guidelines. Am J Transplant 2016; 16 (10) 2932-2942
  • 16 Bruinsma BG, Berendsen TA, Izamis ML, Yeh H, Yarmush ML, Uygun K. Supercooling preservation and transplantation of the rat liver. Nat Protoc 2015; 10 (03) 484-494
  • 17 de Vries RJ, Tessier SN, Banik PD. , et al. Supercooling extends preservation time of human livers. Nat Biotechnol 2019; 37 (10) 1131-1136
  • 18 von Horn C, Minor T. Transient hyperthermia during oxygenated rewarming of isolated rat livers. Transpl Int 2020; 33 (03) 272-278
  • 19 Schlegel A, de Rougemont O, Graf R, Clavien PA, Dutkowski P. Protective mechanisms of end-ischemic cold machine perfusion in DCD liver grafts. J Hepatol 2013; 58 (02) 278-286
  • 20 Dutkowski P, Guarrera JV, de Jonge J, Martins PN, Porte RJ, Clavien PA. Evolving trends in machine perfusion for liver transplantation. Gastroenterology 2019; 156 (06) 1542-1547
  • 21 De Carlis R, Lauterio A, Ferla F, Di Sandro S, Sguinzi R, De Carlis L. Hypothermic machine perfusion of liver grafts can safely extend cold Ischemia for Up to 20 hours in cases of necessity. Transplantation 2017; 101 (07) e223-e224
  • 22 Muller X, Schlegel A, Kron P. , et al. Novel real-time prediction of liver graft function during hypothermic oxygenated machine perfusion before liver transplantation. Ann Surg 2019; 270 (05) 783-790
  • 23 Guarrera JV, Henry SD, Samstein B. , et al. Hypothermic machine preservation in human liver transplantation: the first clinical series. Am J Transplant 2010; 10 (02) 372-381
  • 24 De Deken J, Kocabayoglu P, Moers C. Hypothermic machine perfusion in kidney transplantation. Curr Opin Organ Transplant 2016; 21 (03) 294-300
  • 25 Guarrera JV, Henry SD, Samstein B. , et al. Hypothermic machine preservation facilitates successful transplantation of “orphan” extended criteria donor livers. Am J Transplant 2015; 15 (01) 161-169
  • 26 Op den Dries S, Sutton ME, Karimian N. , et al. Hypothermic oxygenated machine perfusion prevents arteriolonecrosis of the peribiliary plexus in pig livers donated after circulatory death. PLoS One 2014; 9 (02) e88521
  • 27 Dutkowski P, Polak WG, Muiesan P. , et al. 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 (05) 764-770 , discussion 770–771
  • 28 van Rijn R, Karimian N, Matton APM. , et al. Dual hypothermic oxygenated machine perfusion in liver transplants donated after circulatory death. Br J Surg 2017; 104 (07) 907-917
  • 29 Vekemans K, van Pelt J, Komuta M. , et al. Attempt to rescue discarded human liver grafts by end ischemic hypothermic oxygenated machine perfusion. Transplant Proc 2011; 43 (09) 3455-3459
  • 30 Westerkamp AC, Karimian N, Matton AP. , et al. Oxygenated hypothermic machine perfusion after static cold storage improves hepatobiliary function of extended criteria donor livers. Transplantation 2016; 100 (04) 825-835
  • 31 Burlage LC, Karimian N, Westerkamp AC. , et al. Oxygenated hypothermic machine perfusion after static cold storage improves endothelial function of extended criteria donor livers. HPB (Oxford) 2017; 19 (06) 538-546
  • 32 Brüggenwirth IMA, van Leeuwen OB, de Vries Y. , et al. Extended hypothermic oxygenated machine perfusion enables ex situ preservation of porcine livers for up to 24 hours. JHEP Rep 2020; 2 (02) 100092
  • 33 Guarrera JV, Estevez J, Boykin J. , et al. Hypothermic machine perfusion of liver grafts for transplantation: technical development in human discard and miniature swine models. Transplant Proc 2005; 37 (01) 323-325
  • 34 Schlegel A, Kron P, Dutkowski P. Hypothermic oxygenated liver perfusion: basic mechanisms and clinical application. Curr Transplant Rep 2015; 2 (01) 52-62
  • 35 Abudhaise H, Davidson BR, DeMuylder P, Luong TV, Fuller B. Evolution of dynamic, biochemical, and morphological parameters in hypothermic machine perfusion of human livers: a proof-of-concept study. PLoS One 2018; 13 (09) e0203803
  • 36 Monbaliu D, Liu Q, Libbrecht L. , et al. Preserving the morphology and evaluating the quality of liver grafts by hypothermic machine perfusion: a proof-of-concept study using discarded human livers. Liver Transpl 2012; 18 (12) 1495-1507
  • 37 Lüer B, Koetting M, Efferz P, Minor T. Role of oxygen during hypothermic machine perfusion preservation of the liver. Transpl Int 2010; 23 (09) 944-950
  • 38 Schlegel A, Dutkowski P. Role of hypothermic machine perfusion in liver transplantation. Transpl Int 2015; 28 (06) 677-689
  • 39 Debbaut C, Monbaliu D, Casteleyn C. , et al. From vascular corrosion cast to electrical analog model for the study of human liver hemodynamics and perfusion. IEEE Trans Biomed Eng 2011; 58 (01) 25-35
  • 40 Schlegel A, Kron P, De Oliveira ML, Clavien PA, Dutkowski P. Is single portal vein approach sufficient for hypothermic machine perfusion of DCD liver grafts?. J Hepatol 2016; 64 (01) 239-241
  • 41 Brüggenwirth IMA, Burlage LC, Porte RJ, Martins PN. Is single portal vein perfusion the best approach for machine preservation of liver grafts?. J Hepatol 2016; 64 (05) 1194-1195
  • 42 Jomaa A, Gurusamy K, Siriwardana PN. , et al. Does hypothermic machine perfusion of human donor livers affect risks of sinusoidal endothelial injury and microbial infection? A feasibility study assessing flow parameters, sterility, and sinusoidal endothelial ultrastructure. Transplant Proc 2013; 45 (05) 1677-1683
  • 43 van Leeuwen OB, Fujiyoshi M, Ubbink R. , et al. Ex situ machine perfusion of human donor livers via the surgically reopened umbilical vein: a proof of concept. Transplantation 2019; 103 (10) 2130-2135
  • 44 Jain S, Xu H, Duncan H. , et al. Ex-vivo study of flow dynamics and endothelial cell structure during extended hypothermic machine perfusion preservation of livers. Cryobiology 2004; 48 (03) 322-332
  • 45 Ravikumar R, Jassem W, Mergental H. , et al. Liver transplantation after ex vivo normothermic machine preservation: a Phase 1 (first-in-man) clinical trial. Am J Transplant 2016; 16 (06) 1779-1787
  • 46 Selzner M, Goldaracena N, Echeverri J. , et al. Normothermic ex vivo liver perfusion using Steen solution as perfusate for human liver transplantation: first North American results. Liver Transpl 2016; 22 (11) 1501-1508
  • 47 Bral M, Gala-Lopez B, Bigam D. , et al. Preliminary single-center Canadian experience of human normothermic ex vivo liver perfusion: results of a clinical trial. Am J Transplant 2017; 17 (04) 1071-1080
  • 48 Nasralla D, Coussios CC, Mergental H. , et al; Consortium for Organ Preservation in Europe. A randomized trial of normothermic preservation in liver transplantation. Nature 2018; 557 (7703): 50-56
  • 49 Ghinolfi D, Rreka E, De Tata V. , et al. Pilot, open, randomized, prospective trial for normothermic machine perfusion evaluation in liver transplantation from older donors. Liver Transpl 2019; 25 (03) 436-449
  • 50 Liu Q, Hassan A, Pezzati D. , et al. Ex-situ liver machine perfusion: the impact of fresh frozen plasma. Liver Transpl 2020; 26 (02) 215-226
  • 51 van Leeuwen OB, de Meijer VE, Porte RJ. Viability criteria for functional assessment of donor livers during normothermic machine perfusion. Liver Transpl 2018; 24 (10) 1333-1335
  • 52 Perera T, Mergental H, Stephenson B. , et al. First human liver transplantation using a marginal allograft resuscitated by normothermic machine perfusion. Liver Transpl 2016; 22 (01) 120-124
  • 53 Watson CJ, Kosmoliaptsis V, Randle LV. , et al. Preimplant normothermic liver perfusion of a suboptimal liver donated after circulatory death. Am J Transplant 2016; 16 (01) 353-357
  • 54 Jassem W, Xystrakis E, Ghnewa YG. , et al. Normothermic machine perfusion (NMP) inhibits proinflammatory responses in the liver and promotes regeneration. Hepatology 2019; 70 (02) 682-695
  • 55 Marecki H, Bozorgzadeh A, Porte RJ, Leuvenink HG, Uygun K, Martins PN. Liver ex situ machine perfusion preservation: a review of the methodology and results of large animal studies and clinical trials. Liver Transpl 2017; 23 (05) 679-695
  • 56 op den Dries S, Karimian N, Sutton ME. , et al. Ex vivo normothermic machine perfusion and viability testing of discarded human donor livers. Am J Transplant 2013; 13 (05) 1327-1335
  • 57 Sutton ME, op den Dries S, Karimian N. , et al. Criteria for viability assessment of discarded human donor livers during ex vivo normothermic machine perfusion. PLoS One 2014; 9 (11) e110642
  • 58 Matton APM, de Vries Y, Burlage LC. , et al. Biliary Bicarbonate, pH, and glucose are suitable biomarkers of biliary viability during ex situ normothermic machine perfusion of human donor livers. Transplantation 2019; 103 (07) 1405-1413
  • 59 Karimian N, Matton AP, Westerkamp AC. , et al. Ex situ normothermic machine perfusion of donor livers. J Vis Exp 2015; (99) e52688
  • 60 Vogel T, Brockmann JG, Quaglia A. , et al. The 24-hour normothermic machine perfusion of discarded human liver grafts. Liver Transpl 2017; 23 (02) 207-220
  • 61 Liu Q, Nassar A, Buccini L. , et al. Lipid metabolism and functional assessment of discarded human livers with steatosis undergoing 24 hours of normothermic machine perfusion. Liver Transpl 2018; 24 (02) 233-245
  • 62 Mergental H, Stephenson BTF, Laing RW. , et al. Development of clinical criteria for functional assessment to predict primary nonfunction of high-risk livers using normothermic machine perfusion. Liver Transpl 2018; 24 (10) 1453-1469
  • 63 Mergental H, Perera MT, Laing RW. , et al. Transplantation of declined liver allografts following normothermic ex-situ evaluation. Am J Transplant 2016; 16 (11) 3235-3245
  • 64 Watson CJE, Kosmoliaptsis V, Randle LV. , et al. Normothermic perfusion in the assessment and preservation of declined livers before transplantation: hyperoxia and vasoplegia-important lessons from the first 12 cases. Transplantation 2017; 101 (05) 1084-1098
  • 65 Watson CJE, Kosmoliaptsis V, Pley C. , et al. Observations on the ex situ perfusion of livers for transplantation. Am J Transplant 2018; 18 (08) 2005-2020
  • 66 van Leeuwen OB, de Vries Y, Fujiyoshi M. , et al. Transplantation of high-risk donor livers after ex situ resuscitation and assessment using combined hypo- and normothermic machine perfusion: a prospective clinical trial. Ann Surg 2019; 270 (05) 906-914
  • 67 Laing RW, Mergental H, Yap C. , et al. Viability testing and transplantation of marginal livers (VITTAL) using normothermic machine perfusion: study protocol for an open-label, non-randomised, prospective, single-arm trial. BMJ Open 2017; 7 (11) e017733
  • 68 Boteon YL, Laing RW, Schlegel A. , et al. Combined hypothermic and normothermic machine perfusion improves functional recovery of extended criteria donor livers. Liver Transpl 2018; 24 (12) 1699-1715
  • 69 de Vries Y, Matton APM, Nijsten MWN. , et al. Pretransplant sequential hypo- and normothermic machine perfusion of suboptimal livers donated after circulatory death using a hemoglobin-based oxygen carrier perfusion solution. Am J Transplant 2019; 19 (04) 1202-1211
  • 70 Quintini C, Martins PN, Shah S. , et al; American Society of Transplant Surgeons Standards Committee. Implementing an innovated preservation technology: the American Society of Transplant Surgeons' (ASTS) Standards Committee White Paper on Ex Situ Liver Machine Perfusion. Am J Transplant 2018; 18 (08) 1865-1874
  • 71 Carpenter DJ, Chiles MC, Verna EC. , et al. Deceased brain dead donor liver transplantation and utilization in the United States: nighttime and weekend effects. Transplantation 2019; 103 (07) 1392-1404
  • 72 Sotiropoulos GC, Lang H, Saner FH. , et al. Long-term results after liver transplantation with “livers that nobody wants” within Eurotransplant: a center's experience. Transplant Proc 2008; 40 (09) 3196-3197
  • 73 McCormack L, Quiñonez E, Ríos MM. , et al. Rescue policy for discarded liver grafts: a single-centre experience of transplanting livers ‘that nobody wants’. HPB (Oxford) 2010; 12 (08) 523-530
  • 74 Tector AJ, Mangus RS, Chestovich P. , et al. Use of extended criteria livers decreases wait time for liver transplantation without adversely impacting posttransplant survival. Ann Surg 2006; 244 (03) 439-450
  • 75 Halazun KJ, Quillin RC, Rosenblatt R. , et al. Expanding the margins: high volume utilization of marginal liver grafts among >2000 liver transplants at a single institution. Ann Surg 2017; 266 (03) 441-449
  • 76 Muller X, Marcon F, Sapisochin G. , et al. Defining benchmarks in liver transplantation: a multicenter outcome analysis determining best achievable results. Ann Surg 2018; 267 (03) 419-425
  • 77 Stevens LA, Greene T, Levey AS. Surrogate end points for clinical trials of kidney disease progression. Clin J Am Soc Nephrol 2006; 1 (04) 874-884
  • 78 Agopian VG, Harlander-Locke MP, Markovic D. , et al. Evaluation of early allograft function using the liver graft assessment following transplantation risk score model. JAMA Surg 2018; 153 (05) 436-444
  • 79 Rosen HR, Martin P, Goss J. , et al. Significance of early aminotransferase elevation after liver transplantation. Transplantation 1998; 65 (01) 68-72
  • 80 Feng S, Goodrich NP, Bragg-Gresham JL. , et al. Characteristics associated with liver graft failure: the concept of a donor risk index. Am J Transplant 2006; 6 (04) 783-790
  • 81 Radunz S, Paul A, Nowak K, Treckmann JW, Saner FH, Mathé Z. Liver transplantation using donor organs with markedly elevated liver enzymes: how far can we go?. Liver Int 2011; 31 (07) 1021-1027
  • 82 Gadsden MM, Goldberg DS. Hepatic dysfunction in deceased donors in the age of the opioid epidemic. Transplantation 2018; 102 (10) e403
  • 83 Martins PN, Rawson A, Movahedi B. , et al. Single-center experience with liver transplant using donors with very high transaminase levels. Exp Clin Transplant 2019; 17 (04) 498-506
  • 84 Dufour DR, Lott JA, Nolte FS, Gretch DR, Koff RS, Seeff LB. Diagnosis and monitoring of hepatic injury. II. Recommendations for use of laboratory tests in screening, diagnosis, and monitoring. Clin Chem 2000; 46 (12) 2050-2068
  • 85 Thijssen MF, Brüggenwirth IMA, Gillooly A, Khvorova A, Kowalik TF, Martins PN. Gene Silencing With siRNA (RNA Interference): a new therapeutic option during ex vivo machine liver perfusion preservation. Liver Transpl 2019; 25 (01) 140-151
  • 86 Gillooly AR, Perry J, Martins PN. First Report of siRNA Uptake (for RNA Interference) during ex vivo hypothermic and normothermic liver machine perfusion. Transplantation 2019; 103 (03) e56-e57
  • 87 Bruggenwirth IMA, Martins PN. RNA interference therapeutics in organ transplantation: The dawn of a new era. Am J Transplant 2020; 20 (04) 931-941
  • 88 Buchwald JE, Xu J, Bozorgzadeh A, Martins PN. Therapeutics administered during ex vivo liver machine perfusion: An overview. World J Transplant 2020; 10 (01) 1-14
  • 89 Yao CG, Martins PN. Nanotechnology Applications in Transplantation Medicine. Transplantation 2020; 104 (04) 682-693

Address for correspondence

Paulo N. Martins, MD, PhD, FAST, FEBS, FACS
Division of Transplant, Department of Surgery, UMass Memorial Medical Center, University of Massachusetts
55 Lake Avenue, Worcester, MA 01655

Publication History

Article published online:
18 June 2020

© 2020. Thieme. All rights reserved.

Thieme Medical Publishers
333 Seventh Avenue, New York, NY 10001, USA.

  • References

  • 1 Mazzaferro V, Battiston C, Sposito C. Pro (with caution): extended oncologic indications in liver transplantation. Liver Transpl 2018; 24 (01) 98-103
  • 2 Schaefer B, Zoller H, Schneeberger S. Con: liver transplantation for expanded criteria malignant diseases. Liver Transpl 2018; 24 (01) 104-111
  • 3 Kim WR, Lake JR, Smith JM. , et al. OPTN/SRTR 2017 annual data report: liver. Am J Transplant 2019; 19 (Suppl. 02) 184-283
  • 4 Neuberger J. Liver transplantation in the United Kingdom. Liver Transpl 2016; 22 (08) 1129-1135
  • 5 Saidi RF. Utilization of expanded criteria donors in liver transplantation. Int J Organ Transplant Med 2013; 4 (02) 46-59
  • 6 Lindbergh CA. An apparatus for the culture of whole organs. J Exp Med 1935; 62 (03) 409-431
  • 7 Dutkowski P, de Rougemont O, Clavien PA. Alexis Carrel: genius, innovator and ideologist. Am J Transplant 2008; 8 (10) 1998-2003
  • 8 Brettschneider L, Daloze PM, Huguet C. , et al. Successful orthotopic transplantation of liver homografts after eight to twenty-five hours preservation. Surg Forum 1967; 18: 376-378
  • 9 Kestens PJ, Otte JB, Alexandre GP. , et al. [Human orthotopic liver graft]. Acta Chir Belg 1970; 69 (05) 285-303
  • 10 Collins GM, Bravo-Shugarman M, Terasaki PI. Kidney preservation for transportation. Initial perfusion and 30 hours' ice storage. Lancet 1969; 2 (7632): 1219-1222
  • 11 Belzer FO, D'Alessandro AM, Hoffmann RM. , et al. The use of UW solution in clinical transplantation. A 4-year experience. Ann Surg 1992; 215 (06) 579-583 , discussion 584–585
  • 12 Laing RW, Mergental H, Mirza DF. Normothermic ex-situ liver preservation: the new gold standard. Curr Opin Organ Transplant 2017; 22 (03) 274-280
  • 13 Martins PN, Selzner M, Dayangac M. , et al. What is hot and new in basic science in liver transplantation in 2018? report of the Basic Science Committee of the International Liver Transplantation Society. Transplantation 2019; 103 (04) 654-659
  • 14 Taner T, Martins PN, Ling Q. , et al. What is hot and new in basic and translational science in liver transplantation in 2019? Report of the Basic and Translational Research Committee of the International Liver Transplantation Society. Transplantation 2019; 103 (04) 654-659
  • 15 Karangwa SA, Dutkowski P, Fontes P. , et al. Machine perfusion of donor livers for transplantation: a proposal for standardized nomenclature and reporting guidelines. Am J Transplant 2016; 16 (10) 2932-2942
  • 16 Bruinsma BG, Berendsen TA, Izamis ML, Yeh H, Yarmush ML, Uygun K. Supercooling preservation and transplantation of the rat liver. Nat Protoc 2015; 10 (03) 484-494
  • 17 de Vries RJ, Tessier SN, Banik PD. , et al. Supercooling extends preservation time of human livers. Nat Biotechnol 2019; 37 (10) 1131-1136
  • 18 von Horn C, Minor T. Transient hyperthermia during oxygenated rewarming of isolated rat livers. Transpl Int 2020; 33 (03) 272-278
  • 19 Schlegel A, de Rougemont O, Graf R, Clavien PA, Dutkowski P. Protective mechanisms of end-ischemic cold machine perfusion in DCD liver grafts. J Hepatol 2013; 58 (02) 278-286
  • 20 Dutkowski P, Guarrera JV, de Jonge J, Martins PN, Porte RJ, Clavien PA. Evolving trends in machine perfusion for liver transplantation. Gastroenterology 2019; 156 (06) 1542-1547
  • 21 De Carlis R, Lauterio A, Ferla F, Di Sandro S, Sguinzi R, De Carlis L. Hypothermic machine perfusion of liver grafts can safely extend cold Ischemia for Up to 20 hours in cases of necessity. Transplantation 2017; 101 (07) e223-e224
  • 22 Muller X, Schlegel A, Kron P. , et al. Novel real-time prediction of liver graft function during hypothermic oxygenated machine perfusion before liver transplantation. Ann Surg 2019; 270 (05) 783-790
  • 23 Guarrera JV, Henry SD, Samstein B. , et al. Hypothermic machine preservation in human liver transplantation: the first clinical series. Am J Transplant 2010; 10 (02) 372-381
  • 24 De Deken J, Kocabayoglu P, Moers C. Hypothermic machine perfusion in kidney transplantation. Curr Opin Organ Transplant 2016; 21 (03) 294-300
  • 25 Guarrera JV, Henry SD, Samstein B. , et al. Hypothermic machine preservation facilitates successful transplantation of “orphan” extended criteria donor livers. Am J Transplant 2015; 15 (01) 161-169
  • 26 Op den Dries S, Sutton ME, Karimian N. , et al. Hypothermic oxygenated machine perfusion prevents arteriolonecrosis of the peribiliary plexus in pig livers donated after circulatory death. PLoS One 2014; 9 (02) e88521
  • 27 Dutkowski P, Polak WG, Muiesan P. , et al. 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 (05) 764-770 , discussion 770–771
  • 28 van Rijn R, Karimian N, Matton APM. , et al. Dual hypothermic oxygenated machine perfusion in liver transplants donated after circulatory death. Br J Surg 2017; 104 (07) 907-917
  • 29 Vekemans K, van Pelt J, Komuta M. , et al. Attempt to rescue discarded human liver grafts by end ischemic hypothermic oxygenated machine perfusion. Transplant Proc 2011; 43 (09) 3455-3459
  • 30 Westerkamp AC, Karimian N, Matton AP. , et al. Oxygenated hypothermic machine perfusion after static cold storage improves hepatobiliary function of extended criteria donor livers. Transplantation 2016; 100 (04) 825-835
  • 31 Burlage LC, Karimian N, Westerkamp AC. , et al. Oxygenated hypothermic machine perfusion after static cold storage improves endothelial function of extended criteria donor livers. HPB (Oxford) 2017; 19 (06) 538-546
  • 32 Brüggenwirth IMA, van Leeuwen OB, de Vries Y. , et al. Extended hypothermic oxygenated machine perfusion enables ex situ preservation of porcine livers for up to 24 hours. JHEP Rep 2020; 2 (02) 100092
  • 33 Guarrera JV, Estevez J, Boykin J. , et al. Hypothermic machine perfusion of liver grafts for transplantation: technical development in human discard and miniature swine models. Transplant Proc 2005; 37 (01) 323-325
  • 34 Schlegel A, Kron P, Dutkowski P. Hypothermic oxygenated liver perfusion: basic mechanisms and clinical application. Curr Transplant Rep 2015; 2 (01) 52-62
  • 35 Abudhaise H, Davidson BR, DeMuylder P, Luong TV, Fuller B. Evolution of dynamic, biochemical, and morphological parameters in hypothermic machine perfusion of human livers: a proof-of-concept study. PLoS One 2018; 13 (09) e0203803
  • 36 Monbaliu D, Liu Q, Libbrecht L. , et al. Preserving the morphology and evaluating the quality of liver grafts by hypothermic machine perfusion: a proof-of-concept study using discarded human livers. Liver Transpl 2012; 18 (12) 1495-1507
  • 37 Lüer B, Koetting M, Efferz P, Minor T. Role of oxygen during hypothermic machine perfusion preservation of the liver. Transpl Int 2010; 23 (09) 944-950
  • 38 Schlegel A, Dutkowski P. Role of hypothermic machine perfusion in liver transplantation. Transpl Int 2015; 28 (06) 677-689
  • 39 Debbaut C, Monbaliu D, Casteleyn C. , et al. From vascular corrosion cast to electrical analog model for the study of human liver hemodynamics and perfusion. IEEE Trans Biomed Eng 2011; 58 (01) 25-35
  • 40 Schlegel A, Kron P, De Oliveira ML, Clavien PA, Dutkowski P. Is single portal vein approach sufficient for hypothermic machine perfusion of DCD liver grafts?. J Hepatol 2016; 64 (01) 239-241
  • 41 Brüggenwirth IMA, Burlage LC, Porte RJ, Martins PN. Is single portal vein perfusion the best approach for machine preservation of liver grafts?. J Hepatol 2016; 64 (05) 1194-1195
  • 42 Jomaa A, Gurusamy K, Siriwardana PN. , et al. Does hypothermic machine perfusion of human donor livers affect risks of sinusoidal endothelial injury and microbial infection? A feasibility study assessing flow parameters, sterility, and sinusoidal endothelial ultrastructure. Transplant Proc 2013; 45 (05) 1677-1683
  • 43 van Leeuwen OB, Fujiyoshi M, Ubbink R. , et al. Ex situ machine perfusion of human donor livers via the surgically reopened umbilical vein: a proof of concept. Transplantation 2019; 103 (10) 2130-2135
  • 44 Jain S, Xu H, Duncan H. , et al. Ex-vivo study of flow dynamics and endothelial cell structure during extended hypothermic machine perfusion preservation of livers. Cryobiology 2004; 48 (03) 322-332
  • 45 Ravikumar R, Jassem W, Mergental H. , et al. Liver transplantation after ex vivo normothermic machine preservation: a Phase 1 (first-in-man) clinical trial. Am J Transplant 2016; 16 (06) 1779-1787
  • 46 Selzner M, Goldaracena N, Echeverri J. , et al. Normothermic ex vivo liver perfusion using Steen solution as perfusate for human liver transplantation: first North American results. Liver Transpl 2016; 22 (11) 1501-1508
  • 47 Bral M, Gala-Lopez B, Bigam D. , et al. Preliminary single-center Canadian experience of human normothermic ex vivo liver perfusion: results of a clinical trial. Am J Transplant 2017; 17 (04) 1071-1080
  • 48 Nasralla D, Coussios CC, Mergental H. , et al; Consortium for Organ Preservation in Europe. A randomized trial of normothermic preservation in liver transplantation. Nature 2018; 557 (7703): 50-56
  • 49 Ghinolfi D, Rreka E, De Tata V. , et al. Pilot, open, randomized, prospective trial for normothermic machine perfusion evaluation in liver transplantation from older donors. Liver Transpl 2019; 25 (03) 436-449
  • 50 Liu Q, Hassan A, Pezzati D. , et al. Ex-situ liver machine perfusion: the impact of fresh frozen plasma. Liver Transpl 2020; 26 (02) 215-226
  • 51 van Leeuwen OB, de Meijer VE, Porte RJ. Viability criteria for functional assessment of donor livers during normothermic machine perfusion. Liver Transpl 2018; 24 (10) 1333-1335
  • 52 Perera T, Mergental H, Stephenson B. , et al. First human liver transplantation using a marginal allograft resuscitated by normothermic machine perfusion. Liver Transpl 2016; 22 (01) 120-124
  • 53 Watson CJ, Kosmoliaptsis V, Randle LV. , et al. Preimplant normothermic liver perfusion of a suboptimal liver donated after circulatory death. Am J Transplant 2016; 16 (01) 353-357
  • 54 Jassem W, Xystrakis E, Ghnewa YG. , et al. Normothermic machine perfusion (NMP) inhibits proinflammatory responses in the liver and promotes regeneration. Hepatology 2019; 70 (02) 682-695
  • 55 Marecki H, Bozorgzadeh A, Porte RJ, Leuvenink HG, Uygun K, Martins PN. Liver ex situ machine perfusion preservation: a review of the methodology and results of large animal studies and clinical trials. Liver Transpl 2017; 23 (05) 679-695
  • 56 op den Dries S, Karimian N, Sutton ME. , et al. Ex vivo normothermic machine perfusion and viability testing of discarded human donor livers. Am J Transplant 2013; 13 (05) 1327-1335
  • 57 Sutton ME, op den Dries S, Karimian N. , et al. Criteria for viability assessment of discarded human donor livers during ex vivo normothermic machine perfusion. PLoS One 2014; 9 (11) e110642
  • 58 Matton APM, de Vries Y, Burlage LC. , et al. Biliary Bicarbonate, pH, and glucose are suitable biomarkers of biliary viability during ex situ normothermic machine perfusion of human donor livers. Transplantation 2019; 103 (07) 1405-1413
  • 59 Karimian N, Matton AP, Westerkamp AC. , et al. Ex situ normothermic machine perfusion of donor livers. J Vis Exp 2015; (99) e52688
  • 60 Vogel T, Brockmann JG, Quaglia A. , et al. The 24-hour normothermic machine perfusion of discarded human liver grafts. Liver Transpl 2017; 23 (02) 207-220
  • 61 Liu Q, Nassar A, Buccini L. , et al. Lipid metabolism and functional assessment of discarded human livers with steatosis undergoing 24 hours of normothermic machine perfusion. Liver Transpl 2018; 24 (02) 233-245
  • 62 Mergental H, Stephenson BTF, Laing RW. , et al. Development of clinical criteria for functional assessment to predict primary nonfunction of high-risk livers using normothermic machine perfusion. Liver Transpl 2018; 24 (10) 1453-1469
  • 63 Mergental H, Perera MT, Laing RW. , et al. Transplantation of declined liver allografts following normothermic ex-situ evaluation. Am J Transplant 2016; 16 (11) 3235-3245
  • 64 Watson CJE, Kosmoliaptsis V, Randle LV. , et al. Normothermic perfusion in the assessment and preservation of declined livers before transplantation: hyperoxia and vasoplegia-important lessons from the first 12 cases. Transplantation 2017; 101 (05) 1084-1098
  • 65 Watson CJE, Kosmoliaptsis V, Pley C. , et al. Observations on the ex situ perfusion of livers for transplantation. Am J Transplant 2018; 18 (08) 2005-2020
  • 66 van Leeuwen OB, de Vries Y, Fujiyoshi M. , et al. Transplantation of high-risk donor livers after ex situ resuscitation and assessment using combined hypo- and normothermic machine perfusion: a prospective clinical trial. Ann Surg 2019; 270 (05) 906-914
  • 67 Laing RW, Mergental H, Yap C. , et al. Viability testing and transplantation of marginal livers (VITTAL) using normothermic machine perfusion: study protocol for an open-label, non-randomised, prospective, single-arm trial. BMJ Open 2017; 7 (11) e017733
  • 68 Boteon YL, Laing RW, Schlegel A. , et al. Combined hypothermic and normothermic machine perfusion improves functional recovery of extended criteria donor livers. Liver Transpl 2018; 24 (12) 1699-1715
  • 69 de Vries Y, Matton APM, Nijsten MWN. , et al. Pretransplant sequential hypo- and normothermic machine perfusion of suboptimal livers donated after circulatory death using a hemoglobin-based oxygen carrier perfusion solution. Am J Transplant 2019; 19 (04) 1202-1211
  • 70 Quintini C, Martins PN, Shah S. , et al; American Society of Transplant Surgeons Standards Committee. Implementing an innovated preservation technology: the American Society of Transplant Surgeons' (ASTS) Standards Committee White Paper on Ex Situ Liver Machine Perfusion. Am J Transplant 2018; 18 (08) 1865-1874
  • 71 Carpenter DJ, Chiles MC, Verna EC. , et al. Deceased brain dead donor liver transplantation and utilization in the United States: nighttime and weekend effects. Transplantation 2019; 103 (07) 1392-1404
  • 72 Sotiropoulos GC, Lang H, Saner FH. , et al. Long-term results after liver transplantation with “livers that nobody wants” within Eurotransplant: a center's experience. Transplant Proc 2008; 40 (09) 3196-3197
  • 73 McCormack L, Quiñonez E, Ríos MM. , et al. Rescue policy for discarded liver grafts: a single-centre experience of transplanting livers ‘that nobody wants’. HPB (Oxford) 2010; 12 (08) 523-530
  • 74 Tector AJ, Mangus RS, Chestovich P. , et al. Use of extended criteria livers decreases wait time for liver transplantation without adversely impacting posttransplant survival. Ann Surg 2006; 244 (03) 439-450
  • 75 Halazun KJ, Quillin RC, Rosenblatt R. , et al. Expanding the margins: high volume utilization of marginal liver grafts among >2000 liver transplants at a single institution. Ann Surg 2017; 266 (03) 441-449
  • 76 Muller X, Marcon F, Sapisochin G. , et al. Defining benchmarks in liver transplantation: a multicenter outcome analysis determining best achievable results. Ann Surg 2018; 267 (03) 419-425
  • 77 Stevens LA, Greene T, Levey AS. Surrogate end points for clinical trials of kidney disease progression. Clin J Am Soc Nephrol 2006; 1 (04) 874-884
  • 78 Agopian VG, Harlander-Locke MP, Markovic D. , et al. Evaluation of early allograft function using the liver graft assessment following transplantation risk score model. JAMA Surg 2018; 153 (05) 436-444
  • 79 Rosen HR, Martin P, Goss J. , et al. Significance of early aminotransferase elevation after liver transplantation. Transplantation 1998; 65 (01) 68-72
  • 80 Feng S, Goodrich NP, Bragg-Gresham JL. , et al. Characteristics associated with liver graft failure: the concept of a donor risk index. Am J Transplant 2006; 6 (04) 783-790
  • 81 Radunz S, Paul A, Nowak K, Treckmann JW, Saner FH, Mathé Z. Liver transplantation using donor organs with markedly elevated liver enzymes: how far can we go?. Liver Int 2011; 31 (07) 1021-1027
  • 82 Gadsden MM, Goldberg DS. Hepatic dysfunction in deceased donors in the age of the opioid epidemic. Transplantation 2018; 102 (10) e403
  • 83 Martins PN, Rawson A, Movahedi B. , et al. Single-center experience with liver transplant using donors with very high transaminase levels. Exp Clin Transplant 2019; 17 (04) 498-506
  • 84 Dufour DR, Lott JA, Nolte FS, Gretch DR, Koff RS, Seeff LB. Diagnosis and monitoring of hepatic injury. II. Recommendations for use of laboratory tests in screening, diagnosis, and monitoring. Clin Chem 2000; 46 (12) 2050-2068
  • 85 Thijssen MF, Brüggenwirth IMA, Gillooly A, Khvorova A, Kowalik TF, Martins PN. Gene Silencing With siRNA (RNA Interference): a new therapeutic option during ex vivo machine liver perfusion preservation. Liver Transpl 2019; 25 (01) 140-151
  • 86 Gillooly AR, Perry J, Martins PN. First Report of siRNA Uptake (for RNA Interference) during ex vivo hypothermic and normothermic liver machine perfusion. Transplantation 2019; 103 (03) e56-e57
  • 87 Bruggenwirth IMA, Martins PN. RNA interference therapeutics in organ transplantation: The dawn of a new era. Am J Transplant 2020; 20 (04) 931-941
  • 88 Buchwald JE, Xu J, Bozorgzadeh A, Martins PN. Therapeutics administered during ex vivo liver machine perfusion: An overview. World J Transplant 2020; 10 (01) 1-14
  • 89 Yao CG, Martins PN. Nanotechnology Applications in Transplantation Medicine. Transplantation 2020; 104 (04) 682-693

Zoom Image
Fig. 1 Graph representing the dynamics of the metabolism rate according to the machine perfusion temperature used. Based on Van't Hoff's principle (expressed as Q10 = (k2/k1) 10/(t2-t1)) the metabolism rate is much lower in the cold (0–12°C). Types of organ preservation and machine perfusion proposed in accordance with temperature intervals: Hyper MP, hyperthermic machine perfusion (38–42°C); NMP, normothermic machine perfusion (35–38°C); SNMP, subnormothermic machine perfusion (24–34°C); MMP, midthermic machine perfusion (13–24°C); HMP, hypothermic machine perfusion (0–12°C); SCS, static cold storage (0–8°C), super-cooling preservation (−4 to 0°C). (Adapted from Karangwa et al.[15])
Zoom Image
Fig. 2 Panel showing four of the currently available devices available in the market for ex situ liver perfusion. (A) Left: an overview of the Liver Assist machine perfusion device (Organ Assist Products, B.V., Groningen, NL); top right: single hypothermic machine perfusion with a commercial perfusion solution; bottom right: dual normothermic machine perfusion using a perfusion solution based on a hemoglobin-based oxygen carrier. (B) Left: an overview of the OrganOx metra machine perfusion device (OrganOx Limited, Oxford, UK), with a close-up on the perfusion parameters panel; right: a closed container with a liver being perfused using red blood cells as perfusate. (C) Left: an overview on the TransMedics Organ Care System (OCS) Liver (Transmedics Inc, Andover, MA) machine perfusion device; right: close-up of a liver being perfused with red blood cells as a perfusate. (D) Left: an overview of the LiverPort machine perfusion device (Organ Recovery Systems, Itasca, IL), right: dual hypothermic machine perfusion with a commercial perfusion solution.