Open access

Cold Ischaemic Injury in Kidney Transplantation

Written By

Sarah A. Hosgood, James P. Hunter and Michael L. Nicholson

Submitted: 12 May 2012 Published: 19 December 2012

DOI: 10.5772/50666

From the Edited Volume

Current Concepts in Kidney Transplantation

Edited by Sandip Kapur, Cheguevara Afaneh and Meredith J. Aull

Chapter metrics overview

2,656 Chapter Downloads

View Full Metrics

1. Introduction

Kidney transplantation is considered the best treatment for end stage renal failure (ESRF) with longer life expectancy and superior quality of life compared to dialysis therapy [1-3]. However, a major constraint to transplantation is the lack of suitable organ donors. To increase the number of available organs there has been an incentive to use ‘marginal’ donors such as donation after cardiac death (DCD) and expanded criteria donors (ECD), in addition to kidneys from the traditional living and deceased donors [4,5]. Although an important source of organs for transplantation, once transplanted a significant proportion of these kidneys have early graft dysfunction.

There are many attributing factors that influence the outcome of the transplanted graft. Donor and recipient age, creatinine clearance, history of hypertension, poor human leukocyte antigen (HLA) matching, cause of death, ethnicity, the cold ischaemic (CI) time and in the case of DCD donors the warm ischaemic insult have all been described as major determinants of graft function and graft survival [6]. The CI time is perhaps the only modifiable factor that significantly affects graft outcome.

Since the 1970s organ preservation has relied on hypothermic conditions to allow an organ to be preserved outside the body from the time of retrieval until transplantation. This allows the organ to be allocated nationally, to the most suitable and immunologically matched recipient. Nonetheless, hypothermic preservation has its limitations and viability cannot be sustained for an indefinite period of time. Hypothermic preservation has been described as ‘a compromise between the benefits and detriments of cooling’ [7].

Advertisement

2. Standard criteria donor (SCD)

Deceased organ donors fall into three categories. A standard criteria donor is a deceased donor who is declared brain dead after a stroke or other brain injury. Brain death means that there is the irreversible loss of function of the brain.

Advertisement

3. Donation after cardiac death (DCD) donor

Donation after cardiac death donors (DCD) are donors from which the organs are retrieved after the cessation of circulation due to a cardiac arrest. These organs are regarded as marginal organs due to the warm ischaemic (WI) insult that they receive before the onset of preservation. This WI interval causes a degree of injury that can lead to irreversible damage, resulting in an unfavourable outcome after transplantation. Four classifications of DCD donors have been categorised depending on the circumstances of death and when the organs are retrieved [8,9] (Table 1).

CategoryDefinitionType
1Dead on arrivalUncontrolled
2Unsuccessful resuscitationUncontrolled
3Awaiting cardiac arrestControlled
4Cardiac arrest while brain deathControlled/uncontrolled

Table 1.

Maastricht categories of donation after cardiac death donors.

Maastricht type 1 and 2 donors are patients who have died suddenly from a cardiac event or trauma and therefore are usually based in the Accident & Emergency department. After a failed resuscitation, the patient is pronounced dead and a 5 minute ‘hands off’ period allowed to lapse. The organs are perfused in-situ through aortic cannulas inserted through the femoral artery [10].

Maastricht type 3 and 4 are patients who are based on an intensive care unit after a severe brain injury. The patient does not meet the criteria for brain stem death and will maintain spontaneous ventilation. Under controlled conditions with no possibility of recovery withdrawal of treatment is planned. After the cessation of the heartbeat the patient is transferred to the operating theatre and the kidneys retrieved after in-situ cooling. In the uncontrolled situation an unexpected cardiac arrest follows brain stem death. The WI time is usually within the region of 15 minutes for controlled donors but can be considerably longer in the uncontrolled situation.

Advertisement

4. Expanded criteria donors (ECD)

Expanded criteria donors (ECD) are defined as any brain dead donor aged ≥ 60 years or over 50 years with ≥ 2 of the following conditions; Hypertension, terminal serum creatinine equal or greater than 132µmol/L or death resulting from an intracranial haemorrhage.

Advertisement

5. Cold ischaemic injury

Hypothermic preservation is based on the principle that cooling an organ inhibits the enzymatic processes. There is a 2-3 fold decrease in metabolism for every 10°C reduction in temperature [11,12]. This slows the depletion of adenosine triphosphate (ATP) and also inhibits the degrading processes (phospholipid hydrolysis). Nonetheless, under hypothermic conditions the metabolic rate remains at about 10% and therefore over time, the hypoxic conditions cause substantial injury [12] this is termed CI injury.

The depletion of ATP due to the inhibition of oxidative metabolism increases levels of adenosine, inosine and hypoxanthine within the cell leading to the formation of lactic acid [13]. This lowers the intracellular pH causing lysosomal instability and the activation of lytic enzymes [14,15]. The depletion of ATP also reduces a large number of cellular processes. Inactivation of the Na+/K+ ATPase pump allows the accumulation of calcium, sodium and water within the cell causing cellular swelling [15]. The binding of transition metals such as iron to their carrier proteins (transferrin, ferritin) is also inhibited which increases the intracellular concentration of free iron [16,17]. This is a strong catalyst for the generation of oxygen free radicals which promotes the production of other free radicals [14]. The impact of CI injury is evident immediately after transplantation when oxygenated blood is re-introduced into the kidney. The downstream effects of ischaemia reperfusion (I/R) injury results in tubular and vascular damage with the impairment of blood flow to the kidney and reduced urine output after transplantation. The kidney can withstand CI times up to 48 hours. Nonetheless, attempts have been made to reduce CI injury and on average the CI time now falls below 24 hours in most transplant centres.

Advertisement

6. Impact

6.1. Delayed graft function

Renal graft function after transplantation is typically measured as incidence of delayed graft function (DGF). There are several definitions of DGF however the majority of centres define DGF as the requirement for dialysis within the first week after transplantation. The diagnosis is based on low urine output, slow decline in serum creatinine levels and increased metabolic instability. Acute tubular injury, otherwise termed acute tubular necrosis (ATN) caused by ischaemic injury is the main cause of DGF after transplantation [18]. DGF is associated with complications such as acute rejection, increased fibrosis and the risk of poorer long term graft survival. It also has a significant economic cost, can complicate patient treatment and prolong hospital stay [19]. Rates of DGF typically range from 5 to 40% in deceased donor kidney transplants [20]. Rates of DGF in live donor transplantation are significantly less (2-5%) due to the short CI time and healthy younger donors [21].

Many experimental studies have shown that the duration of CI directly influences graft function. Several studies suggest that even after 6 hours of CI, significant injury occurs [22,23]. Clinically, the CI time has been clearly shown as an independent risk factor for DGF and reducing the CI time can reduce the incidence of DGF. In an analysis of a series of DBD transplants the risk of DGF was found to increase by 23% for every 6 hours of CI [24] and Locke et al found that limiting the CI time to less than 12 hours reduced the risk of DGF by 15% [25]. Other studies have shown that the risk of DGF is increased by 3.3 and 4.4 fold by increasing the CI time by 5 and 10 hours [26].

6.2. Graft survival

The CI time is regarded as an independent risk factor for DGF and DGF is associated with reduced graft survival [27,28]. However, recent evidence suggests that the association of CI time and DGF may have less of an impact on graft survival than previously thought. A multicentre analysis of kidney preservation found that only when the preservation period exceeded 18 hours was the CI time associated with reduced graft survival [29]. A large analysis of registry data of paired deceased donor kidneys found that DGF induced by CI injury had a limited impact on the long term outcome. Nonetheless, in other studies the CI time has been found to independently influence graft survival even in live donor transplantation and in young deceased donors [30,31].

The disparity between DGF and survival is perhaps due to the lack of sensitivity of DGF in determining the severity of kidney injury. DGF is a simple and standard method of reporting early graft dysfunction. However, dialysis within the first week after transplantation can be used to correct metabolic instability without the presence of significant kidney injury. As such, it is difficult to determine the impact of DGF. DGF due to CI can be reversible and therefore have no effect on long term outcome [32]. However, in severe cases, DGF can lead to incomplete recovery and reduced graft survival due to the loss of nephron mass [33]. Giral-Classe et al reported that rather than the incidence of DGF, the duration of DGF was the important factor with DGF over six days associated with reduced long term graft survival [34]. More recently, urinary biomarkers have been used to determine the severity of acute kidney injury with cystatin C, neutrophil gelatinase-associated lipocalin (NGAL), interleukin-18 (IL-18] and kidney injury molecule-1 (KIM-1) emerging as the most reliable and sensitive markers of injury [35-37]. Although, not readily used as a diagnostic tool in clinical practice, they may be applied more frequently in the future.

Advertisement

7. Acute rejection

Acute rejection (AR) following renal transplantation can be split into two categories, cell mediated rejection and antibody mediated rejection (also termed vascular rejection). Acute cellular rejection is the more common of the two types and with the introduction of modern immunosuppressive agents rates have dropped from 50% a decade ago to 15-20% today. The typical stimulus for cellular rejection is the presence of so-called ‘passenger leucocytes’ which are immune cells carried within the blood vessels and tissues of the donor organ. Following transplantation they are exposed to the recipient immune system which recognises them as foreign and results in activation of host lymphocytes which attack the donor kidney. Antibody mediated rejection is less common and usually more severe and if left untreated can rapidly destroy the graft.

Acute rejection is an important factor in early outcomes of transplantation and is closely associated with delayed graft function (DGF) [38-41]. The precise link between DGF, acute rejection and CI time is difficult to fully elucidate. Prolonged CI has been shown to be one of the main risk factors for DGF and DGF is an independent risk factor for AR [42]. However, DGF is a result of a number of factors and it is over simplistic to ascribe acute rejection to just one of those factors. Nonetheless there is evidence that the CI time, alongside other factors, including duration of dialysis, number of HLA mismatches, panel reactive antibodies more than 5% are independent predictors of AR. A large retrospective analysis of 611 transplants demonstrated that CI time was the strongest predictor of DGF [42]. The risk of DGF increased from 9.6% with 12 hours CI time to 21.5% with 24 hours CI time. In the same analysis the risk of AR was increased by 4% for each additional hour of CI time and the risk of rejection in patients receiving kidneys with less than 24 hours CI time was 14.1% compared to 29.3% in kidneys with greater than 24 hours CI time. Furthermore, death-censored graft survival is significantly reduced in patients in whom AR complicates DGF. In addition CI duration of greater than 24 hours has a significantly reduced death-censored graft survival in comparison with durations of less than 24 hours [42].

Advertisement

8. Donor specific effects

Kidneys from DCD and ECD donors commonly present with high rates of DGF compared to SCD and live donors. [43]. DGF typically ranges from 22% to 84% in DCD kidneys compared to 14% to 40% in DBD donors [25, 44-47]. Evidence suggests that the outcome of kidneys from uncontrolled DCDs is poorer when compared to the controlled DCDs with significantly higher rates of DGF, as a response to the longer duration of warm ischaemic (WI) injury under the uncontrolled situation [48].

Kidneys from ECD have a 70% increased risk of graft loss and higher rates of DGF [25,49,50]. The prognosis is even poorer in DCD kidneys from older donors (over 50 years) with the risk of graft failure rising to 80% [25].

In addition to DGF, a small but significant proportion of kidneys from DCD donors also have primary non function (PNF) with rates reported to range from 4 to 19% amongst transplant centres over the last 30 years [51,52]. PNF is particularly detrimental as the patient is exposed to surgery and immunosuppressive therapies without benefit. Furthermore, they may become sensitized to donor antigens, reducing the opportunity for future transplants.

The WI insult in DCD kidneys and the reduced capacity of kidneys from ECDs to recover and regenerate are certainly major contributing factors for early graft dysfunction. Experimental evidence suggests that the combined effect of WI and CI injury exacerbates the injury during reperfusion and the duration of CI has been found to have a strong influence on graft outcome [53]. However, the impact of CI in clinical transplantation is again varied. It appears that as in SCDs, long term graft survival is not necessarily affected by DGF and CI not necessarily an independent predictor of graft survival. Recent evidence from clinical DCD and DBD programmes have reported similar rates of graft survival after 5 and 10 years [45,54-57]. In a series of 112 uncontrolled DCD kidneys, DGF rates were 84% compared to 22% in DBD donors [54]. Nevertheless, the graft survival rates were similar in both groups of patients, 69.3% versus 75.5% at 5 years and 50.3% versus 57.9% at 10 years, respectively. The link between WI, CI and graft survival is not well documented. However, it appears that prolonged CI after a period WI may not be as detrimental to graft survival as previously thought and that kidneys can recover from ischaemic injury with no long term effects [58].

Advertisement

9. Preservation techniques

Organ preservation was first introduced into clinical transplantation in the 1960s. Until this time without proper preservation conditions, kidneys were transplanted as soon as possible after retrieval to minimize the injury. It was then recognized that in order to improve the outcome of transplantation, better methods of preservation were required. Experimental studies in the 1950s by Lapchinsky [59] in the Soviet Union and the early work by Carrel and Lindbergh, showed that ischaemic injury could be minimized by reducing the temperature [60]. In 1963, Calne et al used the concept of hypothermic temperatures to extend the preservation time and successfully transplant canine kidneys after 12 hours of storage [61]. This led to the application and development of preservation techniques and solutions that are used today.

Advertisement

10. Static cold storage

Static cold storage (CS) is undoubtedly the simplest and most widely utilised method of hypothermic preservation. The kidney is flushed with cold preservation solution to remove the blood and cool the organ. The kidney is then stored in solution surrounded by crushed ice. Preservation solutions have been designed to counteract the detrimental effects of CI injury. There are a number of commercially available preservation solution, which all contain the same basic formula. This includes an impermeant to minimise swelling and provide stability to the ultra-structure of the cell. A buffer and a balanced electrolyte composition with either a high or low Na+ / K+ ratio to prevent the build up of intracellular acidosis and further minimize cellular swelling (Table 2). Solutions with a high potassium concentration are classified as intracellular and those with a high sodium concentration extracellular solutions.

Components
Impermeantsglucose, lactobionate, mannitol, raffinose, sucrose
Colloidhydroxyethyl starch (HES), polyethylene glycol (PEG)
Bufferscitrate, histidine, phosphate
Electrolytescalcium, chloride, magnesium, magnesium sulphate, potassium, sodium
Anti-oxidantsallopurinol, glutathione, mannitol, trytophan
Additivesadenosine, glutamic acid, ketoglutarate

Table 2.

Components commonly used in preservation solutions

11. Static cold storage solutions

11.1. Euro Collins

In 1969 Geoffrey Collins developed the first acellular preservation solution (Collins solution) containing a high concentration of potassium and glucose [62]. Collins solution was later modified omitting some of the ingredients such as magnesium, heparin, procain and replacing glucose with mannitol to provide better osmotic properties and lower the viscosity [63-65]. It was renamed Euro Collins solution and was widely used amongst the transplant community.

11.2. Hyperosmolar citrate

Hyperosmolar citrate (HOC) or more commonly known as Soltran or Marshall’s solution was first developed in the 1970s as an alternative to Collins solution [66,67]. It is has a high potassium content and contains basic ingredients using citrate as a buffer. Its hypertonicity is designed to prevent fluid entry into cells. It is a relatively inexpensive, non-viscose solution that is still commonly used throughout the UK in kidney transplantation. It is not recommended for DCD or marginal kidneys despite the fact that there is little evidence to support this view.

12. University of Wisconsin solution

University of Wisconsin (UW) solution has a high potassium concentration to maintain the intracellular ionic balance. It is a more complex preservation solution compared to Euro Collin and HOC, containing trisaccharide raffinose and the anion lactobionate as osmotic impermeants, a phosphate buffer, anti-oxidants (glutathione) to scavenge oxygen free radicals, allopurinol to block the activity of xanthine oxidase and adenosine, an ATP precursor. It also contains the colloid hydroxyethyl starch (HES), to prevent cellular swelling [68]. However, it is debatable whether this is it necessary in a static storage solution and there is some evidence showing that HES can increase tubular damage and cause red blood cell aggregation. Another potential disadvantage of UW solution is the high concentrations of potassium. Although thought important in the prevention of the build up of intracellular calcium, potassium can induce cellular depolarization, reduce cellular 5’-triphosphate content and activate voltage-dependent channels, such as calcium channels [69]. Nonetheless, due to its composition UW solution had, and still has, a significant advantage over other preservation solutions enabling kidneys to be stored for longer periods with better function and less histological injury after transplantation. It is still considered the ‘gold standard’ preservation solution today.

13. Histidine-Tryptophan-Ketoglutarate (HTK)

HTK was originally developed as a cardioplegic solution but because of its low viscosity was quickly adopted for clinical preservation of the kidney, pancreas and liver [70-72]. It is an extracellular solution and uses the impermeant mannitol and histidine as a buffer. It also contains 2 amino acids, tryptophan, to stabilize cellular membranes and prevent oxidant damage and ketoglutararate, a substrate to support anaerobic metabolism. Recent concerns have been raised regarding its use for ECD and DCD kidneys or for kidneys with prolonged storage times [73]. Some clinical studies have associated its use with the increased risk of PNF and early graft loss [74]. Nonetheless, it is a popular preservation solution widely used throughout Europe and the UK.

14. Celsior solution

Celsior is an extracellular solution and was initially designed for heart transplantation. It contains a high sodium concentration with histidine as a buffer, lactobionate and mannitol to prevent oedema and glutathione as an antioxidant. The solution has proved beneficial in heart, liver, pancreas and in kidney transplantation [75-78].

15. Outcome

An abundance of experimental studies have investigated the efficacy of one solution over another with the majority of studies labelling UW solution as the most superior. However, clinically the evidence is sparse. UW, HTK and Celsior appear to be the better preservation solutions with little difference in rates of DGF between the solutions its usage. Euro Collin solution is not widely used and is regarded as inferior with the suggestion of increasing the risk of DGF [79]. The outcome of individual preservation solutions is more apparent when the CI time is extended beyond 24 hours with UW fairing significantly better than other solutions.

16. Hypothermic machine perfusion

Since the introduction of CS techniques in the 1970s there has been much debate about whether CS or hypothermic machine perfusion (HMP) is the best method of kidney preservation. Undoubtedly, the simplicity of CS has a significant advantage over HMP. However, HMP is it thought to be a better method of preservation in that it allows a continual flush of the microcirculation, prevents the accumulation of waste products, sustains a higher metabolic rate, protects against depolarization of the endothelial cell membrane and reduces free radical formation [80].

Folkert O Belzer was the first to develop a portable HMP system [81,82] in the 1960s. However, with the introduction and success of CS in the 1970s there was little development of this technique in subsequent decades. Nonetheless, with the increasing use of DCD and ECD kidneys over the last decade, there has been renewed interest into the use of HMP. New simpler and portable systems have been developed such as the Lifeport Kidney Transporter (Organ Recovery System, US) which has encouraged the use of this technology. Many experimental studies have found HMP to improve preservation [7,12] and the quality of the kidney. The largest multicentre clinical trial conducted in Europe comparing CS and HMP in deceased donors found that HMP reduced the risk of DGF compared to CS (adjusted odds ratio, 0.57; P=0.01] and improved 1 and 3 year graft survival [83,84]. Although the overall rate of DGF was only reduced by 6%.

The evidence suggests that HMP may be more beneficial in reducing DGF rates in marginal kidneys. In a sub-analysis of 82 pairs of DCD kidneys from the European trial, the DGF rate in the HMP group was 53.7% compared to 69.5% in kidneys that were statically stored [85]. However, there was no significant difference in graft survival at 1 or 3 years. In a further sub-analysis of ECD donors in this trial, HMP reduced rates of DGF from 29.7% to 22% and also improved 1 and 3 year graft survival in ECD kidneys [84,86]. In contrast to this support for HMP, a multicentre UK trial found no beneficial effects of HMP. 45 pairs of controlled DCD kidneys were randomized to HMP or CS [87]. The DGF rates were 58% vs 56% in the HMP and CS groups respectively. However, this trial has been criticised for the sequential design and the small number of patients [88].

HMP techniques are still open to criticism with the suggestion of increased endothelial injury, as found in a recent study of porcine livers [89], risk of trauma to the vessels and the question of cost effectiveness compared to static storage techniques [90]. Nonetheless, it appears that HMP may hold a significant advantage in reducing CI injury compared to CS techniques. The experimental evidence is strong and there is a growing abundance of evidence from clinical studies to suggest an advantage. However, the evidence is not conclusive and there is a need for more clinical trials to determine the superior method of preservation.

17. Normothermic machine perfusion

Maintaining an organ under normothermic conditions is an alternative technique of preservation. Continuous perfusion of the kidney at warmer temperatures with the delivery of nutrients and oxygen has the advantage of avoiding hypothermic injury and hypoxia. In addition, it also may aid recovery and prevent further injury.

Early attempts at normothermic preservation were generally unsuccessful due to the inability to maintain cellular integrity and support renal metabolism. However, advances have been made over the last few decades with the use of technology borrowed from cardiac surgery. The development of less traumatic perfusion pumps and the recognition of the necessity for the delivery of nutrients and oxygen to achieve successful perfusion has made normothermic preservation a realistic contender in clinical transplantation.

Normothermic perfusion can be applied in various ways. The concept of extracorporeal membrane oxygenation (ECMO) to maintain extracorporeal circulation at normal room or body temperature with hyperoxygenated blood can be used to maintain tissue perfusion after the heart has stopped. Normothermic recirculation has proved beneficial in the retrieval of hearts, lungs and abdominal organs. Valero et al assessed the effects of implementing this technique in clinical practice in small group of DCD donors [91]. Circulation was maintained for 60 minutes before total body cooling. The incidence of DGF and PNF was reduced after normothermic recirculation compared to standard in situ or total body cooling. Gravel et al described a DGF rate of 11% in controlled DCD donors [92] and Lee et al found similar 5 year graft survival rates to DBD and living donors [93]. Maintaining circulation before retrieval is thought to condition the organs by up-regulating adenosine receptors which may protect against preservation injury [91]. Reznik et al, recently reported the application of extracorporeal normothermic recirculation in uncontrolled DCD donors using leukocyte depleted blood [94,95]. Initial graft function was achieved in 6 out of the 16 patients. In the kidney, more evidence is needed to determine how normothermic recirculation before retrieval correlates with early and longer term graft function.

In consideration of the logistical problems of prolonged preservation a great deal of research has focused on using normothermic preservation in combination with hypothermic techniques. Experimentally, intermediate periods of normothermic preservation have been used to restored energy metabolism with replenishment of adenosine levels, effectively ‘resuscitating’ the organ and retaining viability compared to kidneys stored under hypothermic conditions [96,97].

Brasile et al found that a period of warm ex-vivo perfusion at the end of the preservation period could resuscitate the kidney after warm and cold ischaemic injury [98,99]. More prolonged normothermic preservation periods have also been more beneficial than hypothermic techniques [100,101]. The only report of a normothermic kidney perfusion technique in clinical practice is by Hosgood and Nicholson [102]. In this single case report of a short period of normothermic perfusion of a marginal kidney with an oxygenated packed red blood cell based solution, the recipient had immediate graft function compared to DGF in the recipient of the paired CS kidney. Further results of the ongoing series at Leicester are awaited. Nonetheless, despite the potential benefits, normothermic preservation is logistically difficult to carry out requiring technical support and expensive perfusion systems.

18. Biomarkers

Measuring the amount of ischaemic injury during preservation would be advantageous as the quality of the kidney could be assessed and a decision made upon its viability. This would be particularly beneficial for marginal kidneys to reduce the likelihood of PNF. Viability is normally assessed by numerous factors including donor history, duration of cardiac arrest, the quality of in-situ perfusion, CI interval and visual inspection of the kidney. Ultimately this relies on the judgement of an experienced surgeon. To avoid PNF, surgeons are typically cautious and therefore many kidneys are deemed unsuitable for transplantation and are discarded [57]. HMP has been used to assess viability. Two aspects can be measured; Firstly, the continuous recirculation of preservation solution through the kidney allows the perfusate flow to be measured and intra-renal resistance can be calculated. Secondly, the perfusate can be sampled to measure cellular injury.

Clinically, the perfusion flow index (PFI) has been used as a measure of flow and resistance [103,104]. This is based on a minimum flow being obtained for a given pressure. The Transplant Group at Newcastle, UK recommend that a PFI of greater than 0.6ml/min/mmHg/100 gram of kidney is needed for a kidney to be deemed suitable for transplantation [105]. However, the ability of these parameters to predict DGF or PNF in clinical practice is limited. Jochman et al recently reported that although renal resistance (RR) at the end of HMP was an independent risk factor for DGF and 1 year graft survival, it had a low predictive power and could not be relied on as a sole measure of viability [106]. This is in agreement with other small clinical studies by Sonnenday [107] and Guarrera [108] et al that showed that kidneys with poor perfusion parameters had a similar outcome to those with good parameters.

Viability can also be measured by sampling the perfusate for biomarkers of cellular injury. Markers such as redox free iron, glutathione S-transferase (GST), total glutathione S-transferase (tGST), lactate dehydrogenase (LDH), N-acetyl-β-D-glucosaminidase (NAG), heart-type fatty acid binding protein (H-FABP) and alanine aminopeptidase (Ala-AP) have all been used to determine injury [104-106,109]. There is little information on their predictive value. However, Jochman et al recently published the results from the European HMP trial in which perfusate samples were taken for the assessment of biomarkers at the end of HMP [106]. GST, NAG, and H-FABP were found to be independent predictors for DGF but not for graft survival in the first year after transplantation. LDH, ASAT, and Ala-AP were found to have no predictive potential for post transplant outcome. Furthermore, the biomarkers did not correlate with intra renal resistance. The evidence suggests that viability assessment during HMP cannot be used independently but may be used collectively with the kidney characteristics and donor demographics to determine the suitability of a kidney for transplantation.

Normothermic preservation techniques may hold more promise in the assessment of viability compared to HMP techniques. During normothermic perfusion renal function and metabolism are restored. In experimental models, low levels of blood flow, reduced renal function and low oxygen consumption have been associated with increased ischaemic injury. Furthermore, these functional measures could be combined with injury biomarkers to assess the quality of the kidney.

19. Experimental studies

19.1. Oxygenation

There is a growing body of evidence in support of recovering ischaemically damaged organs with oxygenated preservation techniques at low temperatures. Historically, oxygenation was considered an essential component of hypothermic kidney preservation in order to support mitochondrial resynthesis of ATP and to delay the injury process. However, with the introduction of the modern day preservation solutions, and the rapid adoption of simple CS techniques, oxygen was not thought to be a vital ingredient and as such is not commonly applied in the clinical setting. Various techniques have been used to apply oxygen under CS and HMP conditions.

Retrograde oxygen persufflation is a simple technique whereby filtered and humidified oxygen is bubbled directly through the renal vasculature during CS. The gas is then allowed to escape through small perforations in the surface of the organ. Reports of its application date back to the 1970s [110,111]. Experimentally, there has been renewed interest in this technique showing a beneficial effect on graft function when compared to CS and HMP techniques [112,113].

Hyperbaric oxygenation is the delivery of oxygen under increased atmospheric pressure. Hyperbaric oxygenation is normally used to treat decompression sickness, carbon monoxide poisoning, gas embolism, circulatory disorders and to promote wound healing [114-116]. However, it has been used in organ preservation. Under normal atmospheric pressure there is a limit to the amount of oxygen that can be carried in the blood. Increasing the atmospheric pressure at which it is delivered, increases the amount of dissolved oxygen in the plasma allowing deeper penetration into the tissue (Henry’s Law). Therefore, tissues can be adequately oxygenated in the absence of a blood flow, a particular advantage in organ preservation [114,115]. Although an interesting concept and benefits have been demonstrated in liver and bowel transplantation, there has been little evidence of its use in kidney preservation in recent times.

Oxygen can also be added during HMP. At present, HMP is not supplemented with oxygen based on the presumption that air equilibration in perfusates sufficiently supports energy metabolism and that oxygen consumption at 4ºC is around 5% of that found at body temperature [117]. However, ATP can be restored in part, with the addition of oxygen and energy substrates during perfusion [118]. Short periods of oxygenated perfusion after CS have also been used to resuscitate and condition organs, correcting ATP loss, reducing levels of oxidative stress and improving organ viability [119]. The addition of free radial scavengers such as superoxide dismutase (SOD) to the preservation solution has been found to be beneficial [119,120] in preventing the generation of oxygen free radicals in this highly oxygenated environment.

20. Oxygenated solutions

Oxygen can also be effectively administered during preservation by the use of artificial oxygen carriers. Perfluorocarbons (PFC) are inert solutions that have a high capacity for dissolving oxygen. They release oxygen down a concentration gradient creating a highly oxygenated environment which is not affected by temperature [121,122]. They can be added simply during CS in a technique called the two layer method (TLM). The density of the PFC allows two layers to be formed, PFC on the bottom and the preservation solution on top. The organ is placed in the solution and remains between the two layers. Oxygen can be continuously added allowing adequate diffusion through the organ. TLM has been particularly beneficial for pancreas preservation, allowing a sufficient amount of ATP to be generated to improve organ viability [121,123]. The use of TLM has shown potential in other organs but has failed to gain much support as the ability of oxygen to penetrate deep into tissue in more densely capsulated organs has been questioned. In the kidney its beneficial effect was found in a rat model, however, when applied in a porcine model the results showed no advantage [121,124-126].

PFC can also be formulated as an emulsion for continuous perfusion and was applied during early attempts at machine perfusion [126-129]. However, the instability and adverse effects of the emulsions at that time prevented their continued application [121].

Other novel oxygen carriers have recently been applied experimentally in kidney preservation. Hemarina-M101 (M101] is a respiratory pigment derived from a marine invertebrate, Arenicola marina [130]. It has an extremely high affinity for oxygen and functions over a large range of temperatures (4-37ºC) releasing oxygen against a gradient. Using a porcine kidney model Thuiller et al recently showed in that adding M101 to UW or HTK solution during CS for 24 hours improved renal function and reduced fibrosis after transplantation. Micro-bubbles derived from Dodecafluoropentane (DDFPe) are also being investigated as oxygen replacement therapies and may in the future be applied during organ preservation [131,132].

In addition to hypothermic conditions, perfluorochemical and haemoglobin solutions can also be used to deliver oxygen at normothermic temperatures [133]. Brasile et al originally developed an acellular normothermic solution based on a modified cell culture medium and PFC emulsion (Perflubron) [134]. The perfusate was made up of a highly enriched tissue culture-like medium containing essential and non-essential amino acids, lipids, carbohydrates.

Historically, haemoglobin based solutions such as Stroma-free haemoglobin failed to demonstrate benefit experimentally because of toxic effects on the kidney. However, a newly developed solution, pyridoxalated haemoglobin-polyoxyethylene (PHP) has been deemed to be a more stable solution [133]. New more stable 2nd and 3rd generation PFCs are being developed and several are undergoing clinical trials to assess their safety. Humphreys et al recently used a commercially made PFC ‘Oxygent’ to provide oxygenation and reduced ischaemic injury to the kidney during a period of warm ischaemia by retrograde infusion through the urinary collecting system [135].

Other solutions such as Lifor, a new artificial preservation medium containing a non protein oxygen carrier that can be used at room temperature may also be used for preservation [136, 137]. These new solutions may hold more promise for future development of normothermic preservation perfusates. Nonetheless, the use of these normothermic perfusates in clinical practice is still awaited.

21. Experimental agents

I/R injury involves a cascade of events centralised by activated endothelial cells immediately after transplantation. One of the first inflammatory responses is the infiltration of neutrophils into the tissue. Cell adhesion molecules are recognised by leukocytes which interact with tissue cells to allow the movement of immune cells and mediators to the injury site [138,139]. This is mainly mediated through the up-regulation of endothelial adhesion molecules (ICAM-1, VCAM-1 and E-Selectin) [138]. The release of pro-inflammatory cytokines and chemokines, activation of the complement system and production of reactive oxygen species (ROS) [139] also cause significant cellular injury.

A vast number of therapies have been investigated to ameliorate the detrimental effects of I/R injury such as vasodilatory agents [140,141], antioxidants [142-144], anti-inflammatory agents [145,146] and growth factors [147] and in the experimental setting many of these have proved beneficial. Of particular interest are the therapies that collectively target several mechanisms of I/R injury, these include the endogenous gaseous molecules nitric oxide (NO) [148,149], carbon monoxide (CO) [150,151] and hydrogen sulphide (H2S) [152,153]. Experimental models have shown their ability to reduce inflammation, oxidative damage, apoptosis and promote smooth muscle relaxation causing vasodilation to enhance renal blood flow. However, their application into clinical practice is awaited.

There is no single agent used as standard clinical practice to treat I/R injury and reduce DGF. Nonetheless, there are several agents of interest that have recently been examined in clinical trials. Recombinant human erythropoietin (EPO) is a treatment for anaemia in renal patients however it also has cytoprotective properties and has been shown to protect against kidney injury in experimental models [154, 155]. However, the results from two clinical trials contradict the majority of animal studies and showed no benefit of EPO in reducing rates of DGF [156,157]. Furthermore, in one trial concerns of the increase in the incidence of graft thrombosis where raised [157]. Other trials to assess the effects of EPO are ongoing and the results are pending. It has been suggested that EPO mediates protection through a tissue receptor that is distinct from the classical EPO‐receptor that is known to mediate erythropoiesis [158]. A new compound has been formulated, pyroglutamate helix B surface peptide (pHBSP) that has the tissue‐protective properties similar to those of EPO but without causing erythropoiesis [158]. Early experimental models suggest that this agent is beneficial in reducing kidney injury and may hold promise for future clinical trials.

Several volatile anaesthetic agents sevoflurane and desflurane are also being trialled in clinical transplantation to reduce kidney injury. These agents are thought to have a conditioning effect that up-regulates protective mechanisms to reduce the I/R injury response [159]. The conditioning effect can also be applied by short intervals of ischaemia either directly to the organ or remotely to a limb [160]. It can be applied to the donor or recipient and again experimental models have shown the benefits of conditioning techniques. They are particularly attractive for clinical transplantation in that no pharmacological intervention is required and therefore the technique is expected to have a high safety profile. The results of several clinical trials are eagerly awaited. Propofol is another anaesthetic agent that may reduce I/R injury [161,162]. Experimental models have highlighted the anti-oxidant and anti-apoptotic properties of the agent [161,162].

There has been a great deal of emphasis on stem cell therapy to reduce kidney injury. The ability of stem cells to differentiate into multiple lineages with the capacity to stimulate the regeneration of renal tissue is particularly attractive in kidney transplantation. Bone marrow derived mesenchymal stem cells have been used in the rat kidney to reduce inflammation and oxidative damage [163-165]. However, there has been no clinical application of this therapy in kidney transplantation.

Immunosuppressant therapies used on induction can be used to reduce I/R injury and DGF. They suppress leukocyte infiltration and reduce endothelial injury. Anti-CD25 [166] and antithymocyte globulin (ATG) [167] are amongst some of the agents being currently being studied to reduce the incidence of DGF.

22. Conclusion

CI injury is detrimental to early graft function and as such early graft dysfunction is associated with reduced graft survival and complications after transplantation. However, the direct impact of CI on long term graft survival is less clear. Clinical studies suggest that CI may not necessarily be an independent risk factor for reduced graft survival. Nonetheless, further evidence is needed to examine the relationship between CI injury and graft survival. Hypothermic preservation techniques are designed to counteract the detrimental effect of CI injury and hypothermic machine perfusion is emerging as a superior method of preservation compared with static cold storage. Other preservation techniques are being developed such as normothermic perfusion and the addition of oxygen and oxygen carriers during hypothermic preservation. These techniques may hold promise for the future to limit the damage caused by CI injury. Therapeutic agents administered to the recipient may also prove beneficial in reducing early graft dysfunction. Nonetheless, translation of these therapies from animal models to clinical practice remains difficult and the search for the optimal agent or therapy is ongoing.

References

  1. 1. RaoP. SSchaubelD. EWeiGFentonS. SEvaluating the survival benefit of kidney retransplantation.Transplantation2006Sep 15;825669674
  2. 2. SchnuellePLorenzDTredeMVan Der WoudeF. JImpact of renal cadaveric transplantation on survival in end-stage renal failure: evidence for reduced mortality risk compared with hemodialysis during long-term follow-up. J Am Soc Nephrol 1998Nov;91121352141
  3. 3. WolfeR. AAshbyV. BMilfordE. LOjoA. OEttengerR. EAgodoaL. Yet alComparison of mortality in all patients on dialysis, patients on dialysis awaiting transplantation, and recipients of a first cadaveric transplant.N Engl J Med 1999Dec 2;3412317251730
  4. 4. DaemenJ. WOomenA. PKeldersW. PKootstraGThe potential pool of non-heart-beating kidney donors.Clin Transplant 1997Apr;112149154
  5. 5. AlvarezJdel Barrio R, Arias J, Ruiz F, Iglesias J, de Elias R, et al. Non-heart-beating donors from the streets: an increasing donor pool source.Transplantation2000Jul 27;702314317
  6. 6. NybergGNilssonBNordenGKarlbergIOutcome of renal transplantation in patients over the age of 60: a case-control study.Nephrol Dial Transplant 19951019194
  7. 7. TaylorM. JBaicuS. CCurrent state of hypothermic machine perfusion preservation of organs: The clinical perspectiveCryobiology2010Jul;60(3 Suppl):S2035
  8. 8. TerasakiP. IChoY. WCeckaJ. MStrategy for eliminating the kidney shortage.Clin Transpl 19971997265267
  9. 9. KootstraGDaemenJ. HOomenA. PCategories of non-heart-beating donors.Transplant Proc 1995Oct;27528932894
  10. 10. SnoeijsM. GDekkersA. JBuurmanW. Avan den Akker L, Welten RJ, Schurink GW, et al. In situ preservation of kidneys from donors after cardiac death: results and complications. Ann Surg 2007Nov;2465844852
  11. 11. MitchellTSabaHLaakmanJParajuliNMacMillan-Crow LA. Role of mitochondrial-derived oxidants in renal tubular cell cold-storage injuryFree Radic Biol Med 2010Nov 1;49812731282
  12. 12. FullerB. JLeeC. YHypothermic perfusion preservation: the future of organ preservation revisited? Cryobiology 2007Apr;542129145
  13. 13. KosieradzkiMRowinskiWIschemia/reperfusion injury in kidney transplantation: mechanisms and preventionTransplant Proc 2008Dec;401032793288
  14. 14. SalahudeenA. KCold ischemic injury of transplanted kidneys: new insights from experimental studies.Am J Physiol Renal Physiol 2004Aug;287(2):F1817
  15. 15. WeinbergJ. MThe cell biology of ischemic renal injury.Kidney Int 1991Mar;393476500
  16. 16. CardenD. LGrangerD. NPathophysiology of ischaemia-reperfusion injury.J Pathol 2000Feb;1903255266
  17. 17. PizanisNGillnerSKamlerMDe GrootHJakobHRauenUCold-induced injury to lung epithelial cells can be inhibited by iron chelators- implications for lung preservation. Eur J Cardiothorac Surg 2011Oct;404948955
  18. 18. KwiatkowskiAWszolaMPerkowska-ptasinskaAOstrowskiKDomagalaPFesolowiczSet alInfluence of preservation method on histopathological lesions of kidney allograftsAnn Transplant 2009Jan-Mar;1411013
  19. 19. BrookN. RWallerJ. RRichardsonA. CAndrew Bradley J, Andrews PA, Koffman G, et al. A report on the activity and clinical outcomes of renal non-heart beating donor transplantation in the United Kingdom.Clin Transplant 2004Dec;186627633
  20. 20. YarlagaddaS. GCocaS. GFormicaR. NJr, Poggio ED, Parikh CR. Association between delayed graft function and allograft and patient survival: a systematic review and meta-analysisNephrol Dial Transplant 2009Mar;24310391047
  21. 21. GokM. AShentonB. KPelsersMWhitwoodAMantleDCornellCet alReperfusion injury in renal transplantation: comparison of LD, HBD and NHBD renal transplants.Ann Transplant 2004923334
  22. 22. DragunDHoffUParkJ. KQunYSchneiderWLuftF. Cet alProlonged cold preservation augments vascular injury independent of renal transplant immunogenicity and function.Kidney Int 2001Sep;60311731181
  23. 23. WilhelmS. MSimonsonM. SRobinsonA. VStoweN. TSchulakJ. ACold ischemia induces endothelin gene upregulation in the preserved kidney.J Surg Res 1999Jul;851101108
  24. 24. OjoA. OWolfeR. AHeldP. JPortF. KSchmouderR. LDelayed graft function: risk factors and implications for renal allograft survival.Transplantation1997Apr 15;637968974
  25. 25. 7717971807Locke JE, Segev DL, Warren DS, Dominici F, Simpkins CE, Montgomery RA. Outcomes of kidneys from donors after cardiac death: implications for allocation and preservation. Am J Transplant 2007 Jul;7(7):1797-1807
  26. 26. KaylerL. KSrinivasT. RScholdJ. DInfluence of CIT-induced DGF on kidney transplant outcomes.Am J Transplant 2011Dec;111226572664
  27. 27. DittrichSGronebergD. AVon LoeperJLippekFHegemannOGrosse-siestrupCet alInfluence of cold storage on renal ischemia reperfusion injury after non-heart-beating donor explantationNephron Exp Nephrol 2004e97102
  28. 28. SiedleckiAIrishWBrennanD. CDelayed graft function in the kidney transplant.Am J Transplant 2011Nov;111122792296
  29. 29. OpelzGDohlerBMulticenter analysis of kidney preservation.Transplantation2007Feb 15;833247253
  30. 30. SimpkinsC. EMontgomeryR. AHawxbyA. MLockeJ. EGentryS. EWarrenD. Set alCold ischemia time and allograft outcomes in live donor renal transplantation: is live donor organ transport feasible? Am J Transplant 2007Jan;7199107
  31. 31. HernandezDEstupinanSPerezGRufinoMGonzalez-posadaJ. MLuisDet alImpact of cold ischemia time on renal allograft outcome using kidneys from young donors. Transpl Int 2008Oct;2110955962
  32. 32. ChatziantoniouCDussauleJ. CIs kidney injury a reversible process? Curr Opin Nephrol Hypertens 2008Jan;1717681
  33. 33. BarrientosAPortolesJHerreroJ. ATorralboAPratsDGutierrez-milletVet alGlomerular hyperfiltration as a nonimmunologic mechanism of progression of chronic renal rejection.Transplantation1994Mar 15;575753756
  34. 34. Giral-classeMHourmantMCantarovichDDantalJBlanchoGDaguinPet alDelayed graft function of more than six days strongly decreases long-term survival of transplanted kidneysKidney Int 1998Sep;543972978
  35. 35. ParikhC. RJaniAMishraJMaQKellyCBaraschJet alUrine NGAL and IL-18 are predictive biomarkers for delayed graft function following kidney transplantation.Am J Transplant 2006Jul;6716391645
  36. 36. HallI. EYarlagaddaS. GCocaS. GWangZDoshiMDevarajanPet alIL-18 and urinary NGAL predict dialysis and graft recovery after kidney transplantationJ Am Soc Nephrol 2010Jan;211189197
  37. 37. KoynerJ. LVaidyaV. SBennettM. RMaQWorcesterEAkhterS. Aet alUrinary biomarkers in the clinical prognosis and early detection of acute kidney injuryClin J Am Soc Nephrol 2010Dec;51221542165
  38. 38. MclarenA. JJassemWGrayD. WFuggleS. VWelshK. IMorrisP. JDelayed graft function: risk factors and the relative effects of early function and acute rejection on long-term survival in cadaveric renal transplantationClin Transplant 1999Jun;133266272
  39. 39. LebranchuYHalimiJ. MBockAChapmanJDussolBFritscheLet alDelayed graft function: risk factors, consequences and parameters affecting outcome-results from MOST, A Multinational Observational Study.Transplant Proc 2005Jan-Feb;371345347
  40. 40. RosenthalJ. TDanovitchG. MWilkinsonAEttengerR. BThe high cost of delayed graft function in cadaveric renal transplantation.Transplantation1991May;51511151118
  41. 41. SanfilippoFVaughnW. KSpeesE. KLucasB. AThe detrimental effects of delayed graft function in cadaver donor renal transplantation.Transplantation1984Dec;386643648
  42. 42. MikhalskiDWissingK. MGhisdalLBroedersNToulyMHoangA. Det alCold ischemia is a major determinant of acute rejection and renal graft survival in the modern era of immunosuppression.Transplantation 2008Apr 15;85(7 Suppl):S39
  43. 43. NicholsonM. LMetcalfeM. SWhiteS. AWallerJ. RDoughmanT. MHorsburghTet alA comparison of the results of renal transplantation from non-heart-beating, conventional cadaveric, and living donors. Kidney Int 2000Dec;58625852591
  44. 44. GagandeepSMatsuokaLMateoRChoY. WGenykYSherLet alExpanding the donor kidney pool: utility of renal allografts procured in a setting of uncontrolled cardiac death.Am J Transplant 2006Jul;6716821688
  45. 45. DAlessandroA. MFernandezL. AChinL. TShamesB. DTurgeonN. AScottDL, et al. Donation after cardiac death: the University of Wisconsin experience.Ann Transplant 2004916871
  46. 46. WhitingJ. FDelmonicoFMorrisseyPBasadonnaGJohnsonSLewisW. Det alClinical results of an organ procurement organization effort to increase utilization of donors after cardiac death.Transplantation2006May 27;811013681371
  47. 47. TojimbaraTFuchinoueSIwadohKKoyamaISannomiyaAKatoYet alImproved outcomes of renal transplantation from cardiac death donors: a 30-year single center experienceAm J Transplant 2007Mar;73609617
  48. 48. SummersD. MJohnsonR. JAllenJFuggleS. VCollettDWatsonC. Jet alAnalysis of factors that affect outcome after transplantation of kidneys donated after cardiac death in the UK: a cohort studyLancet 2010Oct 16;376974913031311
  49. 49. SungR. SGuidingerM. KChristensenL. LAshbyV. BMerionR. MLeichtmanA. Bet alDevelopment and current status of ECD kidney transplantationClin Transpl 200520053755
  50. 50. SaidiR. FEliasNKawaiTHertlMFarrellM. LGoesNet alOutcome of kidney transplantation using expanded criteria donors and donation after cardiac death kidneys: realities and costsAm J Transplant 2007Dec;71227692774
  51. 51. HordijkWHoitsmaA. JVan Der VlietJ. AHilbrandsL. BResults of transplantation with kidneys from non-heart-beating donors. Transplant Proc 2001Feb-Mar;33(1-2):1127-1128.
  52. 52. DaemenJ. HDe VriesBOomenA. PDemeesterJKootstraGEffect of machine perfusion preservation on delayed graft function in non-heart-beating donor kidneys--early results.Transpl Int 1997104317322
  53. 53. JohnsonR. JFuggleS. VONeillJStartSBradleyJ. AForsytheJL, et al. Factors influencing outcome after deceased heart beating donor kidney transplantation in the United Kingdom: an evidence base for a new national kidney allocation policyTransplantation2010Feb 27;894379386
  54. 54. BarlowA. DMetcalfeM. SJohariYElwellRVeitchP. SNicholsonM. LCase-matched comparison of long-term results of non-heart beating and heart-beating donor renal transplantsBr J Surg 2009Jun;966685691
  55. 55. SinghR. PFarneyA. CRogersJZuckermanJReeves-danielAHartmannEet alKidney transplantation from donation after cardiac death donors: lack of impact of delayed graft function on post-transplant outcomes.Clin Transplant 2011Mar-Apr;252255264
  56. 56. SnoeijsM. GSchaubelD. EHeneRHoitsmaA. JIduM. MIjzermansJ. Net alKidneys from donors after cardiac death provide survival benefitJ Am Soc Nephrol 2010Jun;21610151021
  57. 57. HooglandE. RSnoeijsM. GWinkensBChristaansM. HVan HeurnL. WKidney Transplantation from Donors after Cardiac Death: Uncontrolled versus Controlled DonationAm J Transplant 2011Jul;11714271434
  58. 58. TaylorC. JKosmoliaptsisVSharplesL. DPrezziDMorganC. HKeyTet alTen-year experience of selective omission of the pretransplant crossmatch test in deceased donor kidney transplantationTransplantation2010Jan 27;892185193
  59. 59. LapchinskyA. GRecent results of experimental transplantation of preserved limbs and kidneys and possible use of this technique in clinical practice.Ann N Y Acad Sci 1960May 31;87539571
  60. 60. CarrelALindberghC. AThe Culture of Whole Organs.Science 1935Jun 21;812112621623
  61. 61. CalneR. YPeggD. EPryse-daviesJBrownF. LRenal Preservation by Ice-Cooling: an Experimental Study Relating to Kidney Transplantation from Cadavers.Br Med J 1963Sep 14;25358651655
  62. 62. CollinsG. MBravo-shugarmanMTerasakiP. IKidney preservation for transportationInitial perfusion and 30 hours’ ice storage. Lancet 1969Dec 6;2763212191222
  63. 63. OpelzGTerasakiP. IKidney preservation: perfusion versus cold storage-1975.Transplant Proc 1976Mar;81121125
  64. 64. CollinsG. MHalaszN. ACurrent aspects of renal preservation.Urology1977Jul;10(1 Suppl):2232
  65. 65. CollinsG. MHalaszN. ASimplified 72-hr kidney storage.Surg Forum 1974250275277
  66. 66. SlapakMWilsonAClyneCBagshawHNaikR. BLeeH. AHyperosmolar citrate versus perfudex: a functional comparison in clinical kidney preservation.Transplant Proc 1979Mar;111478481
  67. 67. JablonskiPHowdenBMarshallVScottDEvaluation of citrate flushing solution using the isolated perfused rat kidney.Transplantation1980Oct;304239243
  68. 68. BelzerF. OGlassN. RSollingerH. WHoffmannR. MSouthardJ. HA new perfusate for kidney preservation.Transplantation1982Mar;333322323
  69. 69. HauetTHanZDoucetCRamella-virieuxSHadj Aissa A, Carretier M, et al. A modified University of Wisconsin preservation solution with high-Na+ low-K+ content reduces reperfusion injury of the pig kidney graft. Transplantation2003Jul 15;7611827
  70. 70. GroenewoudA. FIsemerF. EStadlerJHeidecheC. DFlorackGHoelscherMA comparison of early function between kidney grafts protected with HTK solution versus Euro-Collins solution.Transplant Proc 1989Feb;21(1 Pt 2):1243 EOF4 EOF
  71. 71. MinorTOlschewskiPTolbaR. HAkbarSKocalkovaMDombrowskiFLiver preservation with HTK: salutary effect of hypothermic aerobiosis by either gaseous oxygen or machine perfusionClin Transplant 2002Jun;163206211
  72. 72. Fridell JA, Mangus RS, Tector AJ. Clinical experience with histidine-tryptophan-ketoglutarate solution in abdominal organ preservation: a review of recent literature. Clin Transplant 2009 Jun-Jul;23 3 305312 .
  73. 73. LynchR. JKubusJChenaultR. HPelletierS. JCampbellD. AEnglesbeM. JComparison of histidine-tryptophan-ketoglutarate and University of Wisconsin preservation in renal transplantationAm J Transplant 2008Mar;83567573
  74. 74. StevensR. BSkorupaJ. YRigleyT. HYannamG. RNielsenK. JSchrinerM. Eet alIncreased primary non-function in transplanted deceased-donor kidneys flushed with histidine-tryptophan-ketoglutarate solutionAm J Transplant 2009May;9510551062
  75. 75. BokuNTanoueYKajiharaNEtoMMasudaMMoritaSA comparative study of cardiac preservation with Celsior or University of Wisconsin solution with or without prior administration of cardioplegiaJ Heart Lung Transplant 2006Feb;252219225
  76. 76. BoudjemaKGrandadamSCompagnonPSalameEWolfPDucerfCet alEfficacy and safety of Celsior preservation fluid in liver transplantation: one-year follow up of a prospective, multicenter, non-randomized study.Clin Transplant 2011Apr 21.
  77. 77. FridellJ. AMangusR. SPowelsonJ. AOrgan preservation solutions for whole organ pancreas transplantationCurr Opin Organ Transplant 2010Dec 9.
  78. 78. NunesPMotaAFigueiredoAMacarioFRoloFDiasVet alEfficacy of renal preservation: comparative study of Celsior and University of Wisconsin solutionsTransplant Proc 2007Oct;39824782479
  79. 79. OCallaghanJ. MKnightS. RMorganR. DMorrisPJ. Preservation solutions for static cold storage of kidney allografts: a systematic review and meta-analysis.Am J Transplant 2012Apr;124896906
  80. 80. VaziriNThuillierRFavreauF. DEugeneMMilinSChatauretN. Pet alAnalysis of machine perfusion benefits in kidney grafts: a preclinical studyJ Transl Med 2011Jan 25;9:15.
  81. 81. BelzerF. OAshbyB. SDunphyJ. E24-Hour and 72-Hour Preservation of Canine Kidneys.Lancet1967Sep 9;27515536538
  82. 82. BelzerF. OCurrent methods of kidney storage.Cryobiology1969May-Jun;56444446
  83. 83. MoersCSmitsJ. MMaathuisM. HTreckmannJVan GelderFNapieralskiB. Pet alMachine perfusion or cold storage in deceased-donor kidney transplantationN Engl J Med 2009Jan 1;3601719
  84. 84. MoersCPirenneJPaulAPloegR. JMachine Preservation Trial Study Group. Machine perfusion or cold storage in deceased-donor kidney transplantation. N Engl J Med 2012Feb 23;3668770771
  85. 85. JochmansIMoersCSmitsJ. MLeuveninkH. GTreckmannJPaulAet alMachine perfusion versus cold storage for the preservation of kidneys donated after cardiac death: a multicenter, randomized, controlled trial. Ann Surg 2010Nov;2525756764
  86. 86. TreckmannJMoersCSmitsJ. MGallinatAMaathuisM. HVan Kasterop-kutzMet alMachine perfusion versus cold storage for preservation of kidneys from expanded criteria donors after brain death.Transpl Int 2011Jun;246548554
  87. 87. WatsonC. JWellsA. CRobertsR. JAkohJ. AFriendP. JAkyolMet alCold machine perfusion versus static cold storage of kidneys donated after cardiac death: a UK multicenter randomized controlled trial.Am J Transplant 2010Sep;10919911999
  88. 88. JochmansIMoersCPloegRPirenneJTo perfuse or not to perfuse kidneys donated after cardiac deathAm J Transplant 2011Feb;112409410
  89. 89. MonbaliuDHeedfeldVLiuQWylinTVan PeltJVekemansKet alHypothermic machine perfusion of the liver: is it more complex than for the kidney? Transplant Proc 2011Nov;43934453450
  90. 90. BondMPittMAkohJMoxhamTHoyleMAndersonRThe effectiveness and cost-effectiveness of methods of storing donated kidneys from deceased donors: a systematic review and economic model.Health Technol Assess 2009Aug;13(38):iii-iv, xi-xiv, 1156
  91. 91. ValeroRCabrerCOppenheimerFTriasESanchez-ibanezJDe CaboF. Met alNormothermic recirculation reduces primary graft dysfunction of kidneys obtained from non-heart-beating donors.Transpl Int 2000134303310
  92. 92. GravelM. TArenasJ. DChenaultRnd, Magee JC, Rudich S, Maraschio M, et al. Kidney transplantation from organ donors following cardiopulmonary death using extracorporeal membrane oxygenation support.Ann Transplant 2004915758
  93. 93. LeeC. YTsaiM. KKoW. JChangC. JHuR. HChuehS. Cet alExpanding the donor pool: use of renal transplants from non-heart-beating donors supported with extracorporeal membrane oxygenationClin Transplant 2005Jun;193383390
  94. 94. ReznikOBagnenkoSSkvortsovAAnanyevASenchikKLoginovIet alRehabilitation of ischemically damaged human kidneys by normothermic extracorporal hemoperfusion in situ with oxygenation and leukocyte depletionTransplant Proc 2010Jun;42515361538
  95. 95. ReznikOBagnenkoSScvortsovALoginovIAnanyevASenchikKet alThe use of in-situ normothermic extracorporeal perfusion and leukocyte depletion for resuscitation of human donor kidneys20102010Sep;255343348
  96. 96. MaessenJ. GVan Der VusseG. JVorkMKootstraGIntermediate normothermic perfusion during cold storage of ischemically injured kidneys. Transplant Proc 1989Feb;21(1 Pt 2):1252-1253.
  97. 97. MaessenJ. GVan Der VusseG. JVorkMKootstraGThe beneficial effect of intermediate normothermic perfusion during cold storage of ischemically injured kidneys. A study of renal nucleotide homeostasis during hypothermia in the dog. Transplantation 1989Mar;473409414
  98. 98. BrasileLStubenitskyB. MBoosterM. HArenadaDHaischCKootstraGTransfection and transgene expression in a human kidney during ex vivo warm perfusion.Transplant Proc 2002Nov;34(7):2624 EOF
  99. 99. BrasileLStubenitskyB. MBoosterM. HArenadaDHaischCKootstraGHypothermia--a limiting factor in using warm ischemically damaged kidneys. Am J Transplant 2001Nov;14316320
  100. 100. BrasileLStubenitskyB. MBoosterM. HLindellSAranedaDBuckCet alOvercoming severe renal ischemia: the role of ex vivo warm perfusion.Transplantation2002Mar 27;736897901
  101. 101. BrasileLStubenitskyB. MBoosterM. HHaischCKootstraGNOS: the underlying mechanism preserving vascular integrity and during ex vivo warm kidney perfusionAm J Transplant 2003Jun;36674679
  102. 102. HosgoodS. ANicholsonM. LFirst in Man Renal Transplantation After Ex Vivo Normothermic Perfusion.Transplantation 2011Aug 11.
  103. 103. AsherJWilsonCGokMShentonB. KStampSWongY. Tet alTransplantation from non heart beating donors in Newcastle upon Tyne.Ann Transplant 2004915961
  104. 104. GokM. APelzersMGlatzJ. FShentonB. KBuckleyP. EPeastonRet alDo tissue damage biomarkers used to assess machine-perfused NHBD kidneys predict long-term renal function post-transplant? Clin Chim Acta 2003Dec;338(1-2):33-43.
  105. 105. GokM. APelsersMGlatzJ. FShentonB. KPeastonRCornellCet alUse of two biomarkers of renal ischemia to assess machine-perfused non-heart-beating donor kidneys.Clin Chem 2003Jan;491172175
  106. 106. JochmansIPirenneJGraft quality assessment in kidney transplantation: not an exact science yet! Curr Opin Organ Transplant 2011Apr;162174179
  107. 107. SonnendayC. JCooperMKrausEGageFHandleyCMontgomeryR. AThe hazards of basing acceptance of cadaveric renal allografts on pulsatile perfusion parameters alone.Transplantation2003Jun 27;751220292033
  108. 108. GuarreraJ. VGoldsteinM. JSamsteinBHenrySReverteCArringtonBet alWhen good kidneys pump badly’: outcomes of deceased donor renal allografts with poor pulsatile perfusion characteristics. Transpl Int 2010Apr 1;234444446
  109. 109. De VriesBSnoeijsM. GVon BonsdorffLErnest van Heurn LW, Parkkinen J, Buurman WA. Redox-active iron released during machine perfusion predicts viability of ischemically injured deceased donor kidneys. Am J Transplant 2006Nov;61126862693
  110. 110. RossHEscottM. LGaseous oxygen perfusion of the renal vessels as an adjunct in kidney preservation.Transplantation1979Nov;285362364
  111. 111. FischerJ. HKulusDHansen-schmidtIIsselhardWAdenine nucleotide levels of canine kidneys during hypothermic aerobic or anaerobic storage in Collins solution.Eur Surg Res 1981132178188
  112. 112. TreckmannJ. WPaulASaadSHoffmannJWaldmannK. HBroelschC. Eet alPrimary organ function of warm ischaemically damaged porcine kidneys after retrograde oxygen persufflation.Nephrol Dial Transplant 2006Jul;21718031808
  113. 113. TreckmannJNagelschmidtMMinorTSanerFSaadSPaulAFunction and quality of kidneys after cold storage, machine perfusion, or retrograde oxygen persufflation: results from a porcine autotransplantation modelCryobiology2009Aug;5911923
  114. 114. EdwardsM. LHyperbaric oxygen therapy. Part 1: history and principles. J Vet Emerg Crit Care (San Antonio) 2010Jun;203284288
  115. 115. EdwardsM. LHyperbaric oxygen therapy. Part 2: application in disease. J Vet Emerg Crit Care (San Antonio) 2010Jun;203289297
  116. 116. MuralidharanVChristophiCHyperbaric oxygen therapy and liver transplantationHPB (Oxford) 200793174182
  117. 117. GuibertE. EPetrenkoA. YBalabanC. LSomovA. YRodriguezJ. VFullerB. JOrgan Preservation: Current Concepts and New Strategies for the Next DecadeTransfus Med Hemother 2011382125142
  118. 118. PeggD. EWustemanM. CForemanJMetabolism of normal and ischemically injured rabbit kidneys during perfusion for 48 hours at 10 C.Transplantation1981Nov;325437443
  119. 119. StegemannJHirnerARauenUMinorTGaseous oxygen persufflation or oxygenated machine perfusion with Custodiol-N for long-term preservation of ischemic rat livers? Cryobiology 2009Feb;5814551
  120. 120. MinorTIsselhardWYamamotoYObaraMSaadSThe effects of allopurinol and SOD on lipid peroxidation and energy metabolism in the liver after ischemia in an aerobic/anaerobic persufflation.Surg Today 1993238728732
  121. 121. HosgoodS. ANicholsonM. LThe role of perfluorocarbon in organ preservationTransplantation 2010May 27;891011691175
  122. 122. ClarkL. CJr, Gollan F. Survival of mammals breathing organic liquids equilibrated with oxygen at atmospheric pressure.Science 1966Jun 24;15273017551756
  123. 123. BrandhorstHAsifSAnderssonKTheisingerBAnderssonH. HFelldinMet alA new oxygen carrier for improved long-term storage of human pancreata before islet isolationTransplantation2010Jan 27;892155160
  124. 124. MalufD. GMasV. RYanekKStoneJ. JWeisRMasseyDet alMolecular markers in stored kidneys using perfluorocarbon-based preservation solution: preliminary resultsTransplant Proc 2006Jun;38512431246
  125. 125. MaradaTZacharovovaKSaudekFPerfluorocarbon improves post-transplant survival and early kidney function following prolonged cold ischemiaEur Surg Res 2010170 EOF178 EOF
  126. 126. HosgoodS. AMohamedI. HNicholsonM. LThe two layer method does not improve the preservation of porcine kidneys.Med Sci Monit 2011Jan;17(1):BR2733
  127. 127. BerkowitzH. DMendhamJMillerL. DImportance of circulating microparticles for optimal renal perfusion.Surg Forum 197324293295
  128. 128. BerkowitzH. DMccombsPSheetySMillerL. DSloviterHFluorochemical perfusates for renal preservation.J Surg Res 1976Jun;206595600
  129. 129. HondaKFundamental and clinical studies on intracadaveric organ perfusion with Fluosol-DA.Prog Clin Biol Res 1983122327330
  130. 130. ThuillierRDutheilDTrieuM. TMalletVAllainGRousselotMet alSupplementation With a New Therapeutic Oxygen Carrier Reduces Chronic Fibrosis and Organ Dysfunction in Kidney Static Preservation.Am J Transplant 2011Sep;11918451860
  131. 131. JohnsonJ. LDolezalM. CKerschenAMatsunagaT. OUngerE. CIn vitro comparison of dodecafluoropentane (DDFP), perfluorodecalin (PFD), and perfluoroctylbromide (PFOB) in the facilitation of oxygen exchange.Artif Cells Blood Substit Immobil Biotechnol 2009374156162
  132. 132. LundgrenC. EBergoeG. WTyssebotnI. MIntravascular fluorocarbon-stabilized microbubbles protect against fatal anemia in rats.Artif Cells Blood Substit Immobil Biotechnol 2006345473486
  133. 133. DanielsF. HMccabeR. EJr, Leonard EF. The use of hemoglobin solutions in kidney perfusions.Crit Rev Biomed Eng 198494315345
  134. 134. BrasileLDelVecchio P, Amyot K, Haisch C, Clarke J. Organ preservation without extreme hypothermia using an Oxygen supplemented perfusate.Artif Cells Blood Substit Immobil Biotechnol 199422414631468
  135. 135. HumphreysM. RErethM. HSeboT. JSlezakJ. MDongYBluteM. Let alCan the kidney function as a lung? Systemic oxygenation and renal preservation during retrograde perfusion of the ischaemic kidney in rabbitsBJU Int 2006Sep;983674679
  136. 136. GageFLeeserD. BPorterfieldN. KGraybillJ. CGillernSHawksworthJ. Set alRoom temperature pulsatile perfusion of renal allografts with Lifor compared with hypothermic machine pump solutionTransplant Proc 2009Nov;41935713574
  137. 137. RegnerK. RNilakantanVRyanR. PMortensenJWhiteS. MShamesB. Det alProtective effect of Lifor solution in experimental renal ischemia-reperfusion injuryJ Surg Res 2010Dec;164(2):e2917
  138. 138. BonventreJ. VYangLCellular pathophysiology of ischemic acute kidney injuryJ Clin Invest 2011Nov 1;1211142104221
  139. 139. BonventreJ. VPathophysiology of AKI: injury and normal and abnormal repair.Contrib Nephrol 2010165917
  140. 140. HosgoodS. ABagulAKaushikMRimoldiJGadepalliR. SNicholsonM. LApplication of nitric oxide and carbon monoxide in a model of renal preservation.Br J Surg 2008Aug;95810601067
  141. 141. OrucOInciKAkiF. TZeybekDMuftuogluS. FKilincKet alSildenafil attenuates renal ischemia reperfusion injury by decreasing leukocyte infiltrationActa Histochem 2010Jul;1124337344
  142. 142. NafarMSahraeiZSalamzadehJSamavatSVaziriN. DOxidative stress in kidney transplantation: causes, consequences, and potential treatment.Iran J Kidney Dis 2011Nov;56357372
  143. 143. SavasMYeniECiftciHYildizFGulumMKeserB. Set alThe antioxidant role of oral administration of garlic oil on renal ischemia-reperfusion injury.Ren Fail 2010Jan;323362367
  144. 144. ChatterjeeP. KNovel pharmacological approaches to the treatment of renal ischemia-reperfusion injury: a comprehensive reviewNaunyn Schmiedebergs Arch Pharmacol 2007Oct;376(1 EOF43 EOF
  145. 145. HaugC. EColvinR. BDelmonicoF. LAuchinclossHJr, Tolkoff-Rubin N, Preffer FI, et al. A phase I trial of immunosuppression with anti-ICAM-1 (CD54) mAb in renal allograft recipients.Transplantation1993Apr;55476672discussion 772-3.
  146. 146. HarlanJ. MWinnR. KLeukocyte-endothelial interactions: clinical trials of anti-adhesion therapy.Crit Care Med 2002May;30(5 Suppl):S2149
  147. 147. EdelsteinC. LLingHSchrierR. WThe nature of renal cell injury.Kidney Int 1997May;51513411351
  148. 148. YatesP. JHosgoodS. ANicholsonM. LA biphasic response to nitric oxide donation in an ex vivo model of donation after cardiac death renal transplantation.J Surg Res 2012Jun 15;1752316321
  149. 149. Dal Secco DMoreira AP, Freitas A, Silva JS, Rossi MA, Ferreira SH, et al. Nitric oxide inhibits neutrophil migration by a mechanism dependent on ICAM-1: role of soluble guanylate cyclaseNitric Oxide2006Aug;1517786
  150. 150. CaumartinYStephenJDengJ. PLianDLanZLiuWet alCarbon monoxide-releasing molecules protect against ischemia-reperfusion injury during kidney transplantation.Kidney Int 2011May;791010801089
  151. 151. HantoD. WMakiTYoonM. HCsizmadiaEChinB. YGalloDet alIntraoperative administration of inhaled carbon monoxide reduces delayed graft function in kidney allografts in Swine.Am J Transplant 2010Nov;101124212430
  152. 152. HosgoodS. ANicholsonM. LHydrogen sulphide ameliorates ischaemia-reperfusion injury in an experimental model of non-heart-beating donor kidney transplantationBr J Surg 2010Feb;972202209
  153. 153. LiuY. HLuMBianJ. SHydrogen sulfide and renal ischemiaExpert Rev Clin Pharmacol 2011Jan;414961
  154. 154. CassisPAzzolliniNSoliniSMisterMAielloSCuginiDet alBoth darbepoetin alfa and carbamylated erythropoietin prevent kidney graft dysfunction due to ischemia/reperfusion in ratsTransplantation2011Aug 15;923271279
  155. 155. HuLYangCZhaoTXuMTangQYangBet alErythropoietin Ameliorates Renal Ischemia and Reperfusion Injury via Inhibiting Tubulointerstitial InflammationJ Surg Res 2011Jul 19.
  156. 156. MartinezFKamarNPalletNLangPDurrbachALebranchuYet alHigh dose epoetin beta in the first weeks following renal transplantation and delayed graft function: Results of the Neo-PDGF StudyAm J Transplant 2010Jul;10716951700
  157. 157. AydinZMallatM. JSchaapherderA. FVan ZonneveldA. JVan KootenCRabelinkT. Jet alRandomized Trial of Short-Course High-Dose Erythropoietin in Donation After Cardiac Death Kidney Transplant RecipientsAm J Transplant 2012Mar 19.
  158. 158. PatelN. SKerr-petersonH. LBrinesMCollinoMRogazzoMFantozziRet alThe delayed administration of pHBSP, a novel non-erythropoietic analogue of erythropoietin, attenuates acute kidney injury. Mol Med 2012Mar 8.
  159. 159. EldaifS. MDeneveJ. AWangN. PJiangRMosunjacMMutrieC. Jet alAttenuation of renal ischemia-reperfusion injury by postconditioning involves adenosine receptor and protein kinase C activationTranspl Int 2010Feb;232217226
  160. 160. SzwarcIMouradGArgilesAPost-conditioning to reduce renal ischaemia/reperfusion injuryNephrol Dial Transplant 2009Jul;24722889author reply 2289-90.
  161. 161. BasuSMeisertIEggenspergerEKriegerEKrennC. GTime course and attenuation of ischaemia-reperfusion induced oxidative injury by propofol in human renal transplantation.Redox Rep 2007124195202
  162. 162. SnoeijsM. GVaahteraLDe VriesE. ESchurinkG. WHaenenG. RPeutz-kootstraC. Jet alAddition of a water-soluble propofol formulation to preservation solution in experimental kidney transplantationTransplantation2011Aug 15;923296302
  163. 163. ZhuoWLiaoLXuTWuWYangSTanJMesenchymal stem cells ameliorate ischemia-reperfusion-induced renal dysfunction by improving the antioxidant/oxidant balance in the ischemic kidneyUrol Int 2011862191196
  164. 164. HaraYStolkMRingeJDehneTLadhoffJKotschKet alIn vivo effect of bone marrow-derived mesenchymal stem cells in a rat kidney transplantation model with prolonged cold ischemia.Transpl Int 2011Nov;241111121123
  165. 165. KwonOMillerSLiNKhanAKadryZUemuraTBone marrow-derived endothelial progenitor cells and endothelial cells may contribute to endothelial repair in the kidney immediately after ischemia-reperfusionJ Histochem Cytochem 2010Aug;588687694
  166. 166. CiancioGBurkeG. WGaynorJ. JRothDKupinWRosenAet alA randomized trial of thymoglobulin vs. alemtuzumab (with lower dose maintenance immunosuppression) vs. daclizumab in renal transplantation at 24 months of follow-upClin Transplant 2008Mar-Apr;222200210
  167. 167. BrennanD. CDallerJ. ALakeK. DCibrikDDel Castillo D, Thymoglobulin Induction Study Group. Rabbit antithymocyte globulin versus basiliximab in renal transplantation. N Engl J Med 2006Nov 9;3551919671977

Written By

Sarah A. Hosgood, James P. Hunter and Michael L. Nicholson

Submitted: 12 May 2012 Published: 19 December 2012