Guidelines For Clinical Practice Open Access
Copyright ©2009 Baishideng. All rights reserved.
World J Gastrointest Surg. Nov 30, 2009; 1(1): 21-25
Published online Nov 30, 2009. doi: 10.4240/wjgs.v1.i1.21
Future of bioartificial liver support
Robert AFM Chamuleau, Department of Hepatology, Academic Medical Center, University of Amsterdam, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands
Author contributions: Chamuleau RAFM contributed solely to this paper.
Correspondence to: Robert AFM Chamuleau, MD, PhD, Department of Hepatology, Academic Medical Center, S-Building, Floor 1, Room 166, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands. r.a.chamuleau@amc.uva.nl
Telephone: +31-20-5666832 Fax: +31-20-5669190
Received: October 21, 2009
Revised: October 28, 2009
Accepted: November 4, 2009
Published online: November 30, 2009

Abstract

Many different artificial liver support systems (biological and non-biological) have been developed, tested pre-clinically and some have been applied in clinical trials. Based on theoretical considerations a biological artificial liver (BAL) should be preferred above the non-biological ones. However, clinical application of the BAL is still experimental. Here we try to analyze which hurdles have to be taken before the BAL will become standard equipment in the intensive care unit for patients with acute liver failure or acute deterioration of chronic liver disease.

Key Words: Acute liver failure, Bioartificial liver, Liver transplantation, Cell transplantation, Tissue engineering, Xenotransplantation



INTRODUCTION

Nowadays intensive care doctors have many artificial devices to support their patients with failing organs. They possess different types of hemodialysis devices, artificial ventilation, artificial heart, aortic balloon pumping, blood oxygenators, heart-lung machines and are able to apply total parenteral nutrition in the patient with short bowel syndrome. However, the patient with acute liver failure (ALF) is still a major challenge[1].

ALF is a devastating clinical syndrome with a high mortality (60%-80%, depending on the cause and the experience of the clinical centre) with most frequent causes of death being brain edema, SIRS (systemic inflammatory response syndrome) and multiple organ failure (MOF). Emergency whole or partial liver transplantation (orthotopic (OLT) or auxiliary) is the only life-saving therapy.

Many attempts have been made to develop artificial liver support devices (ALSD): non-biological ones such as hemodialysis, charcoal hemoperfusion, selective plasma filtration, plasma exchange, hemo-diadsorption, albumin dialysis and biological ones such as whole liver perfusion, liver cell transplantation and bioartificial liver support.

The status of ALSDs has been the subject of many reviews, at least one a year, since 2001[2-14]. It does not seem wise to repeat their contents in this editorial and the reader is referred to the publications for global and/or detailed information. From reading them, at least one common conclusion emerges: devices that only support the failing detoxification function of the severely diseased liver are not sufficient to save the lives of ALF patients. It is generally accepted that the syndrome of ALF is not only determined by failing hepatic detoxification, but also by failing hepatic synthetic and regulatory function. This is also one of the conclusions of a workshop on ALF held in the USA in 2008[1].

The purpose of this editorial is, however, to analyze the critical issues that have to be solved in the near future.

CRITICAL ISSUES
What can we expect from cell or even organ-based ALSDs?

Whole animal liver perfusion as an ALSD seems to be a logic approach. There is some experience in a few case reports[15], but it has never been accepted as a common treatment, because of its complexity and its important xenotransplantation-related problems.

There are a few case reports concerning the more simple technique of liver cell transplantation (LCTX)[16] as a treatment for ALF. LCTX has at least 2 important drawbacks: (1) the availability of sufficient amounts of fully differentiated human liver cells; (2) the so far unsolved problem of transplanting large amounts (at least 10% of the normal parenchymal mass) of cells where there is adequate blood supply.

Ideally, a tissue-engineered transplantable liver should be the final solution. Such a bioengineered liver (BEL) should resemble the native liver as much as possible. This means a composite of parenchymal and non-parenchymal liver cells in a sponge-like configuration in which a vascular system provides, by direct plasma contact, oxygen and nutrients to the liver cells and which is equipped with a biliary outflow system. Ideally this BEL has to be connected to the splanchnic circulation (inflow tract), the caval vein system (outflow tract) and the intestine (biliary tract).

At present, such a BEL is only in a very preliminary and experimental phase[17-22] and, as second best to liver transplantation, patients have to be treated by one of the existing bioartificial livers (BALs) that can only be connected outside the body to the patient’s systemic blood circulation

A BAL is defined as a bioreactor charged with liver cells that is connected outside the body to the blood or plasma circulation of the patient. Since a BAL supports both the failing detoxification and the failing synthetic and regulatory function of the diseased liver, it should have a beneficial effect on the degree of hepatic coma and the severity of MOF and, last but not least, on survival of ALF patients and preferably also of patients with acute on chronic liver disease (AoCLD).

In general, 4 types of BAL bioreactors can be distinguished: hollow fiber; flat plate and monolayer; perfused beds/scaffolds; encapsulation/suspension. Every system has its pros and cons. For details see Allen et al[2].

To prove their right to belong to the standard equipment of an intensive care unit BALs need to be validated in randomized, controlled clinical trials. Several questions have arisen as to whether pre-clinical research on BALs has been sufficient to justify their clinical application.

How good are results in experimental animals?

In general, the answer is positive. In many different models of ALF several BALs based on animal liver cells have shown to prolong survival significantly in comparison to standard treatment[23-33].

Can it contain a sufficient mass of parenchymal liver cells?

It is generally accepted (based on safe surgical resections) that survival is possible with a minimum of 20% of liver mass with optimal functionality. Assuming that the ALF patient still has some residual functioning liver mass, a BAL should contain at least 15% of liver mass. However, the reality is that isolated liver cells in a bioreactor do not have optimal functionality, so more than 15% (preferably 20%-30%) of liver mass will be required[34].

Furthermore, it is well known that parenchymal liver cells function at best in a 3-dimensional (3D) configuration. In addition their functionality increases when they are co-cultured with non-parenchymal cells[35] For these reasons, the ideal BAL should contain at least a mixture of well-differentiated liver cells in a 3D configuration at a mass of at least 20% of the normal liver (200 g cells in 1 kg of liver). Vital Therapies ELAD® (Extracorporeal Liver Assist Device) and Hep-Art AMC-BAL have this capacity.

How is bi-directional mass transport of oxygen, carbon dioxide, nutrients and liver cell products best guaranteed?

In BAL devices, bi-directional mass transfer is needed to provide nutrients to sustain cell viability and allow export of therapeutic cell products. Although most device designs address this, there are important limitations involving the use of semi-permeable membranes as a barrier between plasma and the bio-component. Bioreactors in which direct contact between plasma and the liver cells is guaranteed or those using semi-permeable membranes with high porosity are preferred.

In addition, liver cells need sufficient oxygen supply to function optimally[36]. The amount of oxygen actually dissolved in plasma is insufficient in this respect. Therefore, the cells in the bioreactor should see either full blood (with many problems such as hemolysis, clotting and platelet loss) or plasma with an extra oxygen carrier such as fluorocarbons[37] or locally supplied oxygen by oxygen capillaries interwoven with the cell containing hollow fibers (Modular Extracorporeal Liver Support)[38] or matrix (AMC-BAL)[39] inside the BAL: a so-called internal oxygenator.

Do BALs support drainage of bile?

Another aspect of current BALS is the universal absence of functional biliary excretion into an isolated compartment. Liver cells in 3D configuration can form functional canaliculi, but it is unknown to what extent biliary compounds still accumulate intracellularly and whether these will shorten the vitality of the cells. If some export of biliary compounds occurs at the basal lateral side to the plasma compartment a hybrid system removing them from this compartment is a logic next step. This might mean a modular system in which a BAL is combined with an artificial liver support device such as hemodialysis, charcoal hemoperfusion or albumin dialysis[40].

How long do cells remain viable and functional?

Cell viability is of paramount importance for the life supporting capacity of a BAL. The experience is that primary liver cells in a bioreactor lose functionality over time. With this already being the case under optimal culture conditions, it is especially problematic when the environment of cells is 100% human plasma. A decrease in function is even more marked if cells have to live in the plasma of ALF patients[41-46]. Increased concentrations of toxic products and probably decreased concentrations of essential nutrients play a role in this regard. For this reason, BALs are only temporarily sufficiently functional and have probably to be replaced after a critical time by fresh ones.

Which cells can be used in the BAL?

Freshly isolated or cryopreserved porcine liver cells or a human hepatoma cell line have been most frequently used as the biocomponent in clinically applied bioartificial livers.

Because of the xenotransplantation-related disadvantages of porcine cells (immunological reactions and possible pig endogenous retrovirus transmission)[47-50] and the shortage of primary human hepatocytes, a well-differentiated human liver cell line seems to be the Holy Grail. Such a cell line will have minimal immunogenicity, no risk of xenozoonosis and required functionality and availability.

Primary sources for the development of such human cell lines are human liver tumor derived cell lines, immortalized fetal or adult hepatocytes and stem cells of hepatic, hematopoietic, mesenchymal or embryonic origin. However, in all cell types tested so far, the in vitro differentiation cannot be stimulated to such an extent that functionality reaches that of primary human hepatocytes. The future lies in having more insight into differentiation-promoting factors and the influence of matrix and co-culture conditions on the functionality of liver cell lines[51].

What is the current situation?

A few BAL systems are currently in the process of being commercialized (Table 1).

Table 1 BALs to be commercialized.
CompanyDeviceCharacteristicsClinical experience & future plans
HepaLifeHepa-Mate™ (previously HepatAssist)Cryopreserved porcine cells, treatment 3-6 h for 1-5 d, charcoal column, and centrifugal plasmapheresis. Cell mass previously 60 g, in future trial 160 gPhase II/III with HepatAssist in 171 ALF patients, only 9% improvement in OLT/NR as compared to controls. New trial in preparation
Vital therapiesELAD®Two-chambered hollow fiber cartridge with immortalized human C3A cell line. Treatment up to 10 d. Ultrafiltrate perfusion. 4 replaceable cartridges. Cell mass 4 g × 200 gControlled study with 25 ALF patients completed. 92% recovery OLT/NR. Controlled clinical trial in 49 AoCLD patients in China
Beijing and Nanjing UniversitiesTECA-BALSS/HBALPorcine cells (10-20 billion cells), outside compartment of hollow fiber devicesPhase I, 15 patients ALF and 3 patients AoCLD
Hep-ArtAMC-BALPerfused scaffold, oxygenation in situ, 10-15 billion freshly isolated SPF porcine hepatocytesPhase I/IIa; 14 ALF patients. Safe, no PERV transmission

Vital Therapies just finished a controlled clinical trial in 49 AoCLD patients in China. At its website (http://www.vitaltherapies.com) one can read: “The pivotal China trial enrolled 49 patients and was carried out to support the registration of ELAD in China. It demonstrated statistically significant improvement in transplant free survival for acute-on-chronic liver failure patients treated with ELAD compared to the control group. These were mostly hepatitis B patients. VTI filed an application for marketing approval with the China SFDA in September 2007 and this application remains under review. These results remain to be confirmed in studies outside China”.

HepaLife (http://www.hepalife.com) is promoting the Demetriou system (formerly brought by Circe and Arbios) that is based on cryopreserved porcine liver cells combined with a charcoal column connected to a plasmapheresis circuit. More than 200 patients have been treated by this system. In a multicenter controlled clinical trial in 181 ALF patients, time to death was significantly prolonged only in a subgroup of 83 patients with ALF of known etiology.

The Chinese ALSDs (TECA BALSS and HBAL) and the Dutch AMC-BAL have been tested in Phase 1-2a trials but are not yet commercially available.

Why is clinical proof of efficacy rather limited?

There are a few explanations: (1) The hardware used for bioreactors has not always been optimal. Hollow fiber-based bioreactors will have mass transfer restrictions and the absence of an internal oxygenator will limit cell functionality if plasma perfusion is the approach to the patient’s blood circulation. In addition not all BALs have a 3D configuration of the liver cells. (2) The optimal human liver cell line is still not available. Hepatoma-derived cell lines are not fully differentiated, nor are immortalized liver cell lines. Future developments in this regard are urgently needed. (3) In the already published clinical trials, patient populations have been rather diverse making intention-to-treat analyses disappointing. (4) If BAL treatment is applied in ALF patients to bridge the waiting time for OLT, post-transplantation survival is not only dependent on BAL treatment but also on OLT.

CONCLUSION

Taking all these considerations together there is certainly a future for the BAL, based on pre-clinical data and the lessons that have been drawn from the existing controlled trials. A well-differentiated human liver cell line is still the Holy Grail. If this cell line were available, future clinical trials should be done with it in a BAL consisting of minimal mass transfer restrictions and equipped with cell oxygenation in situ, loaded with a sufficient 3D mass. Eventually it should be combined with albumin dialysis and refreshed after a critical time. The trial population should be as homogeneous as possible and well defined with regard to survival capacity.

POSSIBLE CONFLICT OF INTEREST

Chamuleau RAFM is CSO of Hep-Art Medical Devices B.V. that produces the AMC-BAL.

Footnotes

Peer reviewer: Dr. Gianpiero Gravante, Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester General Hospital, 38 Hospital Close, Leicester, LE54WU, United Kingdom

S- Editor Li LF L- Editor Cant MR E- Editor Lin YP

References
1.  Lee WM, Squires RH Jr, Nyberg SL, Doo E, Hoofnagle JH. Acute liver failure: Summary of a workshop. Hepatology. 2008;47:1401-1415.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Allen JW, Hassanein T, Bhatia SN. Advances in bioartificial liver devices. Hepatology. 2001;34:447-455.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Hayes PC, Lee A. What progress with artificial livers? Lancet. 2001;358:1286-1287.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Strain AJ, Neuberger JM. A bioartificial liver--state of the art. Science. 2002;295:1005-1009.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Planchamp C, Vu TL, Mayer JM, Reist M, Testa B. Hepatocyte hollow-fibre bioreactors: design, set-up, validation and applications. J Pharm Pharmacol. 2003;55:1181-1198.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  van de Kerkhove MP, Hoekstra R, Chamuleau RA, van Gulik TM. Clinical application of bioartificial liver support systems. Ann Surg. 2004;240:216-230.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Wigg AJ, Padbury RT. Liver support systems: promise and reality. J Gastroenterol Hepatol. 2005;20:1807-1816.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Park JK, Lee DH. Bioartificial liver systems: current status and future perspective. J Biosci Bioeng. 2005;99:311-319.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Rozga J. Liver support technology--an update. Xenotransplantation. 2006;13:380-389.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  O'Grady J. Personal view: current role of artificial liver support devices. Aliment Pharmacol Ther. 2006;23:1549-1557.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Santoro A, Mancini E, Ferramosca E, Faenza S. Liver support systems. Contrib Nephrol. 2007;156:396-404.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Stadlbauer V, Jalan R. Acute liver failure: liver support therapies. Curr Opin Crit Care. 2007;13:215-221.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Phua J, Lee KH. Liver support devices. Curr Opin Crit Care. 2008;14:208-215.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Sgroi A, Serre-Beinier V, Morel P, Bühler L. What clinical alternatives to whole liver transplantation? Current status of artificial devices and hepatocyte transplantation. Transplantation. 2009;87:457-466.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Abouna GM, Ganguly P, Jabur S, Tweed W, Hamdy H, Costa G, Farid E, Sater A. Successful ex vivo liver perfusion system for hepatic failure pending liver regeneration or liver transplantation. Transplant Proc. 2001;33:1962-1964.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Galvão FH, de Andrade Júnior DR, de Andrade DR, Martins BC, Marson AG, Bernard CV, Dos Santos SA, Bacchella T, Machado MC. Hepatocyte transplantation: State of the art. Hepatol Res. 2006;36:237-247.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Chan C, Berthiaume F, Nath BD, Tilles AW, Toner M, Yarmush ML. Hepatic tissue engineering for adjunct and temporary liver support: critical technologies. Liver Transpl. 2004;10:1331-1342.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Diekmann S, Bader A, Schmitmeier S. Present and Future Developments in Hepatic Tissue Engineering for Liver Support Systems : State of the art and future developments of hepatic cell culture techniques for the use in liver support systems. Cytotechnology. 2006;50:163-179.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Fiegel HC, Kaufmann PM, Bruns H, Kluth D, Horch RE, Vacanti JP, Kneser U. Hepatic tissue engineering: from transplantation to customized cell-based liver directed therapies from the laboratory. J Cell Mol Med. 2008;12:56-66.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Ishii Y, Saito R, Marushima H, Ito R, Sakamoto T, Yanaga K. Hepatic reconstruction from fetal porcine liver cells using a radial flow bioreactor. World J Gastroenterol. 2008;14:2740-2747.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Linke K, Schanz J, Hansmann J, Walles T, Brunner H, Mertsching H. Engineered liver-like tissue on a capillarized matrix for applied research. Tissue Eng. 2007;13:2699-2707.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Sarkis R, Wen L, Honiger J, Baudrimont M, Delelo R, Calmus Y, Capeau J, Nordlinger B. [Intraperitoneal transplantation of isolated hepatocytes of the pig: the implantable bioartificial liver]. Chirurgie. 1998;123:41-46.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Flendrig LM, Calise F, Di Florio E, Mancini A, Ceriello A, Santaniello W, Mezza E, Sicoli F, Belleza G, Bracco A. Significantly improved survival time in pigs with complete liver ischemia treated with a novel bioartificial liver. Int J Artif Organs. 1999;22:701-709.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Abrahamse SL, van de Kerkhove MP, Sosef MN, Hartman R, Chamuleau RA, van Gulik TM. Treatment of acute liver failure in pigs reduces hepatocyte function in a bioartificial liver support system. Int J Artif Organs. 2002;25:966-974.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Enosawa S, Miyashita T, Saito T, Omasa T, Matsumura T. The significant improvement of survival times and pathological parameters by bioartificial liver with recombinant HepG2 in porcine liver failure model. Cell Transplant. 2006;15:873-880.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Flendrig LM, Chamuleau RA, Maas MA, Daalhuisen J, Hasset B, Kilty CG, Doyle S, Ladiges NC, Jörning GG, la Soe JW. Evaluation of a novel bioartificial liver in rats with complete liver ischemia: treatment efficacy and species-specific alpha-GST detection to monitor hepatocyte viability. J Hepatol. 1999;30:311-320.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Hochleitner B, Hengster P, Bucher H, Ladurner R, Schneeberger S, Krismer A, Kleinsasser A, Barnas U, Klima G, Margreiter R. Significant survival prolongation in pigs with fulminant hepatic failure treated with a novel microgravity-based bioartificial liver. Artif Organs. 2006;30:906-914.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Kamohara Y, Fujioka H, Eguchi S, Kawashita Y, Furui J, Kanematsu T. Comparative study of bioartificial liver support and plasma exchange for treatment of pigs with fulminant hepatic failure. Artif Organs. 2000;24:265-270.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kawazoe Y, Eguchi S, Sugiyama N, Kamohara Y, Fujioka H, Kanematsu T. Comparison between bioartificial and artificial liver for the treatment of acute liver failure in pigs. World J Gastroenterol. 2006;12:7503-7507.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Khalili TM, Navarro A, Ting P, Kamohara Y, Arkadopoulos N, Solomon BA, Demetriou AA, Rozga J. Bioartificial liver treatment prolongs survival and lowers intracranial pressure in pigs with fulminant hepatic failure. Artif Organs. 2001;25:566-570.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Sheil AG, Sun J, Mears DC, Waring M, Woodman K, Johnston B, Horvat M, Watson KJ, Koutalistras N, Wang LS. Preclinical trial of a bioartificial liver support system in a porcine fulminant hepatic failure model. Aust N Z J Surg. 1996;66:547-552.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Sosef MN, Abrahamse LS, van de Kerkhove MP, Hartman R, Chamuleau RA, van Gulik TM. Assessment of the AMC-bioartificial liver in the anhepatic pig. Transplantation. 2002;73:204-209.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Tréhout D, Desille M, Doan BT, Mahler S, Frémond B, Mallédant Y, Campion JP, Desbois J, Beloeil JC, de Certaines J. Follow-up by one- and two-dimensional NMR of plasma from pigs with ischemia-induced acute liver failure treated with a bioartificial liver. NMR Biomed. 2002;15:393-403.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Morsiani E, Brogli M, Galavotti D, Pazzi P, Puviani AC, Azzena GF. Biologic liver support: optimal cell source and mass. Int J Artif Organs. 2002;25:985-993.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Kidambi S, Yarmush RS, Novik E, Chao P, Yarmush ML, Nahmias Y. Oxygen-mediated enhancement of primary hepatocyte metabolism, functional polarization, gene expression, and drug clearance. Proc Natl Acad Sci USA. 2009;106:15714-15719.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Catapano G, De BL. Combined effect of oxygen and ammonia on the kinetics of ammonia elimination and oxygen consumption of adherent rat liver cells. Int J Artif Organs. 2002;25:151-157.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Nieuwoudt M, Engelbrecht GH, Sentle L, Auer R, Kahn D, van der Merwe SW. Non-toxicity of IV injected perfluorocarbon oxygen carrier in an animal model of liver regeneration following surgical injury. Artif Cells Blood Substit Immobil Biotechnol. 2009;37:117-124.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Gerlach JC, Lemmens P, Schön M, Janke J, Rossaint R, Busse B, Puhl G, Neuhaus P. Experimental evaluation of a hybrid liver support system. Transplant Proc. 1997;29:852.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Flendrig LM, la Soe JW, Jörning GG, Steenbeek A, Karlsen OT, Bovée WM, Ladiges NC, te Velde AA, Chamuleau RA. In vitro evaluation of a novel bioreactor based on an integral oxygenator and a spirally wound nonwoven polyester matrix for hepatocyte culture as small aggregates. J Hepatol. 1997;26:1379-1392.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Sauer IM, Zeilinger K, Pless G, Kardassis D, Theruvath T, Pascher A, Goetz M, Neuhaus P, Gerlach JC. Extracorporeal liver support based on primary human liver cells and albumin dialysis--treatment of a patient with primary graft non-function. J Hepatol. 2003;39:649-653.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Newsome PN, Tsiaoussis J, Ansell I, Ross JA, Sethi T, Hayes PC, Plevris JN. Acute liver failure serum reduces hepatocyte-matrix adhesion by a cell death-independent mechanism. J Hepatol. 2001;34 Suppl 1:29.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Anderson C, Thabrew MI, Hughes RD. Assay to detect inhibitory substances in serum of patients with acute liver failure. Int J Artif Organs. 1999;22:113-117.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Newsome PN, Tsiaoussis J, Masson S, Buttery R, Livingston C, Ansell I, Ross JA, Sethi T, Hayes PC, Plevris JN. Serum from patients with fulminant hepatic failure causes hepatocyte detachment and apoptosis by a beta(1)-integrin pathway. Hepatology. 2004;40:636-645.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Hughes RD, Yamada H, Gove CD, Williams R. Inhibitors of hepatic DNA synthesis in fulminant hepatic failure. Dig Dis Sci. 1991;36:816-819.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Mitry RR, Hughes RD, Bansal S, Lehec SC, Wendon JA, Dhawan A. Effects of serum from patients with acute liver failure due to paracetamol overdose on human hepatocytes in vitro. Transplant Proc. 2005;37:2391-2394.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Saich R, Selden C, Rees M, Hodgson H. Characterization of pro-apoptotic effect of liver failure plasma on primary human hepatocytes and its modulation by molecular adsorbent recirculation system therapy. Artif Organs. 2007;31:732-742.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Lee JH, Moran C. Current status of xenotransplantation - A review. Asian Australas J Anim Sci. 2001;14:1497-1504.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Patience C, Patton GS, Takeuchi Y, Weiss RA, McClure MO, Rydberg L, Breimer ME. No evidence of pig DNA or retroviral infection in patients with short-term extracorporeal connection to pig kidneys. Lancet. 1998;352:699-701.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Wang HH, Wang YJ, Liu HL, Liu J, Huang YP, Guo HT, Wang YM. Detection of PERV by polymerase chain reaction and its safety in bioartificial liver support system. World J Gastroenterol. 2006;12:1287-1291.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Xu H, Sharma A, Okabe J, Cui C, Huang L, Wei YY, Wan H, Lei Y, Logan JS, Levy MF. Serologic analysis of anti-porcine endogenous retroviruses immune responses in humans after ex vivo transgenic pig liver perfusion. ASAIO J. 2003;49:407-416.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Chamuleau RA, Deurholt T, Hoekstra R. Which are the right cells to be used in a bioartificial liver? Metab Brain Dis. 2005;20:327-335.  [PubMed]  [DOI]  [Cited in This Article: ]