Minireviews Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Transplant. Dec 24, 2017; 7(6): 317-323
Published online Dec 24, 2017. doi: 10.5500/wjt.v7.i6.317
Hepatocyte transplantation: Consider infusion before incision
Ryan D Heath, Bhupinder S Romana, Jamal A Ibdah, Veysel Tahan, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, United States
Furkan Ertem, Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, United States
ORCID number: Ryan D Heath (0000-0002-2072-4028); Furkan Ertem (0000-0001-6151-7925); Bhupinder S Romana (0000-0002-2244-9701); Jamal A Ibdah (0000-0002-5646-9014); Veysel Tahan (0000-0001-6796-9359).
Author contributions: All authors contributed to the acquisition of data, writing, and revision of this manuscript.
Conflict-of-interest statement: All the authors have no conflicts of interests to declare. All authors of this article declare that there is no conflicting interests (including but not limited to commercial, personal, political, intellectual or religious interests) that are related to the work submitted for consideration of publication. The statement of “No conflict of interest” file is signed by each author and attached.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Veysel Tahan, MD, FACP, FACG, FESBGH, Associate Professor, Division of Gastroenterology and Hepatology, University of Missouri, 1 Hospital Dr, Columbia, MO 65212, United States. tahanv@health.missouri.edu
Telephone: +1-573-8846044 Fax: +1-573-8844595
Received: October 28, 2017
Peer-review started: October 29, 2017
First decision: November 20, 2017
Revised: November 27, 2017
Accepted: December 5, 2017
Article in press: December 6, 2017
Published online: December 24, 2017
Processing time: 54 Days and 16.6 Hours

Abstract

Human hepatocyte transplantation is undergoing study as a bridge, or even alternative, to orthotopic liver transplantation (OLT). This technique has undergone multiple developments over the past thirty years in terms of mode of delivery, source and preparation of cell cultures, monitoring of graft function, and use of immunosuppression. Further refinements and improvements in these techniques will likely allow improved graft survival and function, granting patients higher yield from this technique and potentially significantly delaying need for OLT.

Key Words: Hepatocye, Transplantation, Cell therapy, Liver, Graft, Orthotopic

Core tip: Further human studies involving humans are needed, however, the current collectively suggest progress in terms of improved effectiveness of human hepatocyte transplantation (HTx). With improvements in optimizing delivery technique and assessing proper recipients of livers, monitoring graft function, as well as recognizing and treating graft rejection, HTx may be able to be used more widely in metabolic liver disease and potentially delay necessity of orthotopic liver transplantation.



INTRODUCTION

Human hepatocyte transplantation (HTx) is being studied as a potential future alternative and currently use as a bridge to orthotropic liver transplantation (OLT). Over the last 10 years it has been noted that the number of patients requiring transplant as well as total transplants being performed has been stable (NIHMS). Given the inadequate supply of donor organs in relation to patients who would benefit from transplantation, continued research into alternate therapies for treatment or to prolong time before transplantation becomes necessary is timely. HTx is a technique which has been refined over the past three decades which seeks to improve liver function via transplantation of donor hepatocytes directly, rather than transplanting an entire organ. While a number of disorders have been evaluated for efficacy of therapy with this technique, individuals with inborn errors of metabolism appear the greatest benefit[1,2]. Sustained benefits have not been observed, however, refinements in the practice may lead to greater temporal benefits. While this review aims to summarize use of HTx in studies, it also seeks to highlight potential shortcomings of previously utilized technique and focus on areas of future study which may lead to improved yield of HTx.

PREPARATION OF HEPATOCYTE CULTURES

While many consider avoidance and delay of surgery appealing when considering HTx compared to OLT, it should be noted that the source of hepatocytes utilized for HTx generally come from livers deemed unsuitable for OLT[1-4]. The most common reason for rejection of a liver for OLT being steatosis, which is associated with both lower cell viability and yield[5-7]. Ischemic damage to livers is also a common reason for rejection, similarly affecting the yield and viability of extracted hepatocytes[8]. That stated, there is evidence that high quality hepatocytes may be obtained from cardiac death donors with prolonged warm ischemia, though prolonged episodes of ischemia predictably decreases viability[9,10]. While a current argument in favor of expanding research and use of HTx is that one is able to utilize cells from a larger pool of donor organs, one suspects that use of hepatocytes for HTx cultured from livers deemed suitable for OLT would likely result in greater success of this therapy. Beyond simple increased efficacy, multiple recipients could benefit from a single donor liver. Admittedly, there are concerns regarding evaluating the fitness of a recipient to receive donor hepatocytes. For instance, the cytochrome P450 enzyme is involved metabolism of drugs and steroids, bile synthesis, cholesterol synthesis, and vitamin D production. This enzyme system has been noted to have different levels of expression and function within humans, however, and this variability may be partially responsible for variant viability of HTx[11-13]. Not every person may be fit to receive any donor hepatocyte culture due to pre-existing chronic condition or associated medication they take, however, increasing the donor pool would still likely increase overall access to HTx. Furthermore, cell cultures can be cryopreserved and stored until needed, whereas there is a finite amount of time a whole liver can be stored before it is no longer viable for OLT[14].

CLINICAL INDICATIONS FOR HEPATOCYTE TRANSPLANTATION

Further discussion of refinement in technique warrants first discussing potential clinical indications for its use. As previously noted, congenital metabolic disorders appear to hold the greatest promise for use of HTx as metabolism of substrates in questions occur almost exclusively in hepatocytes. The cases reviewed below demonstrate HTx as a successful bridge to OLT.

Crigler-Najjar syndrome (CN) TypeIis an autosomal recessive condition with complete absence of a uridine diphosphate glucuronosyltransferase (UDPGT) enzymes, resulting in life threatening unconjugated hyperbilirubinemia with long term risk of kernicterus. While phototherapy can be an effective treatment, its effectiveness has been observed to decrease with increased age[15]. The first hepatocyte transplantation was performed in a rat model deficient in UDPGT enzymes[16-18]. A minimal percentage of liver mass comprised of engrafted cells (0.2%), resulted in a 40% decrease in unconjugated bilirubin levels[18]. Humans with CN TypeIhave subsequently undergone HTx with marked improvement in unconjugated bilirubinemia, however, all patients subsequently required OLT anywhere from 4 to 20 mo after HTx due to either loss of graft or insufficient improvement in symptoms[19-23].

Urea cycle disorders, comprising a group of disorder due to deficiencies in one of six different enzymes in the urea cycle, are another group seemingly optimally situated to benefit from HTx. These deficiencies collectively result in hyperammonemia with significant neurologic sequelae. Most patients present as neonates, with current therapy involving protein restriction, hemodialysis, or hemofiltration. Hyperammonemia is still noted despite these treatments, however, with OLT being the only current definitive treatment. Humans have successfully undergone HTx as a bridge to whole organ transplantation, with stabilization of ammonia metabolism noted between 4-13 mo before OLT became necessary[24,25].

Familial hypercholesterolemia (FH) is caused by absence of the low density lipoprotein receptor (LDLR) resulting in early onset severe coronary artery disease. Low density lipoprotein (LDL) apheresis or OLT are the only current treatments, however, a rabbit model of FH undergoing HTx was noted to have decreased levels of serum cholesterol by 30%-60% for 100 d[26-28]. In 1995, 5 patients between the ages of 7 and 41 underwent HTx, demonstrating up to a 20% reduction in LDL in three of the patients, the other 2 not responding to therapy[29].

Glycogen storage disease TypeI(GSD-I) is an autosomal recessive metabolic disorder resulting from deficiency of the hepatic enzymes glucose-6-phosphatase (Ia) or glucose-6-phosphate transporter (Ib), resulting in deficiency in glucose production with noted severe hypoglycemia, lactic acidosis, hyperlipidemia, growth retardation, hyperuricemia, and renal dysfunction. While many patients can be treated with consumption of starch, some are unresponsive to dietary therapy and require OLT to correct the underlying defect[30]. Two patients, 18 and 47 years old, underwent HTx with subsequent ability to maintain unaltered diet for up to 7-9 mo[1,31].

Infantile Refsum disease is an autosomal recessive disorder characterized by impaired peroxisome function, resulting in accumulation of very long chain fatty acids and branched chain fatty acids which are normally degraded in peroxisomes. Patients present with severe neurologic defects and rarely survive beyond age 10, with treatment generally centering around supportive care[32]. One 4 years old female patient underwent HTx, demonstrating significant biochemical improvement for more than 16 mo[33].

HTx has been suggested as a treatment for Hemophilia A and B; with murine models demonstrating in 5%-10% increase in factor VIII and 1%-2% increase in factor IX[34,35]. These increases do result in decreased bleeding time and do provide a therapeutic benefit. In one 2004 study, a 3 mo and 35 mo old patients underwent HTx with 70% reduction in factor VII requirements noted after 6 mo, however, both patients eventually underwent OLT[36].

Progressive familial intrahepatic cholestasis (PFIC) encompasses a group of autosomal recessive liver diseases presenting in infancy and childhood with progressive cholestasis of hepatocellular origin, with three subtypes noted involving different components of bile metabolism[37]. Murine models of this disease process demonstrated improved bile metabolism using intrasplenic HTx[38]. Two children have been treated with HTx, however, both required OLT after 5 and 14 mo. Biopsies of the livers demonstrated extensive fibrosis and no donor cells on pathology before transplantation, the conclusion made that existing fibrosis likely impaired engraftment[39].

Phenylketonuria (PKU) is one of the most common inborn errors of metabolism, a deficiency of the enzyme phenylalanine hydroxyls (PAH) resulting in toxic concentrations of phenylalanine, the only current treatment involving phenylalanine restricted diet[40]. Murine models demonstrate significant improvement in PAH levels[41].

Alpha 1 antitrypsin (A1AT) deficiency - in 1997, a 52 years old patient underwent HTx as a bridge to transplant, with wild type A1AT levels were noted to increase in the interval between OLT, which occurred 2 d later[42].

This list is not exhaustive, however, it serves to illustrate the potential for this route of therapy in a large number of disorders mediated by hepatocyte dysfunction and subsequent metabolic derangements. While temporary improvements have been noted, others suspect the temporal benefits of HTx could be extended with transplantation of adequate cell mass, improved stock and implantation of transplanted hepatocytes, evaluating the ideal route of delivery, and improved and more accurate monitoring of graft function with emphasis on timely detection of rejection (Table 1).

Table 1 Summary of hepatocyte transplantation reports in human patients.
Ref.IndicationNo. of patientsInfusion site Outcome
Ambrosino et al[21]Criggler-Najjar TypeIA 9-year-old boyPortal veinDecreased bilirubin approximately 4 mo, underwent OLT
Lysy et al[22]Criggler-Najjar TypeIA 9-year-old girlJejunal veinDecreased bilirubin approximately 6 mo, underwent OLT
Lysy et al[22]Criggler-Najjar TypeIA 1-year-old girlSplenic veinDecreased bilirubin approximately 4 mo, underwent OLT
Zhou et al[53]Criggler-Najjar TypeI2 (4-mo-old boy and newborn boy)Portal veinDecreased bilirubin approximately 3-4 mo with subsequent OLT
Meyburg et al[24]Urea cycle disorders4 (1-d to 3-year-old)Portal veinStable 4-13 mo before OLT, 1 death at 4-mo
Grossman et al[29]Familial hyper-cholesterolemia5 (7-year-old to 41-year-old)Portal veinThree patients with approximately 40% reduction in LDL lasting 4 mo
Lee et al[31]Glycogen storage disordersA 8-year-old kidPortal veinFollowed for 7 mo, on tacrolimus and able to fast for 7 h without hypoglycemia
Muraca et al[1]Glycogen storage disorders47-year-old, femalePortal veinFollowed for 9 mo, on tacrolimus and able to fast for 7 h without hypoglycemia
Sokal et al[33]Refsum disease4-year-old girlPortal vein16 mo improvement
Dhawan et al[36]Hemophilia A2 (3-mo-old and 35-mo-old)Portal vein6 mo with 70% reduction in Factor VII requirements
Hansel et al[42]A1AT deficiencyA 52-year-oldPortal veinA1AT levels did increase before OLT available 2 d later
Soltys et al[52]Phenyl-ketonuriaA 27-year-old femalePortal vein7 mo of unrestricted diet
METHODS OF DELIVERY

Regarding adequate transplantation, two issues warrant discussion. One, culturing of hepatocytes from livers deemed unsuitable for OLT, has been previously discussed. Another issue regarding viability deals with transplantation of “fresh” vs cryopreserved hepatocytes. Fresh hepatocytes do demonstrate higher viability, with cryopreserved hepatocytes observed to have mitochondrial respiratory chain alterations and decreased ATP production[43]. Furthermore, protein synthesis has been noted to be impaired in cryopreserved cells relative to fresh hepatocytes[44,45]. A 2013 cohort study compared viability of freshly isolated hepatocytes against cryopreserved hepatocytes at 24, 48, and 72 h[46]. Freshly isolated hepatocytes demonstrated mean viability of approximately 81%, while means viability was approximately 61% at 24 h, 52% at 48 h, and 48% at 72 h. There was no noted increased caspase activity, an enzyme involved in apoptosis, though there did appear to some mild derangement in Cytochrome activities, previously noted above to be involved in hepatic metabolism of many different substrates.

Hepatocytes have been transplanted into the liver, spleen, and peritoneal cavity, with intraportal injection being the preferred and most physiological site for clinical transplantation[14,42]. This site may be accessed via percutaneous trans-hepatic puncture, cannulation of the umbilical vein, or open cannulation of a mesenteric vein[14]. Shear stress from catheterization can have an effect on viability, however, it has been demonstrated that catheters as small as 4.2 F are associated with acceptable viability[47]. Portal hypertension and any thrombosis are associated with lower engraftment levels, however, use of heparin infusion has been proposed as a potential mechanism to improve engraftment[48]. In cases of known portal hypertension, the spleen may be used as an alternate engraftment site, however, there are cases of splenic necrosis after injection into the splenic artery[8,49]. The peritoneal cavity is another alternate site, however, engraftment levels and long term viability of the graft have been observed to be significantly lower than portal vein infusion[50,51]. There are studies comparing efficacy of any method of delivery against another, and proper determination of the relative efficacy of each would be invaluable toward design of future studies evaluating HTx.

MONITORING GRAFT FUNCTION

Beyond just the method of delivery, appropriate pre-treatment of the recipient has been evaluated to improve efficacy of HTx. A 2017 case series details use of pre-operative liver-directed radiation[52]. Preoperative liver-directed irradiation has been noted to demonstrate complete correction of the bilirubin conjugation defect noted in rat models of Criger-Najjar syndrome TypeIfollowing HTx[53]. This case series demonstrated improved function of HTx from porcine hepatocytes comparing primates receiving hepatic pre-irradiation vs those who did not. Function was assessed by measuring levels of porcine albumin after HTx; pre-irradiated subjects demonstrated significantly higher levels of this protein than control subjects. Using immunohistochemical staining, spatial analysis of stained recipient liver tissue post HTx demonstrated level of engraftment to be approximately 11.8% in experimental subjects vs approximately 5% in control subjects. Survival of the graft appears improved in the pre-treated group appears improved as well, with no evidence of infiltrating T cells or macrophages noted in cells of the experimental group. Given the promising nature of the above results, two children with urea cycle defects were subsequently infused after undergoing the irradiation preconditioning protocol. One child was 4 mo of the age, the other underwent HTx shortly after birth. Regarding the patient receiving HTx shortly after birth, at 26 h, cell viability was noted to be approximately 63% with ammonia metabolism noted at normal levels[52]. One patient was noted to have intermittent episodes of hyperammonemia, however, it was noted that goal tacrolimus levels post-transplant were not sustained. This patient did eventually undergo OLT at 3.5 mo of age. The other patient maintained normal levels of ammonia for approximately 40 d, however, was not to have intermittent episodes of hyperammonemia after this point. On day 84 acutely increased levels of ammonia, glutamine, and urinary orotic acid suggested graft failure.

This same case series included a 27 years old female patient with PKU also undergoing HTx after irradiation pretreatment, doing well for 7 mo on an unrestricted diet before demonstrating evidence of rejection. Tacrolimus levels were again noted to be below goal level, and the patient was treated with corticosteroids and augmented immunosuppression protocol with phenylalanine tolerance returning. Phenylalanine levels remained normal for over one year, however, the patient’s follow became inconsistent and adequate monitoring of immunosuppression was not performed. Her brother, also afflicted with PKU, was used a control agent. At this point in the study, the graft was assumed to be rejected and immunosuppression discontinued without any adverse effects noted. All three cases demonstrated improved length of graft function after HTx has taken place, and suggests pre-operative irradiation may serve as standard pretreatment to improve HTx efficacy.

Also evident in the above case series, however, is the issue of recognizing and treating graft rejection. The case series did detail how rejection was diagnosed, however, there remains no consensus on pretreatment to reduce risk of rejection, graft monitoring, and treatment once rejection is recognized or suspected. This case series chose to utilize monitoring for CD154+ T-cytotoxic memory cells, previously demonstrated to be sensitive for acute rejection in pediatric liver or intestinal implants[53-56]. Increasing concentration of this T cell was noted to generally correlate with suspected decreased function of the hepatocytes, however, the authors do note that significant daily variance of measured total bilirubin in the CN TypeIpatients and phenylalanine levels in the PKU patient[52]. Reviewing the aforementioned cases and use of immunosuppression, it appears tacrolimus is an acceptable immunosuppressive agent, however, that closer monitoring of drug levels may be necessary to ensure continued appropriate function of the transplanted hepatocytes[1,31,52]. Further prospective cohort studies utilizing different monitoring intervals or alternate immunosuppressive therapy is likely necessary to ensure sustained and optimized graft function.

CONCLUSION

Further human studies involving humans are needed, however, the above collectively suggest progress in terms of improved effectiveness of HTx. With improvements in optimizing delivery technique and assessing proper recipients of livers, monitoring graft function, as well as recognizing and treating graft rejection, HTx may be able to be used more widely in metabolic liver disease and potentially delay necessity of OLT.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Transplantation

Country of origin: United States

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Kin T, Ramsay MA, Sureshkumar KK S- Editor: Ji FF L- Editor: A E- Editor: Yan JL

References
1.  Muraca M, Gerunda G, Neri D, Vilei MT, Granato A, Feltracco P, Meroni M, Giron G, Burlina AB. Hepatocyte transplantation as a treatment for glycogen storage disease type 1a. Lancet. 2002;359:317-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 271]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
2.  Hines RN, McCarver DG. The ontogeny of human drug-metabolizing enzymes: phase I oxidative enzymes. J Pharmacol Exp Ther. 2002;300:355-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 221]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
3.  Popescu I, Dima SO. Domino liver transplantation: how far can we push the paradigm? Liver Transpl. 2012;18:22-28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 53]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
4.  Roels L, Rahmel A. The European experience. Transpl Int. 2011;24:350-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 57]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
5.  Fox IJ, Chowdhury JR, Kaufman SS, Goertzen TC, Chowdhury NR, Warkentin PI, Dorko K, Sauter BV, Strom SC. Treatment of the Crigler-Najjar syndrome type I with hepatocyte transplantation. N Engl J Med. 1998;338:1422-1426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 826]  [Cited by in F6Publishing: 706]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
6.  Donato MT, Lahoz A, Jiménez N, Pérez G, Serralta A, Mir J, Castell JV, Gómez-Lechón MJ. Potential impact of steatosis on cytochrome P450 enzymes of human hepatocytes isolated from fatty liver grafts. Drug Metab Dispos. 2006;34:1556-1562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 106]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
7.  Sagias FG, Mitry RR, Hughes RD, Lehec SC, Patel AG, Rela M, Mieli-Vergani G, Heaton ND, Dhawan A. N-acetylcysteine improves the viability of human hepatocytes isolated from severely steatotic donor liver tissue. Cell Transplant. 2010;19:1487-1492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
8.  Strom SC, Fisher RA, Thompson MT, Sanyal AJ, Cole PE, Ham JM, Posner MP. Hepatocyte transplantation as a bridge to orthotopic liver transplantation in terminal liver failure. Transplantation. 1997;63:559-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 419]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
9.  Mitry RR, Dhawan A, Hughes RD, Bansal S, Lehec S, Terry C, Heaton ND, Karani JB, Mieli-Vergani G, Rela M. One liver, three recipients: segment IV from split-liver procedures as a source of hepatocytes for cell transplantation. Transplantation. 2004;77:1614-1616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 112]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
10.  Kawahara T, Toso C, Douglas DN, Nourbakhsh M, Lewis JT, Tyrrell DL, Lund GA, Churchill TA, Kneteman NM. Factors affecting hepatocyte isolation, engraftment, and replication in an in vivo model. Liver Transpl. 2010;16:974-982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
11.  Gramignoli R, Marongiu F, Tahan V, Dorko K, Geller D, Venkataramanan R, D’Antiga L, Ellis ECS, Strom SC. Isolation and evaluation of human hepatocytes for cell-based therapy. Dig Liver Dis. 2011;43:S96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
12.  Donato MT, Lahoz A, Montero S, Bonora A, Pareja E, Mir J, Castell JV, Gómez-Lechón MJ. Functional assessment of the quality of human hepatocyte preparations for cell transplantation. Cell Transplant. 2008;17:1211-1219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
13.  LeCluyse EL. Human hepatocyte culture systems for the in vitro evaluation of cytochrome P450 expression and regulation. Eur J Pharm Sci. 2001;13:343-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 340]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
14.  Jorns C, Ellis EC, Nowak G, Fischler B, Nemeth A, Strom SC, Ericzon BG. Hepatocyte transplantation for inherited metabolic diseases of the liver. J Intern Med. 2012;272:201-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 95]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
15.  Strauss KA, Robinson DL, Vreman HJ, Puffenberger EG, Hart G, Morton DH. Management of hyperbilirubinemia and prevention of kernicterus in 20 patients with Crigler-Najjar disease. Eur J Pediatr. 2006;165:306-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 101]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
16.  Groth CG, Arborgh B, Björkén C, Eriksson G, Lundren G. Hepatocellular transplantation in glucuronyltransferase-deficient rat. Sven Kir. 1974;31:42.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Groth CG, Arborgh B, Björkén C, Sundberg B, Lundgren G. Correction of hyperbilirubinemia in the glucuronyltransferase-deficient rat by intraportal hepatocyte transplantation. Transplant Proc. 1977;9:313-316.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Matas AJ, Sutherland DE, Steffes MW, Mauer SM, Sowe A, Simmons RL, Najarian JS. Hepatocellular transplantation for metabolic deficiencies: decrease of plasms bilirubin in Gunn rats. Science. 1976;192:892-894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 236]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
19.  Darwish AA, Sokal E, Stephenne X, Najimi M, de Goyet Jde V, Reding R. Permanent access to the portal system for cellular transplantation using an implantable port device. Liver Transpl. 2004;10:1213-1215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 67]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
20.  Dhawan A, Puppi J, Hughes RD, Mitry RR. Human hepatocyte transplantation: current experience and future challenges. Nat Rev Gastroenterol Hepatol. 2010;7:288-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 244]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
21.  Ambrosino G, Varotto S, Strom SC, Guariso G, Franchin E, Miotto D, Caenazzo L, Basso S, Carraro P, Valente ML. Isolated hepatocyte transplantation for Crigler-Najjar syndrome type 1. Cell Transplant. 2005;14:151-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 154]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
22.  Lysy PA, Najimi M, Stephenne X, Bourgois A, Smets F, Sokal EM. Liver cell transplantation for Crigler-Najjar syndrome type I: update and perspectives. World J Transplant. 2008;14:3464-3470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 91]  [Cited by in F6Publishing: 75]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
23.  Allen KJ, Mifsud NA, Williamson R, Bertolino P, Hardikar W. Cell-mediated rejection results in allograft loss after liver cell transplantation. Liver Transpl. 2008;14:688-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 64]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
24.  Meyburg J, Das AM, Hoerster F, Lindner M, Kriegbaum H, Engelmann G, Schmidt J, Ott M, Pettenazzo A, Luecke T. One liver for four children: first clinical series of liver cell transplantation for severe neonatal urea cycle defects. Transplantation. 2009;87:636-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 132]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
25.  Meyburg J, Hoffmann GF. Liver, liver cell and stem cell transplantation for the treatment of urea cycle defects. Mol Genet Metab. 2010;100 Suppl 1:S77-S83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
26.  Eguchi S, Rozga J, Lebow LT, Chen SC, Wang CC, Rosenthal R, Fogli L, Hewitt WR, Middleton Y, Demetriou AA. Treatment of hypercholesterolemia in the Watanabe rabbit using allogeneic hepatocellular transplantation under a regeneration stimulus. Transplantation. 1996;62:588-593.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  Gunsalus JR, Brady DA, Coulter SM, Gray BM, Edge AS. Reduction of serum cholesterol in Watanabe rabbits by xenogeneic hepatocellular transplantation. Nat Med. 1997;3:48-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 77]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
28.  Chowdhury JR, Grossman M, Gupta S, Chowdhury NR, Baker JR Jr, Wilson JM. Long-term improvement of hypercholesterolemia after ex vivo gene therapy in LDLR-deficient rabbits. Science. 1991;254:1802-1805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 293]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
29.  Grossman M, Rader DJ, Muller DW, Kolansky DM, Kozarsky K, Clark BJ 3rd, Stein EA, Lupien PJ, Brewer HB Jr, Raper SE. A pilot study of ex vivo gene therapy for homozygous familial hypercholesterolaemia. Nat Med. 1995;1:1148-1154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 392]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
30.  Chou JY, Jun HS, Mansfield BC. Glycogen storage disease type I and G6Pase-β deficiency: etiology and therapy. Nat Rev Endocrinol. 2010;6:676-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 157]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
31.  Lee KW, Lee JH, Shin SW, Kim SJ, Joh JW, Lee DH, Kim JW, Park HY, Lee SY, Lee HH. Hepatocyte transplantation for glycogen storage disease type Ib. Cell Transplant. 2007;16:629-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 81]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
32.  Steinberg SJ, Dodt G, Raymond GV, Braverman NE, Moser AB, Moser HW. Peroxisome biogenesis disorders. Biochim Biophys Acta. 2006;1763:1733-1748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 338]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
33.  Sokal EM, Smets F, Bourgois A, Van Maldergem L, Buts JP, Reding R, Bernard Otte J, Evrard V, Latinne D, Vincent MF. Hepatocyte transplantation in a 4-year-old girl with peroxisomal biogenesis disease: technique, safety, and metabolic follow-up. Transplantation. 2003;76:735-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 235]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
34.  Ohashi K, Waugh JM, Dake MD, Yokoyama T, Kuge H, Nakajima Y, Yamanouchi M, Naka H, Yoshioka A, Kay MA. Liver tissue engineering at extrahepatic sites in mice as a potential new therapy for genetic liver diseases. Hepatology. 2005;41:132-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 109]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
35.  Tatsumi K, Ohashi K, Shima M, Nakajima Y, Okano T, Yoshioka A. Therapeutic effects of hepatocyte transplantation on hemophilia B. Transplantation. 2008;86:167-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
36.  Dhawan A, Mitry RR, Hughes RD, Lehec S, Terry C, Bansal S, Arya R, Wade JJ, Verma A, Heaton ND. Hepatocyte transplantation for inherited factor VII deficiency. Transplantation. 2004;78:1812-1814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 189]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
37.  Arnell H, Fischler B, Bergdahl S, Schnell PO, Jacobsson H, Nemeth A. Hepatobiliary scintigraphy during cholestatic and noncholestatic periods in patients with progressive familial intrahepatic cholestasis after partial external biliary diversion. J Pediatr Surg. 2011;46:467-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
38.  De Vree JM, Ottenhoff R, Bosma PJ, Smith AJ, Aten J, Oude Elferink RP. Correction of liver disease by hepatocyte transplantation in a mouse model of progressive familial intrahepatic cholestasis. Gastroenterology. 2000;119:1720-1730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 146]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
39.  Loeber JG. Neonatal screening in Europe; the situation in 2004. J Inherit Metab Dis. 2007;30:430-438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 156]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
40.  Blau N, van Spronsen FJ, Levy HL. Phenylketonuria. Lancet. 2010;376:1417-1427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 704]  [Cited by in F6Publishing: 700]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
41.  Hamman KJ, Winn SR, Harding CO. Hepatocytes from wild-type or heterozygous donors are equally effective in achieving successful therapeutic liver repopulation in murine phenylketonuria (PKU). Mol Genet Metab. 2011;104:235-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
42.  Hansel MC, Davila JC, Vosough M, Gramignoli R, Skvorak KJ, Dorko K, Marongiu F, Blake W, Strom SC. The Use of Induced Pluripotent Stem Cells for the Study and Treatment of Liver Diseases. Curr Protoc Toxicol. 2016;67:14.13.1-14.13.27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 60]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
43.  Stéphenne X, Najimi M, Ngoc DK, Smets F, Hue L, Guigas B, Sokal EM. Cryopreservation of human hepatocytes alters the mitochondrial respiratory chain complex 1. Cell Transplant. 2007;16:409-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 76]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
44.  Gómez-Lechón MJ, Lahoz A, Jiménez N, Vicente Castell J, Donato MT. Cryopreservation of rat, dog and human hepatocytes: influence of preculture and cryoprotectants on recovery, cytochrome P450 activities and induction upon thawing. Xenobiotica. 2006;36:457-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 34]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
45.  Terry C, Hughes RD, Mitry RR, Lehec SC, Dhawan A. Cryopreservation-induced nonattachment of human hepatocytes: role of adhesion molecules. Cell Transplant. 2007;16:639-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
46.  Sharma S, Ellis EC, Gramignoli R, Dorko K, Tahan V, Hansel M, Mattison DR, Caritis SN, Hines RN, Venkataramanan R. Hepatobiliary disposition of 17-OHPC and taurocholate in fetal human hepatocytes: a comparison with adult human hepatocytes. Drug Metab Dispos. 2013;41:296-304.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Meyburg J, Alexandrova K, Barthold M, Kafert-Kasting S, Schneider AS, Attaran M, Hoerster F, Schmidt J, Hoffmann GF, Ott M. Liver cell transplantation: basic investigations for safe application in infants and small children. Cell Transplant. 2009;18:777-786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
48.  Baccarani U, Adani GL, Sanna A, Avellini C, Sainz-Barriga M, Lorenzin D, Montanaro D, Gasparini D, Risaliti A, Donini A. Portal vein thrombosis after intraportal hepatocytes transplantation in a liver transplant recipient. Transpl Int. 2005;18:750-754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
49.  Nagata H, Ito M, Shirota C, Edge A, McCowan TC, Fox IJ. Route of hepatocyte delivery affects hepatocyte engraftment in the spleen. Transplantation. 2003;76:732-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
50.  Habibullah CM, Syed IH, Qamar A, Taher-Uz Z. Human fetal hepatocyte transplantation in patients with fulminant hepatic failure. Transplantation. 1994;58:951-952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 223]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
51.  Pilichos C, Perrea D, Demonakou M, Preza A, Donta I. Management of carbon tetrachloride-induced acute liver injury in rats by syngeneic hepatocyte transplantation in spleen and peritoneal cavity. World J Transplant. 2004;10:2099-2102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
52.  Soltys KA, Setoyama K, Tafaleng EN, Soto Gutiérrez A, Fong J, Fukumitsu K, Nishikawa T, Nagaya M, Sada R, Haberman K. Host conditioning and rejection monitoring in hepatocyte transplantation in humans. J Hepatol. 2017;66:987-1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 76]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
53.  Zhou H, Dong X, Kabarriti R, Chen Y, Avsar Y, Wang X, Ding J, Liu L, Fox IJ, Roy-Chowdhury J. Single liver lobe repopulation with wildtype hepatocytes using regional hepatic irradiation cures jaundice in Gunn rats. PLoS One. 2012;7:e46775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
54.  Ashokkumar C, Bentlejewski C, Sun Q, Higgs BW, Snyder S, Mazariegos GV, Abu-Elmagd K, Zeevi A, Sindhi R. Allospecific CD154+ B cells associate with intestine allograft rejection in children. Transplantation. 2010;90:1226-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
55.  Ashokkumar C, Shapiro R, Tan H, Ningappa M, Elinoff B, Fedorek S, Sun Q, Higgs BW, Randhawa P, Humar A. Allospecific CD154+ T-cytotoxic memory cells identify recipients experiencing acute cellular rejection after renal transplantation. Transplantation. 2011;92:433-438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
56.  Ashokkumar C, Talukdar A, Sun Q, Higgs BW, Janosky J, Wilson P, Mazariegos G, Jaffe R, Demetris A, Dobberstein J. Allospecific CD154+ T cells associate with rejection risk after pediatric liver transplantation. Am J Transplant. 2009;9:179-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 66]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]