university of groningen technical aspects of liver transplantation

45
University of Groningen Technical aspects of liver transplantation Polak, Wojciech Grzegorz IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2008 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Polak, W. G. (2008). Technical aspects of liver transplantation. [s.n.]. Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license. More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne- amendment. Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 23-06-2022

Upload: khangminh22

Post on 12-Mar-2023

1 views

Category:

Documents


0 download

TRANSCRIPT

University of Groningen

Technical aspects of liver transplantationPolak, Wojciech Grzegorz

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2008

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):Polak, W. G. (2008). Technical aspects of liver transplantation. [s.n.].

CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license.More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne-amendment.

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

Download date: 23-06-2022

CHAPTER 2

THE EVOLUTION OF SURGICAL TECHNIQUES IN LIVER TRANSPLANTATION - A REVIEW

Polak WG, Peeters PMJG, Slooff MJH

Submitted for publication

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

12 | Chapter 2

ABSTRACT

Currently, liver transplantation (LT) is an accepted method of treatment of end-stage

liver disease, metabolic diseases with their primary defect in the liver and unresectable

primary liver tumors. Surgical techniques in liver transplantation have evolved

considerably over the past 40 years. The developments have led to a safer procedure for

the recipient reflected by continuously improving survival figures after LT. Also the new

techniques offer the possibility of tailoring the operation to the needs and condition

of the recipient as in partial grafting or in different revascularization techniques, or in

techniques of biliary reconstructions. Additionally the new techniques such as split LT,

domino transplantation and LRLT have also brought about an increase in the available

grafts. In this review the evolution of surgical techniques in LT over the past 40 years

and their contribution to the current results are discussed.

The evolution of surgical techniques in liver transplantation - a review | 13

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

INTRODUCTION

Currently, liver transplantation (LT) is an accepted method of treatment of end-stage

liver disease, metabolic diseases with their primary defect in the liver and unresectable

primary liver tumors. The first attempt of LT in humans was done by Thomas Starzl at

the University of Colorado in Denver, in 1963 1. The recipient was a three-years-old

boy with biliary atresia. The child died during transplantation, because of coagulopathy

and bleeding. After several unsuccessful LTs in Denver, Boston and Paris, a voluntary

worldwide moratorium discouraged liver transplantation until 1967 2-4. On July 23,

1967 Thomas Starzl performed the first successful LT. The recipient was 19-month-old

girl with hepatocellular carcinoma 5. One year later, Sir Roy Calne successfully launched

LT in Cambridge, UK 6. Despite the fact that at least the operation seemed to be feasible,

LT remained an experimental procedure until 1983 due to its low one year survival rate,

which at that time was around 30% 7,8. In that year a National Institute of Health

Consensus Development was held in Washington DC. Based on the experiences from

four liver transplant centers in Pittsburgh (US), Cambridge (UK), Hannover (Germany)

and Groningen (the Netherlands) a consensus document was published stating that LT

could be recognized as an accepted method of treatment for patients with end-stage

liver disease 9. By that time the first improved one year survival rate approaching 60%

were reported 10.

From the first successful LT until now many details of the surgical technique of LT have

been improved and adopted in order to achieve a superior outcome. Currently, one year

patient survival after LT is about 85-90% and long-term survival is about 70% 11,12.

In this review the evolution of surgical techniques in LT over the past 40 years and their

contribution to the current results are discussed.

IMPLANTATION TECHNIQUES

Conventional LT

In the early days of LT, the technique in which the native liver is removed together with

the retrohepatic part of the inferior vena cava (IVC) and orthotopically replaced by a

donor liver including the IVC with supra- and infrahepatic IVC anastomoses, was the

standard technique of LT 13. This technique is called the conventional technique and it

was exclusively used until the early 1980’s. The results of the procedure improved over

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

14 | Chapter 2

the years from the poor survival during the early days to 60% one year survival reported

in the early 1980’s 10. The conventional technique requires temporary cross-clamping

of the IVC and portal vein (PV) with important hemodynamic consequences as reduced

back-flow to the heart, congestion in the caval and splanchic bed as well as a reduction

of renal function. The congestion leads to increased bleeding during hepatectomy. To

solve this problems a veno-venous by-pass (VVB) was introduced in the mid 1980’s 14-

15.

Venous-venous by-pass

The first clinical attempts with a passive by-pass from the IVC to the superior vena cava

via the internal jugular vein were often complicated by fatal pulmonary embolism due

to clotting in the plastic cannulae 1. To avoid thrombotic complications Calne et al.

introduced a femoro-femoral partial cardiopulmonary bypass with a pump oxygenator.

This provided the wanted hemodynamic stability 16. However, the need for systemic

heparinisation often led to uncontrolled bleeding. In 1983 Griffith et al. introduced a

VVB with the use of centrifugal pump and heparin coated cannulae 14. Shaw described

the first LT with this type of VVB without the need of systemic heparinization 15.

As shown by several studies the benefits of the VVB include: (1) reduction of hemodynamic

instability in the anhepatic phase; (2) preservation of renal function; (3) reduction of

blood loss and (4) prevention of portal and systemic cogestion 15,17,18. The complication

rate of the VVB varies between 10-30% 19. The most common complications include

wound infection, hematoma, lymphocoele, deep vein thrombosis, and nerve injuries

and they are associated with the insertion of cannulae 19-21. Severe complications as

hypothermia, bleeding complications as a result of hemolysis and platelet depletion or

pulmonary embolism can be life-threatening 22-25. Additionally, the use of the VVB is

associated with higher costs.

Over time modifications of the standard VVB were introduced in order to simplify its use

and reduce complications. In the presence of porto-caval or mesenterico-caval shunts

single-limb bypass from the IVC was used 26. Also cannulation of the PV was replaced in

some centers by cannulation of the inferior mesenteric vein 27. A percutaneous technique

for establishing the VVB for femoral and axillary cannula was introduced in 1994 by

Oken et al. to reduce complications due to the dissection in the axilla and in the groin 28. A prospective randomized study showed that this percutaneous technique shortens

operative time, provides better shunt flow and is associated with a lower complication

rate in comparison to the open technique 29.

The evolution of surgical techniques in liver transplantation - a review | 15

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

Despite the advantages of the VVB several centers reported excellent results of LT

without the VVB and recommended to use it selectively 30-33. The development of an

alternative graft implantation technique technique with preservation of the IVC, the so

called piggyback technique ultimately made the VVB redundant.

“Piggyback” technique

In the piggyback technique in the contrast to the conventional technique the native IVC

remains in situ during hepatectomy. To achieve this, the native liver is dissected from

the IVC.

Although this technique was already used in the pioneering period of

clinical LT in Denver (US) and Cambridge (UK), it was popularized by Tzakis et al. in 1989 5,6,34.

In the original report of Tzakis piggyback LT was still performed with the use of a

VVB [34]. Venous outflow reconstruction was done between the suprahepatic end of

the donor IVC and a common orifice created of two or three hepatic veins (left and

middle hepatic vein or right and middle hepatic vein). This is the “classical” piggyback

technique 34.

Currently, the piggyback technique is the dominating one in LT and it is feasible in most

of the cases 35-37. Several studies, including one randomized trial compared the results

of the conventional technique and the piggyback technique. The results of these studies

showed shorter operation time, shorter warm ischemia time, shorter anhepatic phase,

reduction of blood loss and reduction of the cost of the procedure in favour of the

piggyback technique 38-42.

Modifications of the piggyback technique

The “Achilles heel” of the piggyback technique is the venous outflow reconstruction.

Many reports about outflow problems are published, showing a frequency of outflow

complications of 2.5-4.6% 43,44. This includes major graft congestion, caval stenosis or

thrombosis, acute and chronic Budd-Chiari syndrome, and torsion or stenosis of caval

anastomosis.

Belghitti et al. described a side-to-side anastomosis between donor and recipient IVC

without the routine use of VVB 45. In this modification both supra- and infrahepatic

end of the donor IVC is closed and the anastomosis is created between two new

incisions, one made on the recipient IVC and another on the donor IVC. Cherqui et al.

introduced another venous outflow reconstruction. He enlarged the common orifice of

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

16 | Chapter 2

three hepatic veins by the caudal incision on the anterior wall of the recipient IVC and

anastomosed it with the suprahepatic end of the donor IVC (end-to-side anastomosis) 46. This technique was further modified by closing all hepatic veins and creating an

anastomosis between a new incision on the anterior wall of recipient IVC and a V-shape

incision on the donor IVC 47,48.

In some particular situations leading to a short suprahepatic IVC cuff such as in domino

liver transplantation or in case of outflow complications after piggyback implantation

some authors advocated the use of the infrahepatic cavo-caval anastomosis 49,50.

All these modifications of venous outflow reconstruction were meant to improve the

venous outflow from the graft and to reduce outflow complications.

There are few studies comparing different techniques of venous outflow reconstruction

in piggyback LT 44,51,52. A French retrospective, multicenter study showed that the

“classical” piggyback technique was associated with a higher bleeding complication

from the caval anastomosis as well as with a higher incidence of Budd-Chiari syndrome

in comparison to the side-to-side or end-to-side cavocavostomy 44. In contrast, the

result of a prospective study from Belgium demonstrated that although the “classical”

piggyback technique was associated with a higher incidence of bleeding complications

compared to the side-to-side technique, all outflow complications occurred only in the

latter technique 51. In a recent study Cescon et al. compared three types of orifices used

for caval anastomosis in “classical” piggyback LT and he found that a cuff of left and

middle hepatic veins with a >1 cm transversal cavoplasty was associated with the lowest

rate of complications related to caval anastomosis (stenosis, thrombosis or kinking) 52.

Temporary portocaval shunt (TPCS)

As an alternative to the VVB authors recommended the use of a TPCS with the

piggyback technique in order to provide better hemodynamic stability and decompress

the splanchic area during the anhepatic phase 53,54. A prospective randomized trial

assessing the use of TPCS in piggy-back LT demonstrated improvement of hemodynamic

status, reduction of intraoperative transfusion requirements and preservation of renal

function 55. Molmenti et al. reported good results with a novel technique facilitating

TPCS when a direct shunt could not be created 56,57. Either a donor iliac vein or plastic

cannula was used to bridge the distance between portal vein and the IVC.

In contrast, several reports demonstrated that excellent results with piggyback LT are

feasible without a TPCS 41,47,58.

The TPCS probably has its own indication field in patients tending to hemodynamic

The evolution of surgical techniques in liver transplantation - a review | 17

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

instability, patients with a difficult hepatectomy (i.e. large caudate lobe) or patients with

few portosystemic collaterals.

Pre-reperfusion flushing and IVC venting

Liver grafts need to be flushed before reperfusion to prevent air emboli and to remove

toxic products from the graft after the preservation period. If too high concentration of

vasoactive substances and potassium enters the systematic circulation after reperfusion

it may cause a variety of hemodynamic changes ranging from arrhythmias, hypotension

to cardiac arrest, known as postreperfusion syndrome 59. This syndrome occurs in

approximately 30% of all transplant patients 59,60.

Flushing solutions

Ringer’s solution has been the most common flushing solution since the early years of

LT 3. However, experimental as well as clinical studies showed that rinsing with cold

Ringer’s solution causes endothelial cell swelling, cellular rounding, protrusion of cells

into the sinusoidal space, which can be deleterious for graft function 61,62. Therefore,

some centers replaced Ringer’s solution with other flushing solutions as saline, serum

albumin, plasmanate (serum substitute), Carolina Rinse Solution or autologous blood 60,63,64.

In a randomized prospective study Adam et al. investigated two flushing protocols,

one with Ringer’s lactate and another with human albumin 63. The results of this study

showed that the degree of hepatocellular injury was significantly lower in grafts flushed

with human albumin probably due to their protective effects as oxygen radicals scavenger

and by the maintenance of normal oncotic pressure. Bachmann et al. compared in a

prospective randomized trial three different flushing protocols using human albumin,

Carolina Rinse solution and blood 64. In this study Carolina Rinse Solution proved to

prevent reperfusion injury and improve graft function compared to human albumin and

blood, which is explained by protection of sinusoidal endothelial cells and decreased

activation of Kupfer cells. In all aforementioned studies the liver graft was flushed when

the caval anastomosis was made and subsequently it was reperfused. The liver is not

uniformly rinsed by this technique 65,66. Therefore, toxic products such as potassium

can still enter the systematic circulation after recirculation in too high concentration. To

prevent this some authors proposed additional flush with portal or arterial blood and

the use of vena cava venting.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

18 | Chapter 2

IVC venting

The technique of additional flushing with portal blood was already mentioned in the

late 1970s by Wall et al., however in his report only the first 100 ml of portal blood

reperfusing the graft was discarded 67. This technique was refreshed by Fukuzawa et al.,

who showed that additional flush with 500 ml of portal blood was associated with better

early graft function, better hemodynamic stability and smaller shifts in serum potassium

compared to Ringer’s solution alone 60. Additionally, Fukuzawa et al. proposed IVC

venting with a catheter placed through the infrahepatic anastomosis, which allowed

the external drainage of the effluent before the caval clamp was released. Another

study showed also that portal blood flush alone together with IVC venting prior to

reperfusion was sufficient and optimized graft function without risk of hyperkalemia 68. The advantage of IVC venting was shown in a prospective study by Brems et al. 69. He showed that graft flushing with crystalloid solution and afterwards portal blood

with IVC venting resulted in stable serum potassium and lower rate of postreperfusion

syndrome in comparison to the technique without IVC venting.

However, different results were obtained in two other studies. In a randomized study,

Millis et al. compared four different flushing techniques with a solution of Ringer’s

lactate and serum albumin: initial portal flush with or without IVC venting, and initial

arterial flush with or without IVC venting 70. In both groups with IVC venting additional

flush with 500 ml portal blood was performed. He did not find any differences in

postoperative graft function between the four groups, however the lowest incidence

of postreperfusion syndrome was observed in the group of portal flush without

IVC venting. Similarly, in a more recent retrospective study comparing two flushing

techniques, portal vein flush with Ringer’s without IVC venting and portal blood flush

with IVC venting, the authors demonstrated that the first technique was associated with

better hemodynamic recovery, higher lactate clearance and better early postoperative

graft function compared to the second one 71.

Based on the available data it seems that flushing alone provides inadequate cleaning

of the liver with increased possibility of an unwanted high potassium influx in the

systematic circulation. Addition of venting seems to be more effective with lower

chances of deleterious effects after reperfusion. The debate about which solution is

superior for flushing is still not definitely concluded.

The evolution of surgical techniques in liver transplantation - a review | 19

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

REVASCULARIZATION PROTOCOLS

During the first attempts of LT in humans the liver grafts were reperfused first via hepatic

artery 5. Nowadays, the most commonly used sequence of revascularization is initial

reperfusion via the PV (initial portal reperfusion – IPR) and subsequent reconstruction

of the hepatic artery. This order is based on the experience from liver surgery that

portal blood flow alone can provide adequate hepatocellular function 72. However, IPR

increases the risk of warm ischemic damage to the bile ducts, which depend solely on

arterial blood supply. Due to the increasing incidence of ischemic biliary complications

the order of revascularization protocols during LT became subject of investigation.

Sequential revascularization

Sequential revascularization techniques can start either in anterograde fashion via PV or

hepatic artery (HA), or in retrograde fashion via the inferior vena cava (IVC) (retrograde

reperfusion).

Several authors advocated the use of sequential revascularization with arterial blood first

(initial arterial reperfusion – IAR) 73-76. In a prospective randomized study, Durcef et al.

showed more stable hemodynamics, better graft reperfusion and lower peak value of

serum aspartate aminotranferase in patients with IAR compared to patients with IPR 73.

The result of a recent prospective, randomized study comparing hemodynamic profile

and tissue oxygenation during IPR and IAR showed in contrast to the previous study that

IPR offers better graft perfusion and metabolic function than IAR 74. Due to the larger

blood volume IPR causes an acute increase in pulmonary vascular load, which can be

detrimental in patients with pulmonary hypertension or cardiomyopathy. IAR, therefore

may be indicated in patients with poor pulmonary and cardiac reserve. In another

study, Noun et al. demonstrated that IAR provides better graft reperfusion and lower

requirements of blood transfusion as well as shorter postreperfusion fase in comparison

to IPR 75. However, no differences in postoperative aspartate aminotransferase, bile

flow and early vascular and biliary complications were observed between these two

revascularization protocols. Similar results were obtained by Sadler et al., who showed

no differences in the postoperative liver function, intraoperative blood products use and

30-day and 1-year mortality between IPR and IAR 76.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

20 | Chapter 2

Retrograde reperfusion

A new technique of retrograde reperfusion via the IVC followed by subsequent anterograde

reperfusion via PV has recently been proposed 77. In this technique the vascular clamp

on the IVC is removed immediately after completing the IVC anastomosis, allowing

retrograde reperfusion while the PV anastomosis is being constructed. The rationale for

this technique is that it shortens the warm ischemia time (WIT) and efficiently removes

perfusion fluid from the graft before anterograde blood flow is reestablished. The results

of a recent prospective, randomized clinical trial comparing retrograde revascularization

with simultaneous revascularization demonstrated significantly reduction of serum

transaminases and bilirubin levels in the first week posttransplantation as well as lower

incidence of primary non-function and initial poor function in the retrograde reperfused

grafts compared to simultaneously reperfused grafts 78. However, this novel technique

of reperfusion was associated with a higher incidence of ischemic-type biliary lesions

(ITBL).

Simultaneous revascularization

In the simultaneous revascularization the liver graft is reperfused simultaneously via

PV and HA. The motivation for this approach is the same as for the IAR: to reduce

the incidence of biliary complications 79. An additional advantage of simultaneous

reperfusion is that, in case of a problem with one of the two anastomoses, this can

be repaired without completely interrupting blood flow to the graft. The disadvantage

of simultaneous revascularization is prolongation of the WIT and the anhepatic phase,

which can be detrimental to postoperative graft function, survival and morbidity 80,81.

Two retrospective studies compared IPR with simultaneous reperfusion 79,82. No

differences were found in patient and graft survival rates and the incidence of primary non-

function between both techniques of reperfusion. However, simultaneously reperfused

grafts showed significantly lower incidence of biliary complications. In contrast to these

studies, the results of a recent retrospective study showed no differences in patient and

graft survival, morbidity rates, incidence and severity of acute rejection, recuperation

of liver function, or the incidence of ITBL, when comparing IPR versus simultaneous

reperfusion 83.

From the data published to date it is clear that further randomized studies are needed to

prove the exact value of each revascularization technique. So far IPR remains the main

technique of revascularization, except for specific conditions, where IAR or simultaneous

revascularization can be used. The role of retrograde revascularization remains to be

established.

The evolution of surgical techniques in liver transplantation - a review | 21

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

TECHNIQUES OF VASCULAR RECONSTRUCTIONS

Portal vein reconstruction

PV reconstruction is usually performed in end-to-end fashion. It is very important to

prevent stenosis in the portal anastomosis in order to avoid portal hypertension 84. To

achieve this goal different techniques can be used. Starzl et al. proposed a so called

“growth factor”, which allows portal anastomosis to distend to its maximal diameter

after releasing a clamp from the PV and estabilishing portal flow 85. To increase this

effect it is helpful to clamp the donor PV for a short period as described by Calleja et

al. 86. In cases with a size discrepancy between donor and recipient PV (pediatric LT,

partial grafts) or PV hypoplasia the reconstruction of the PV vein can be challenging and

requires special techniques. The same is true for portal vein thrombosis.

Size mismatch or hypoplasia of the native PV

The size mismatch between donor and recipient PV is frequently located at the smaller

recipient side. In such cases the donor PV can be anastomosed to a branch patch of

left and right recipient PV or even to the branch patch of recipient PV and coronary

vein 87. If this is not possible such as in hypoplastic PV, an incision on the ventral side

of the native PV over the full length of the narrowing enlarges the opening on the PV 88. This defect can be covered by a patch or if not too ong covered by the end-to-end

anastomosis.

If the donor PV is long enough, the narrow or hypoplastic recipient portal vein can

be resected and the anastomosis is made at the level of the confluence of splenic and

superior mesenteric vein 88. If the donor PV is too short an interposition graft fashioned

from the donor iliac vein can be used 88,89.

Portal vein thrombosis

PV thrombosis used to be an absolute contraindication for LT. However, several groups

started to perform eversion thrombectomy in case of PV thrombosis with a success

rate of 95% and one-year patient survival above 80% 90-92. If PV thrombosis is not

only restricted to the PV, but involves the portal confluence, there is necessary to use

a “jump-graft” with a donor iliac vein between recipient superior mesenteric vein and

donor PV 93,94. All recipients after thrombectomy of the PV as well as after a “jump-

graft” reconstruction require anticoagulation therapy to prevent recurrent thrombosis 95,96. However, the 5-year patient survival in patients with PV reconstruction for massive

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

22 | Chapter 2

PV thrombosis is still inferior compared to the patients without it 97 .

In case of massive portal system thrombosis special, so called salvage revascularization

techniques were introduced. Cavoportal hemitransposition was proposed by Tzakis et al.

in cases where no portal inflow could be achieved 98. In this technique the infrahepatic

part of the recipient IVC is ligated and the donor PV is anastomosed to the recipient

IVC 98,99.

Sheil et al. described another salvage technique; renoportal anastomosis 100. This

technique is preferably used in patients with preexistent patent distal splenorenal shunt

either spontaneous or after shunt surgery 101. A recent meta-analysis of cavoportal

hemitransposition and renoportal anastomosis showed a high postoperative complication

rate in this procedures and an actuarial patient survival of 74% 102.

Arterialization of the portal vein is the third technique used to provide adequate portal

blood inflow 103. In this technique, the graft receives blood inflow to the portal vein

from the aorta via an iliac conduit or from the common hepatic artery 104,105. The

experiences with this technique are still limited and modest results are reported 105.

The ultimate refugium in cases where no portal flow can be established is multivisceral

organ transplantation. However, so far one and five year patient survival of 65% and

49% are reported 106.

Portal hyperpefusion

A particular complication of partial LT is the “small-for-size-syndrome”. This syndrome

can range from mild hepatic dysfunction with hyperbilirubinemia to the graft failure

leading to retransplantation or death. The problem occurs when a too small for size

graft receives all native portal inflow. Increased portal flow results in the subsequent

reduction of hepatic arterial inflow by the so-called hepatic arterial buffer response,

which can increase the risk for hepatic artery thrombosis (HAT) and ITBL 107. The most

important measure to prevent this syndrome is adequate size matching based on volume

measurements 108. Surgically portal hyperperfusion can be reduced either by splenic

artery ligation combined with portal vein banding or by partial portosystemic shunting

as reported by the Ghent group 109.

The evolution of surgical techniques in liver transplantation - a review | 23

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

ARTERIAL RECONSTRUCTION

Standard arterial reconstruction

To avoid early HAT, which ultimately requires retransplantation, a patent arterial

anastomosis is mandatory. The preferred method for standard arterial reconstruction is

a direct anastomosis between donor and recipient HA in end-to-end fashion. Whenever

possible it should be done with Carrel patches on donor and recipient side, as arterial

anastomoses without patches was found to be associated with a significantly higher

incidence of HAT 110,111. In order to further decrease the incidence of HAT a branch-patch

technique was introduced 112. In this technique the arterial anastomosis is performed

between bifurcations of the donor and recipient artery (i.e. bifurcation of the proper

HA to the left and right HA or bifurcation of the common HA to the proper HA and the

gastroduodenal artery) 113,114.

Unsuitable recipient artery

In some cases the standard arterial anastomosis cannot be made because of unsuitable

recipient arteries due to stenosis, thrombosis, atherosclerosis, intimal dissection or other

pathology.

In order to overcome these problems alternative methods of arterial reconstruction

have been developed. One of these methods is the use of an arterial conduit from

the donor iliac artery directly to the aorta 115,116. The conduit can be anastomosed

either to the infrarenal or to supratruncal aorta 117-119. If the donor iliac artery is not

available some authors recommended use of saphenous vein, autologous radial artery

or donor superior mesenteric artery (SMA) as the conduit for arterial revascularization 120-122. The use of cryopreserved grafts as the conduit has been abandoned, because

of the high complication rate (stenosis, aneurysm formation) 123. Two studies indicated

that use of donor arterial conduits was associated with excellent long term outcomes 124,125. However, several other studies confirmed that independently of the site of

anastomosis, the use of conduits significantly increases the risk of HAT, especially in

the later posttransplant period 126-129. Therefore, other authors recommended direct

anastomosis between an aortic Carrel patch of the donor celiac axis to the recipient

supratruncal aorta or use the recipient’s splenic artery for arterial reconstruction with

the similar results as the standard anastomosis 119,130,131.

The arterial inflow from the recipient side may be compromised by celiac trunk

compression by the arcuate ligament. This can be diagnosed intraoperatively either

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

24 | Chapter 2

by pressure or flow measurements and it can be solved by cleavage of the ligament 132,133. When there is still low arterial inflow, an arterial conduit directly to the aorta is

recommended.

Size discrepancy and multiple arteries

Partial grafting and living donor liver transplantation (LDLT) brought new challenges for

arterial reconstruction with newer techniques and instruments.

In order to overcome size discrepancies between donor and recipient HA the Kyoto

group recommended several techniques for arterial reconstruction 134. In case of a small

caliber difference an undersized artery is cut obliquely or by fish-mouth method, whereas

in a case of a marked diameter difference a funnelization technique is preferred. In the

fish-mouth method two longitudinal contralateral (180° apart) incisions are made on the

undersized artery, whereas in the funnelization technique the caliber of the oversized

artery is reduced gradually with vascular sutures. Testa et al. adopted the fish-mouth

technique 135. The arteriotomy is performed on the posterior wall of donor artery and

on the arterior wall of the recipient artery (mirror incision). Then the back wall is sutured

first from the inside with the running suture followed by running suture on the anterior

wall, which prevents turnover of the anastomosed arteries. Okazaki et al. introduced

the double-needle microsuture technique to avoid the intimal dissection due to turning

over the microclamps on the anastomosed arteries 136. In this technique the back wall

of the anastomosis is suture first with the use of double needle microsuture allowing

the optimal adaptation of the intima. The double-needle technique is especially useful

for arteries in which the tunica intima is separated from tunica media.

In a substantial number of donor livers (24-32%) variant arterial anatomy is present 132,137,138. The presence of aberrant arterial anatomy was found to be a risk factor for

arterial injuries during liver procurement with subsequently increased risk for HAT after

LT 139.

If an accessory artery is present it usually has to be reconstructed on the back-table

prior to implantation. Depending of the length and size of the accessory HA it can

be reinserted to the stump of splenic artery or gastroduodenal artery, or in case of

accessory right HA from the SMA a “fold-over” technique can be used in which the

anastomosis between patches of SMA and celiac trunk is made with a subsequent

anastomosis of the distal end of splenic artery to the recipient artery 140.

The presence of multiple arteries posed another problem for arterial reconstruction in

LDLT. It is not always necessary to reconstruct all arteries, as sometimes the reconstruction

The evolution of surgical techniques in liver transplantation - a review | 25

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

of the dominant artery is sufficient when a pulsatile back-bleeding from the stumps of

other artery/arteries is confirmed. In the absence of such backflow all arteries need

to be reconstructed 141,142. Marcos et al. demonstrated in a recent report of arterial

reconstruction in adult right lobe LDLT excellent results with a novel technique using

an “Y” extension graft or a reversed extension bifurcated graft from the recipient

proper HA with its bifurcation 143. All aforementioned microsurgical techniques are

routinely performed with the use of either surgical loupes or a surgical microscope,

which substantially decreased the number of arterial complications 144,145. Although

meticulous arterial reconstruction and introduction of microsurgical techniques were

recommended by pioneers of LT, the development of partial liver grafts and especially

LDLT established a place for microsurgical techniques in LT 7.

BILIARY ANASTOMOSIS

The biliary anastomosis is called the “Achilles heel” of LT, because biliary and biliary

related complications are the most common source of morbidity and mortality after LT 146,147. Depending on the definition and diagnostics as well as type of the liver graft

the reported incidence of biliary complications varies from 7-38% 148-156.

During the first human LT a loop cholecystojejunostomy with ligation of the donor

common bile duct was used 5. Many different types of bilio-digestive anastomoses

such as a loop cholecystojejunostomy, Roux-en-Y cholecystojejunostomy and

cholecystoduodenostomy have been used over the years 7,8,157-159. All this techniques

were abandoned because of a high rate of biliary complications (obstructions and

leakage), which were mostly related to obstruction of the cystic duct, “the locus minoris

resistentiae” of those techniques.

Currently, two main techniques of biliary tract reconstruction are used:

choledochocholedochostomy (CDC) and Roux-en-Y hepatico-/choledochojejunostomy.

The choice of the technique depends mostly on the recipient’s bile duct status.

Duct-to-duct anastomosis

The first experiences with choledochocholedochostomy (CDC) were discouraging 7,158.

The CDC was associated with a high incidence of bile leak often due to ischemia of the

common bile duct. The role of the common bile duct vascularization was not realized

yet. However, after a pivotal report of Terblanche in 1983 clarifying the importance of

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

26 | Chapter 2

the vascularization of the common bile duct, and its relevance for bile duct anastomosis

the results of CDC substantially improved 147,156,160,161.

If the recipient’s bile duct is normal the CDC is preferable, as it creates physiological bile

flow to the duodenum with preservation of the sphincter of Oddi function, it allows

endoscopic diagnostics and interventions, and it avoids intestinal contamination of the

bile ducts 161,162. It can be done in end-to-end or side-to-side fashion The side-to-side

technique seems to offer greater technical feasibility and easier reconstruction in the

presence of major discrepancy between donor and recipient bile duct, whereas the end-

to-end technique doesn’t require long bile ducts for anastomosis and it reported to be

safer in case of large choledochal varices 151.

A prospective randomized trial comparing end-to-end versus side-to-side biliary showed

no differences in the outcome and the incidence of biliary complication proving that

both techniques are equally effective 163.

Another prospective randomized trial comparing interrupted and continuous suture in

CDC, showed no differences between both suture types with respect to the incidence of

leaks and strictures 164. A new technique of end-to-end CDC using vascular staples was

proposed recently by Thomas et al. 165. In this study tissue everting clips were compared

to a conventional handsewn technique showing no differences in biliary complications

between both techniques.

Hepaticojejunostomy/choledochojejunostomy

The only bilo-digestive anastomosis, which routinely found a place in LT was the Roux-

en-Y hepaticojejunostomy or choledochojejunostomy 147,156.

Both techniques are indicated when the recipient bile duct is abnormal due to the

underlying disease (primary sclerosing cholangitis, cystic fibrosis) or absent as in biliary

atresia. Also when the distance between donor and recipient bile duct is too large

one of these techniques should be used in order to avoid traction on the anastomosis.

Taking into account blood supply of the bile ducts, hepaticojejunostomy as a high

bilodigestive anastomosis provides better conditions for preventing biliary strictures

than choledochojejunostomy 166,167.

A randomized trial comparing CDC with hepaticojejunostomy is not sensible, because

both techniques have their own indication field. In retrospective series both techniques

demonstrated similar outcome, however increased bacterial and fungal colonization

of biliary tract, bile leakage and biliary strictures were more common when Roux-en-Y

hepatico-/choledochojejunostomy was used 146,156,168,169.

J

The evolution of surgical techniques in liver transplantation - a review | 27

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

Although choledochoduodenostomy has almost been abandoned as a technique of

biliary reconstruction, it can play a role as an alternative or a rescue technique on rare

occasions. Such as in patients with difficult access due to previous operations or patients

with a relatively short bowel due to previous resections or in patients with primary

sclerosing cholangitis 170.

Biliary drainage

The possible benefits of the use of biliary drainage include monitoring of bile production,

decompression of the biliary tract and easy radiological access. On the other hand, the

use of biliary drainage needs an additional opening in the recipient bile duct with a risk

of exit site leakage and complications after biliary drain removal [168]. Moreover, a biliary

drain is a foreign body and it may cause partial obstruction by dislodgement, it may

increase bile sludge and stone formation, as well as risk for cholangitis 171. It also needs

to be present for 6-12 weeks after transplantation often to the patients’ discomfort. To

avoid complications associated with the use of biliary drainage several authors proposed

some technical modifications 172-176. Sawyer et al. used a polypropelene drainage

catheter introduced through the stump of cystic duct instead of standard T-tube,

showing significant reduction of tube-related complications, but not the frequency of

other biliary complications 172. Internal stenting was used by other authors 173,174. Johnson et al. recommended use of double-J catheter as an internal stent as a safe

alternative to standard T-tube 173. Other centres proposed a modified technique of

T-tube removal with radiological placing of a temporary catheter at the time of T-drain

removal or using the T-tube itself as a drain under fluoroscopy guidance at the level of

the former entrance place in the common bile duct 175,176. The results of two prospective randomized studies comparing the use of a T-tube in CDC

did not show any benefits of the use of the T-tube 177,178. In a French multicenter study

patients with the CDC over the T-tube had a significantly higher biliary complications

rate (33.3% vs 15.5%) compared to the patients without the T-tube 177. This was

mostly due to T-tube related complications. The results of the second study showed

that the use of a T-tube was associated with a significantly higher number of septic

complications 178. In a prospective randomized trial Bawa et al. compared CDC with

and without using polyvinyl internal stent 174. He found that patient with an internal

biliary stent had a significantly higher bilary complications rate compared to patients

without biliary stenting. Additionally, the use of T-tube was associated with significantly

higher cost as it was shown first in a retrospective study and then confirmed by results

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

28 | Chapter 2

of randomized trial 179,180.

In summary the progress in biliary reconstruction was related to the recognition of the

importance of the arterial circulation of the bile ducts, which made a safe use of CDC

possible. Duct-to-duct anastomosis without routine use of biliary stenting is a preferable

technique of biliary reconstruction in LT. If it is not possible the hepaticojejunostomy is

an equivalent alternative.

PARTIAL LIVER TRANSPLANTATION

Extended waiting time and an unacceptable high mortality on the waiting list for LT in

children as a result of scarcity of pediatric donors were obvious reasons for searching

new techniques to obtain suitable grafts for this specific age group.

Reduced-size liver transplantation

The first reduced-size liver transplantation was performed in Denver in 1975, however

this case was not reported until description of a new technique for reducing adult donor

livers to a left lateral liver graft in 1984 by Bismuth in Paris and by Broelsch in Hannover 181-183. These techniques were rapidly implemented by several major pediatric transplant

centers 184,185.

The results of reduced-size LT were equal to the results of full-size LT and waiting time

and mortality on the waiting list for children were significantly reduced 186,187. As a

logical consequence of these favorable results of partial grafts and the fact that for that

purpose adult livers were withdrawn from the adult donor pool, the emergence of the

split liver technique became a fact 188.

Split liver transplantation

Split LT was first performed by Pichlmayr in Hannover and Bismuth in Paris in 1988 189,190. The technique of liver splitting allows the division of the adult donor liver

together with its vascular and biliary structures into two functional grafts, which can be

transplanted into two recipients. Usually grafts were split in a left or left lateral liver for

a child and a right lobe graft for an adult recipient.

The application of split LT was subject to important adaptations over the years. Two

important items emerged after the initial experiences 191-195. Firstly, applying the

technique in an emergency situation should be done with caution and secondly donor

The evolution of surgical techniques in liver transplantation - a review | 29

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

livers eligible for splitting should fulfill certain selection criteria as formulated by Ville de

Goyet 196. These criteria include: donors less that 45 years old, hemodynamically stable,

low or mild inotropic support, normal or slightly altered liver tests, less then five days in

the intensive care unit and normal macroscopic aspect at procurement. Only when such

conditions are met the results of split LT are equal to whole liver grafting 197,198.

A major debate in split LT concerned the subject of in situ or ex-situ splitting 199-201. In

the beginning the liver splitting was usually performed ex-situ, which means that the

whole liver was procured and subsequently divided into two grafts on the back-table 202. Rogiers et al. proposed a new technique, in which the liver is splitted during organ

procurement in the heart-beating donor 203. This technique was developed from left

lateral segmentectomy for living donation. The main advantage of the in-situ technique

is a shorter WIT, better hemostasis and lower rate of biliary complications 199,204.

However, this technique significantly prolongs the time of organ procurement and it

might cause hemodynamic instability during the donor procedure. Also procurement

teams may outstand their welcome in donor hospitals due to the prolonged operation

times. So far from the available literature there is no consensus which technique is

superior as both techniques demonstrate similar patient and graft survival compared

to whole liver grafting 200,201,204. In a retrospective study, Rayes et al. compared ex-

situ and in-situ splitting techniques with respect to patient and graft survival 205. He

found that the in-situ technique provided significantly higher one-year graft survival

in pediatric recipients compared to the ex-situ technique. However, this difference did

not translate in higher one-year patient survival in pediatric recipients. Moreover, there

were no differences in one-year patient and graft survival in adult recipients between

both techniques.

Nowadays split LT can be regarded as a routine procedure in experienced centers. The

application of this technique however is still developing.

Although liver splitting is used to be performed in adult donor livers Cescon et al. recently

reported also excellent results of split LT from pediatric donors below 40 kg 206.

Until 2005 only grafts from stable heart-beating donors were considered for splitting.

However, recently Muiesan et al. reported the first experiences with split and reduced-

size liver transplantation from non-heart beating donors 207.

Due to the increasing shortage of donor grafts especially for adults the technique of left/

right splitting of a graft for two adults re-emerged 208-211. The value of this technique is

currently under evaluation in experienced centers, as left lobe grafts in adult recipients

had inferior outcome in the past 212-214.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

30 | Chapter 2

Recently, left lateral graft procurement was further developed to create monosegmental

grafts and therefore facilitating LT in very small children 215-217.

Living donor liver transplantatation (LDLT)

Evolution of surgical techniques related to partial LT as well as the continuing shortage of

deceased donors led to the development of LDLT. Initially it was developed for pediatric

recipients as the mortality rate in children awaiting LT was high. The first attempts were

performed by Raia et al. in 1988 and 1989 218. The first successful LDLT was reported

in 1990 by Strong et al., who transplanted a left lateral segment from a mother to her

17-months-old son with biliary atresia 219. Since then many centers, especially in Japan

where liver grafts from deceased donors were not available, further developed LDLT 220. LDLT for pediatric recipients turned out to be a great success, as patient and graft

survival was comparable and in most centers superior to that in split and reduced size LT

and even in whole LT 12,221-223. At the same time it appeared a relatively safe procedure

for the donor, with a complications rate of 15-20% as reported in large series and a

mortality rate of between 0.1% and 0.4% 224-226.

The excellent results of LDLT in children together with increasing waiting time for adult

patients with malignant liver tumors prompted the use of living donor liver grafts also

for adults 227. First left lobe grafts were used 228. However, the volume of the left liver

lobe seemed not sufficient for adults and small-for-size syndrome appeared to be a

frequent complication as well as the results were not satisfactory 222,229. Nowadays left

lobes are only used for small adults or older adolescents, or they are used in a dual graft

setting 228,230,231. Therefore, right lobe grafting is the most applied technique for LDLT

in adults. The long-term results of LDLT for adults are nowadays approaching those

of whole LT. However, the rate of biliary complications is high 12,232-234. Additionally,

morbidity in living donors of right liver lobes is significantly higher than morbidity after

living donation for pediatric recipients 233-236.

The techniques of LT with living donor grafts are in essence the same as for implantation

of partial grafts from deceased donors. However, vascular and biliary reconstructions

are often more complex due to multiple vessels or ducts. Except for an adequate graft

size match in LDLT, good venous drainage of the anterior sector of the right liver lobe

was found to be extremely important for postoperative liver function 237. Many different

approaches have been adopted to avoid venous congestion of the anterior sector. These

include reconstruction of the hepatic veins of segment V and VIII into the recipient IVC

using an interposition vein graft or a routine inclusion of the middle hepatic vein in the

The evolution of surgical techniques in liver transplantation - a review | 31

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

graft 238-240. In LDLT the same techniques of biliary reconstructions can be used as in

LT from deceased donors. Roux-en-Y hepaticojejunostomy has been standard biliary

reconstruction in pediatric LDLT, however recently duct-to-duct reconstruction is used

more frequently, especially in adult LDLT 240-243. In case of multiple bile ducts if they

are close to each other they can be unified into a single orifice (ductoplasty) before

anastomosis 244-245.

OTHER LIVER TRANSPLANTATION TECHNIQUES

Domino liver transplantation (DLT)

The concept of DLT was first developed during the international workshop in liver

transplantation for familial amyloidosis polyneuropathy in Stockholm in 1993 246. Two

years later Furtado performed the first successful DLT in Portugal 247. This technique is

possible in patients with a primary genetic defect restricted to the liver with secondary

debilitating extrahepatic manifestations in vital organs such as kidneys, heart and brain.

In order to stop the progression of the disease the liver needs to be replaced 248. This

liver is suitable for donation to another patient with liver failure, because it takes one or

two decades, before the disease becomes symptomatic in a recipient 247-249. Therefore,

such domino livers are given to elderly patients or patients with a malignant disease in

the liver 250. The advantage for the domino recipients is that they can get a graft in

earlier course of their disease. Familial amyloid polyneuropathy is a classical example of

the disease, where domino LT can be used.

The results of DLT are rewarding. Furtado et al. reported one-year patients survival of

70% and 60% 3-year survival, however a recent report Yamamoto et al. showed 1-

year and 5-year patient survival of 95% and 92% in recipients transplanted in the last

decade 250-251 .

Domino donation evolved over the years. In the early years the liver was implanted with

the native IVC, while nowadays the domino liver is harvested without the IVC 252-254. This

requires reconstruction of the suprahepatic caval vein cuff for piggyback implantation 255,256. The advantage of leaving the native IVC in situ is the improved hemodynamic

stability in these already hemodynamic compromised patients. Azoulay et al. reported

also split LT from domino livers from patients with familial amyloid polyneuropathy 257.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

32 | Chapter 2

Auxiliary liver transplantation (ALT)

In the early days of LT the risks of the recipient hepatectomy were appreciated. This

caused an interest in ALT, in which the native liver remained in place and a new graft was

transplanted in a heterotopic position. This is the so called heterotopic auxiliary LT. The

first successful heterotopic auxiliary LT was reported by Fortner et al. in 1970, however

the long-term results were disappointing and inferior to orthotopic LT 258,259. The main

reasons for this were technical complications due to a high venous outflow pressure,

portal vein flow diversion as well as the high risks for the development of hepatocellular

carcinoma in the native liver. As showed in a recent case-control study comparing the

results ALT and orthotopic LT, ALT can not be an alternative for orthotopic LT for chronic

liver disease due to the inferior long-term outcome 260. Therefore, the technique of

heterotopic auxiliary LT has been abandoned for chronic liver diseases.

Terpstra et al. reintroduced the concept of heterotopic ALT for acute liver failure 261. It

based on the fact that after recuperation of the native liver the auxiliary graft could be

removed. This elegant concept was shown to be viable. However, it gained no support

in the transplant community, because of technical problems implied by portal vein

thrombosis and graft dysfunction, which led to the inferior patient survival compared

to orthotopic LT. To solve this problem in a recent report Ringers et al. described a

new technique of ALT in which dissection of the recipient hepatoduodenal ligament is

avoided 262. In this technique end-to-end anastomose was performed between donor

portal vein and recipient left renal vein, hepatic artery was anastomosed to the aorta

using an iliac arterial graft conduit and the bile duct was anastomosed to the stomach.

In 1991 Gubernatis et al. introduced a new technique of ALT for fulminant hepatic

failure, in which part of the native liver was removed and a partial liver graft was

implanted orthotopically (auxiliary partial orthotopic LT) 263. The technique of auxiliary

partial orthotopic LT tried to avoid the disadvantages of ALT while keeping the idea of a

temporary auxiliary graft. Several studies except for one from Japan reported favorable

outcome after auxiliary orthotopic LT for fulminant hepatic failure, however it was

associated with a higher postoperative morbidity 264-272. As shown in one study it

should be offered only for patients with high chances of liver regeneration 265.

Auxiliary orthotopic liver transplantation was also performed with good results for

metabolic liver disorders that cause life-threatening extrahepatic complications as for

example Crigler-Najjar syndrome type 1 273. The rationale for this treatment is to provide

sufficient liver mass to correct the metabolic disorder while keeping the majority of the

native liver.

The evolution of surgical techniques in liver transplantation - a review | 33

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

An interesting variant of auxiliary orthotopic LT was developed in Japan to overcome

small-for-size syndrome in LRLT 274. First, a left hemihepatectomy is performed in the

recipient with subsequent implantation of the left lobe from the living donor. The right

part of the native liver is kept in place until sufficient graft regeneration. Subsequently

the remaining native liver is removed during a second-stage operation.

Although conceptually promising, the ALT plays a minimal role in the treatment of

patients with acute or chronic liver failure mostly because of the high complications

rate, its complexity as well as the fact that orthotopic LT remains a gold standard of the

treatment.

SUMMARY

Surgical techniques in liver transplantation have evolved considerably over the past

40 years. The developments have led to a safer procedure for the recipient reflected

by continuously improving survival figures after LT. Also the new techniques offer the

possibility of tailoring the operation to the needs and condition of the recipient as in

partial grafting or in different revascularization techniques, or in techniques of biliary

reconstructions. Additionally the new techniques such as split LT, domino transplantation

and LRLT have also brought about an increase in the available grafts.

REFERENCES

1. Starzl TE, Marchioro TL, Von Kaulla KN, Hermann G, Brittain RS, Waddell WR.

Homotransplantation of the liver in humans. Surg Gynecol Obstet 1963;117:659-76.

2. Moore FD, Birtch AG, Dagher F, Veith F, Krisher JA, Order SE et al. Immunosuppression and

vascular insufficiency in liver transplantation. Ann N Y Acad Sci 1964;120:729-38.

3. Demirleau J, Noureddine M, Vignes C, Prawerman A, Reziciner S, Larraud P et al.Tentative

d’homogreffe hepatique. [Attempted hepatic homograft.] Mem Acad Chir (Paris)

1964;90:177-9.

4. Starzl TE, Marchioro TL, Rowlands DT Jr, Kirkpatrick CH, Wilson WE, Rifkind D et al.

Immunosuppression after experimental and clinical homotransplantation of the liver. Ann

Surg 1964;160:411-39.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

34 | Chapter 2

5. Starzl TE, Groth CG, Brettschneider L, Penn I, Fulginiti VA, Moon JB et al. Orthotopic

homotransplantation of the human liver. Ann Surg 1968;168:392-415.

6. Calne RY, Williams R. Liver transplantation in man. I. Observations on technique and

organization in five cases. Br Med J 1968; 4:535-40.

7. Starzl TE, Porter KA, Putnam CW, Schroter GP, Halgrimson CG, Weil R 3rd et al. Orthotopic

liver transplantation in ninety-three patients. Surg Gynecol Obstet 1976;142:487-505.

8. Calne RY, Williams R. Orthotopic liver transplantation: the first 60 patients. Br Med J 1977;

1:471-6.

9. Liver transplantation. National Institutes of Health Consensus Development. Natl Inst Health

Consens Dev Conf Summ 1983; 4:15 p.

10. Krom RA, Gips CH, Houthoff HJ, Newton D, van der Waaij D, Beelen J et al. Orthotopic liver

transplantation in Groningen, The Netherlands (1979-1983). Hepatology 1984;4:61S-65S.

11. 2005 Annual Report of the U.S. Organ Procurement and Transplantation Network and

the Scientific Registry of Transplant Recipients: Transplant Data 1995-2006. Department

of Health and Human Services, Health Resources and Services Administration, Healthcare

Systems Bureau, Division of Transplantation, Rockville, MD; United Network for Organ

Sharing, Richmond, VA; Arbor Research Collaborative for Health (formerly URREA), Ann

Arbor, MI.

12. European Liver Transplant Registry. Data Analysis Booklet 05/1968-12/2003.

13. Starzl TE. Putnam CW. Experience in hepatic transplantation. 1969, WB Saunders Company:

Philadelphia. pp 131-135.

14. Griffith BP, Shaw BW Jr, Hardesty RL, Iwatsuki S, Bahnson HT, Starzl TE. Veno-venous bypass

without systemic anticoagulation for transplantation of the human liver. Surg Gynecol

Obstet 1985; 160:270-2.

15. Shaw BW Jr, Martin DJ, Marquez JM, Kang YG, Bugbee AC Jr, Iwatsuki S et al. Venous

bypass in clinical liver transplantation. Ann Surg 1984;200:524-34.

16. Calne RY, Smith DP, McMaster P, Craddock GN, Rolles K, Farman JV et al. Use of partial

cardiopulmonary bypass during the anhepatic phase of orthotopic liver grafting. Lancet

1979;2:612-4.

17. Cheema SP, Hughes A, Webster NR, Bellamy MC. Cardiac function during orthotopic liver

transplantation with venovenous bypass. Anaesthesia 1995;50:776-8.

18. Estrin JA, Belani KG, Ascher NL, Lura D, Payne W, Najarian JS. Hemodynamic changes on

clamping and unclamping of major vessels during liver transplantation. Transplant Proc

1989; 21:3500-5.

The evolution of surgical techniques in liver transplantation - a review | 35

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

19. Chari RS, Gan TJ, Robertson KM, Bass K, Camargo CA, Greig PD et al. Venovenous bypass in

adult orthotopic liver transplantation: routine or selective use? J Am Coll Surg 1998;186:683-

90.

20. Budd JM, Isaac JL, Bennett J, Freeman JW. Morbidity and mortality associated with large-

bore percutaneous venovenous bypass cannulation for 312 orthotopic liver transplantations.

Liver Transpl 2001;7:359-62.

21. Katirji MB. Brachial plexus injury following liver transplantation. Neurology 1989;39:736-8.

22. Eleborg L, Sallander S, Tollemar J. Minimal hemolytic effect of veno-venous bypass during

liver transplantation. Transpl Int 1991;4:157-60.

23. Navalgund AA, Kang Y, Sarner JB, Jahr JS, Gieraerts R. Massive pulmonary thromboembolism

during liver transplantation. Anesth Analg 1988; 67:400-2.

24. Khoury GF, Kaufmann RD, Musich JA. Hypothermia related to the use of venovenous bypass

during liver transplantation. Eur J Anesthesiol 1990;7:501-503.

25. Khoury GF, Mann ME, Porot MJ, Abdul-Rasool IH, Busuttil RW. Air embolism associated with

veno-venous bypass during orthotopic liver transplantation. Anesthesiology 1987;67:848-

51.

26. Stegall MD, Mandell S, Karrer F, Kam I. Liver transplantation without venovenous bypass.

Transplant Proc 1995;27:1254-5.

27. Slooff MJ, Bams JL, Sluiter WJ, Klompmaker IJ, Hesselink EJ, Verwer R. A modified cannulation

technique for veno-venous bypass during orthotopic liver transplantation. Transplant Proc

1989;21: 2328-9.

28. Oken AC, Frank SM, Merritt WT, Fair J, Klein A, Burdick J et al. A new percutaneous technique

for establishing venous bypass access in orthotopic liver transplantation. J Cardiothorac

Vasc Anesth 1994;8:58-60.

29. Tisone G, Mercadante E, Dauri M, Colella D, Anselmo A, Romagnoli J et al. Surgical versus

percutaneous technique for veno-venous bypass during orthotopic liver transplantation: a

prospective randomized study. Transplant Proc 1999;31:3162-3.

30. Wall WJ, Grant DR, Duff JH, Kutt JL, Ghent CN, Bloch MS. Liver transplantation without

venous bypass. Transplantation 1987;43:56-61.

31. Grande L, Rimola A, Cugat E, Alvarez L, Garcia-Valdecasas JC, Taura P, Beltran J et al.. Effect

of venovenous bypass on perioperative renal function in liver transplantation: results of a

randomized, controlled trial. Hepatology 1996;23:1418-28.

32. Johnson MW, Powelson JA, Auchincloss H Jr, Delmonico FL, Cosimi AB. Selective use of

veno-venous bypass in orthotopic liver transplantation. Clin Transplant 1996;10:181-5.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

36 | Chapter 2

33. Kuo PC, Alfrey EJ, Garcia G, Haddow G, Dafoe DC. Orthotopic liver transplantation with

selective use of venovenous bypass. Am J Surg 1995;170:671-5.

34. Tzakis A, Todo S, Starzl TE. Orthotopic liver transplantation with preservation of the inferior

vena cava. Ann Surg 1989:210:649-652.

35. Fleitas MG, Casanova D, Martino E, Maestre JM, Herrera L, Hernanz F et al. Could the

piggyback operation in liver transplantation be routinely used? Arch Surg 1994;129:842-5.

36. Belghiti J, Ettorre GM, Durand F, Sommacale D, Sauvanet A, Jerius JT et al. Feasibility and

limits of caval-flow preservation during liver transplantation. Liver Transpl 2001; 7:983-7.

37. Lerut J, Ciccarelli O, Roggen F, Laterre PF, Danse E, Goffette P et al. Cavocaval adult liver

transplantation and retransplantation without veno-venous bypass and without portocaval

shunting: a prospective feasibility study in adult liver transplantation. Transplantation

2003,75:1740-5.

38. Gonzalez FX, Garcia-Valdecasas JC, Grande L, Pacheco JL, Cugat E, Fuster J et al.. Vena cava

vascular reconstruction during orthotopic liver transplantation: a comparative study. Liver

Transpl Surg 1998:4: 33.

39. Hosein Shokouh-Amiri M, Osama Gaber A, Bagous WA, Grewal HP, Hathaway DK, Vera SR

et al. Choice of surgical technique influences perioperative outcomes in liver transplantation

Ann Surg 2002:231:814-823.

40. Jovine E, Mazziotti A, Grazi GL, Ercolani G, Masetti M, Morganti M et al. Piggy-back versus

conventional technique in liver transplantation: report of a randomized trial. Transpl Int

1997:10:109-112.

41. Lerut JP, Molle G, Donataccio M, De Kock M, Ciccarelli O, et al. Cavocaval liver transplantation

without venovenous bypass and without temporary portocaval shunting: the ideal technique

for adult liver grafting? Transpl Int 1997:10:171-179.

42. Miyamoto S, Polak WG, Geuken E, Peeters PM, de Jong KP, Porte RJ et al. Liver transplantation

with preservation of the inferior vena cava. A comparison of conventional and piggyback

techniques in adults. Clin Transplant 2004;18:686-693.

43. Parrilla P, Sanchez-Bueno F, Figueras J, Jaurrieta E, Mir J, Margarit C et al. Analysis of

the complications of the piggy-back technique in 1,112 liver transplants. Transplantation

1999;67:1214-1217.

44. Navarro F, Le Moine MC, Fabre JM, Belghiti J, Cherqui D, Adam R et al. Specific vascular

complications of orthotopic liver transplantation with preservation of the retrohepatic vena

cava: review of 1361 cases. Transplantation 1999;68:646-650.

45. Belghiti J, Panis Y, Sauvanet A, Gayet B, Fekete F. A new technique of side to side caval

anastomosis during orthotopic hepatic transplantation without inferior vena caval occlusion.

Surg Gynecol Obstet 1992:175:270-272.

The evolution of surgical techniques in liver transplantation - a review | 37

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

46. Cherqui D, Lauzet JY, Rotman N, Duvoux C, Dhumeaux D, Julien M et al. Orthotopic liver

transplantation with preservation of the caval and portal flows. Technique and results in 62

cases. Transplantation 1994:58:793-796.

47. Polak WG, Nemes BA, Miyamoto S, Peeters PM, de Jong KP, Porte RJ, Slooff MJ. End-to-side

caval anastomosis in adult piggyback liver transplantation. Clin Transplant 2006;20:609-

16.

48. Dasgupta D, Sharpe J, Prasad KR, Asthana S, Toogood GJ, Pollard SG et al. Triangular and

self-triangulating cavocavostomy for orthotopic liver transplantation without posterior

suture lines: a modified surgical technique. Transpl Int 2006;19:117-21.

49. Merenda R, Gerunda GE, Neri D, Barbazza F, Di Marzio E, Bruttocao A et al. Infrahepatic

terminolateral cavo-cavostomy as a rescue technique in complicated “modified” piggyback

liver transplantation. J Am Coll Surg 1997;185:576-9.

50. Nishida S, Pinna A, Verzaro R, Levi D, Kato T, Nery JR et al. Domino liver transplantation with

end-to-side infrahepatic vena cavocavostomy. J Am Coll Surg 2001;192:237-40.

51. Hesse UJ, Berrevoet F, Torisi R, Pattyn P, Mortier E, Decruyenaere J et al. Hepato-venous

reconstruction in orthotopic liver transplantation with preservation of the recipients’ inferior

vena cava and veno-venous bypass. Langenbeck’s Arch Surg 2000;385:350-356.

52. Cescon M, Grazi GL, Varotti G, Ravaioli M, Ercolani G, Gardini A et al. Venous outflow

reconstructions with the piggyback technique in liver transplantation: a single-center

experience of 431 cases. Transpl Int 2005;18:318-325.

53. Tzakis AG, Reyes J, Nour B, Marino IR, Todo S, Starzl TE. Temporary end to side portacaval

shunt in orthotopic hepatic transplantation in humans. Surg Gynecol Obstet 1993:176:180-

182.

54. Belghiti J, Noun R, Sauvanet A. Temporary portocaval anastomosis with preservation of

caval flow during orthotopic liver transplantation. Am J Surg 1995;169:277-279.

55. Figueras J, Llado L, Ramos E, Jaurrieta E, Rafecas A, Fabregat J et al. Temporary portocaval

shunt during liver transplantation with vena cava preservation. Results of a prospective

randomized study. Liver Transpl 2001;7:904-911.

56. Molmenti EP, Marsh JW, Molmenti H, Reyes J, Fung JJ. Modified temporary end-to-side

portocaval shunt in liver and small bowel transplantation. Pediatr Transplant 2001;5:381-2.

57. Molmenti EP, Marsh JW, Fung JJ, Casavilla FA. Rapid temporary portosystemic bypass. Dig

Dis Sci 2002;47:448-9.

58. Muscari F, Suc B, Aguirre J, Di Mauro GL, Bloom E, Duffas JP et al. Orthotopic liver

transplantation with vena cava preservation in cirrhotic patients: is systematic temporary

portacaval anastomosis a justified procedure? Transplant Proc 2005;37:2159-62.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

38 | Chapter 2

59. Aggarwal S, Kang Y, Freeman JA, Fortunato FL, Pinsky MR. Postreperfusion syndrome:

cardiovascular collapse following hepatic reperfusion during liver transplantation. Transplant

Proc 1987;19(4 Suppl 3):54-5.

60. Fukuzawa K, Schwartz ME, Acarli K, Katz E, Gabrielson G, Gettes M et al. Flushing with

autologous blood improves intraoperative hemodynamic stability and early graft function

in clinical hepatic transplantation. J Am Coll Surg 1994;178:541-7.

61. Gao WS, Takei Y, Marzi I, Lindert KA, Caldwell-Kenkel JC, Currin RT et al. Carolina rinse

solution--a new strategy to increase survival time after orthotopic liver transplantation in

the rat. Transplantation 1991;52:417-24.

62. Bachmann S, Caldwell-Kenkel JC, Currin RT, Tanaka Y, Takei Y, Marzi I et al. Ultrastructural

correlates of liver graft failure from storage injury: studies of graft protection by Carolina

rinse solution and pentoxifylline. Transplant Proc 1993;25:1620-4.

63. Adam R, Astarcioglu I, Castaing D, Bismuth H. Ringer’s lactate vs serum albumin as a flush

solution for UW preserved liver grafts: results of a prospective randomized study. Transplant

Proc 1991;23:2374-5.

64. Bachmann S, Bechstein WO, Keck H, Lemmens HP, Brandes N, John AK et al. Pilot study:

Carolina Rinse Solution improves graft function after orthotopic liver transplantation in

humans. Transplant Proc 1997;29:390-2.

65. Paulsen AW, Valek TR, Ramsay M, Swygert T, Klintmalm G. Determination of an adequate

flush volume for removal of preservation fluid prior to revascularization of the donor liver.

Transplant Proc 1989; 21: 2349-50.

66. Fukuzawa K, Schwartz ME, Emre S, Miller CM. The effect of allograft flushing with portal

blood on the reperfusion syndrome and graft function in clinical liver transplantation.

Hepatology 1991;14:52A.

67. Wall WJ, Calne RY, Herbertson BM, Baker PG, Smith DP, Underwood J et al. Simple

hypothermic preservation for transporting human livers long distances for transplantation.

Report of 12 cases. Transplantation 1977;23:210-6.

68. Emre S, Schwartz ME, Mor E, Kishikawa K, Yagmur O, Thiese N et al. Obviation of

prereperfusion rinsing and decrease in reservation/reperfusion injury in liver transplantation

by portal blood flushing. Transplantation 1994;57:799-803.

69. Brems JJ, Takiff H, McHutchison J, Collins D, Biermann LA, Pockros P. Systemic versus

nonsystemic reperfusion of the transplanted liver. Transplantation 1993;55:527-9.

70. Millis JM, Melinek J, Csete M, Imagawa DK, Olthoff KM, Neelankanta G et al. Randomized

controlled trial to evaluate flush and reperfusion techniques in liver transplantation.

Transplantation 1997;63:397-403.

The evolution of surgical techniques in liver transplantation - a review | 39

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

71. Gruttadauria S, Cintorino D, Musumeci A, Arcadipane A, Burgio G, Clarizia S et al. Comparison

of two different techniques of reperfusion in adult orthotopic liver transplantation. Clin

Transplant 2006;20:159-62.

72. Rosenberg PM, Friedman LS. The liver in circulatory failure. In: Schiff ER, Sorrel MF, Maddrey

WS (eds). Schiff’s diseases of the liver. Lippincott Williams & Wilkins Publishers; 2002:1327.

73. Ducerf C, Mechet I, Landry JL, DeLaRoche E, Berthoux N, Bourdeix O et al. Hemodynamic

profiles during piggyback liver grafts using arterial or portal revascularization. J Am Coll

Surg 2000;190:89-93.

74. Moreno C, Sabaté A, Figueras J, Camprubí I, Dalmau A, Fabregat J et l. Hemodynamic

profile and tissular oxygenation in orthotopic liver transplantation: Influence of hepatic

artery or portal vein revascularization of the graft. Liver Transpl 2006;12:1607-14.

75. Noun R, Sauvanet A, Belghiti J. Appraisal of the order of revascularization in human liver

grafting: a controlled study. J Am Coll Surg 1997;185:70-3.

76. Sadler KM, Walsh TS, Garden OJ, Lee A. Comparison of hepatic artery and portal vein

reperfusion during orthotopic liver transplantation. Transplantation 2001;72:1680-1684.

77. Kniepeiss D, Iberer F, Grasser B, Schaffellner S, Stadlbauer V, Tscheliessnigg KH. A

single-center experience with retrograde reperfusion in liver transplantation. Transpl Int

2003;16:730-5.

78. Heidenhain C, Heise M, Jonas S, Ben-Asseur M, Puhl G, Mittler J et al. P. Retrograde

reperfusion via vena cava lowers the risk of initial nonfunction but increases the risk of

ischemic-type biliary lesions in liver transplantation--a randomized clinical trial. Transpl Int

2006;19:738-48.

79. Sankary HN, McChesney L, Frye E, Cohn S, Foster P, Williams J. A simple modification

in operative technique can reduce the incidence of nonanastomotic biliary strictures after

orthotopic liver transplantation. Hepatology 1995;21:63-9.

80. Piratvisuth T, Tredger JM, Hayllar KA, Williams R. Contribution of true cold and rewarming

ischemia times to factors determining outcome after orthotopic liver transplantation. Liver

Transpl Surg 1995;1:296-301.

81. Platz KP, Mueller AR, Schafer C, Jahns S, Guckelberger O, Neuhaus P. Influence of warm

ischemia time on initial graft function in human liver transplantation. Transplant Proc

1997;29:3458-9.

82. Massarollo PC, Mies S, Raia S. Simultaneous arterial and portal revascularization in liver

transplantation. Transplant Proc 1998;30:2883-4.

83. Polak WG, Miyamoto S, Nemes BA, Peeters PM, de Jong KP, Porte RJ et al. Sequential

and simultaneous revascularization in adult orthotopic piggyback liver transplantation. Liver

Transpl 2005: 11: 934-940.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

40 | Chapter 2

84. Scantlebury VP, Zajko AB, Esquivel CO, Marino IR, Starzl TE. Successful reconstruction of

late portal vein stenosis after hepatic transplantation. Arch Surg 1989;124:503-5.

85. Starzl TE, Iwatsuki S, Shaw BW Jr. A growth factor in fine vascular anastomoses. Surg

Gynecol Obstet 1984;159:164-5.

86. Calleja IJ, Polo JR, Garci a-Sabrido JL, Ferreiroa JP, Valdecantos E. Two-clamp method to

avoid portal anastomotic stenosis in liver transplantation. Am J Surg 1993; 165: 367-8.

87. Tanaka K, Uemoto S, Tokunaga Y, Fujita S, Sano K, Nishizawa T et al. Surgical techniques

and innovations in living related liver transplantation. Ann Surg 1993;217: 82-91.

88. Marwan IK, Fawzy AT, Egawa H, Inomata Y, Uemoto S, Asonuma K et al. Innovative

techniques for and results of portal vein reconstruction in living-related liver transplantation.

Surgery 1999;125: 265-70.

89. Mitchell A, John PR, Mayer DA, Mirza DF, Buckels JA, De Ville De Goyet J. Improved

technique of portal vein reconstruction in pediatric liver transplant recipients with portal

vein hypoplasia. Transplantation 2002;73:1244-7.

90. Molmenti EP, Roodhouse TW, Molmenti H, Jaiswal K, Jung G, Marubashi S et al.

Thrombendvenectomy for organized portal vein thrombosis at the time of liver

transplantation. Ann Surg 2002;235:292-6.

91. Dumortier J, Czyglik O, Poncet G, Blanchet MC, Boucaud C, Henry L et al. Eversion

thrombectomy for portal vein thrombosis during liver transplantation. Am J Transplant

2002;2:934-8.

92. Robles R, Fernandez JA, Hernandez Q, Mara n C, Rami rez P, Sanchez-Bueno F et al. Eversion

thromboendovenectomy in organized portal vein thrombosis during liver transplantation.

Clin Transplant 2004;18:79-84.

93. Shaked A, Busuttil RW. Liver transplantation in patients with portal vein thrombosis and

central portacaval shunts. Ann Surg 1991;214:696-702.

94. Lerut JP, Mazza D, van Leeuw V, Laterre PF, Donataccio M, de Ville de Goyet J, Van Beers B,

Bourlier P, Goffette P, Puttemans T, Otte JB. Adult liver transplantation and abnormalities of

splanchnic veins: experience in 53 patients. Transpl Int 1997;10:125-32.

95. Tzakis A, Todo S, Stieber A, Starzl TE. Venous jump grafts for liver transplantation in patients

with portal vein thrombosis. Transplantation 1989; 48: 530-1.

96. Moreno Gonzalez E, Garci a Garci a I, Gomez Sanz R, Gonzalez-Pinto I, Loinaz Segurola C,

Jimenez Romero C. Liver transplantation in patients with thrombosis of the portal, splenic

or superior mesenteric vein. Br J Surg. 1993;80:81-5.

97. Yerdel MA, Gunson B, Mirza D, Karayalcin K, Olliff S, Buckels J et al. Portal vein thrombosis in

adults undergoing liver transplantation: risk factors, screening, management, and outcome.

Transplantation 2000;69:1873-81.

The evolution of surgical techniques in liver transplantation - a review | 41

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

98. Tzakis AG, Kirkegaard P, Pinna AD, Jovine E, Misiakos EP, Maziotti A et al.. Liver transplantation

with cavoportal hemitransposition in the presence of diffuse portal vein thrombosis.

Transplantation 1998;65:619-24.

99. Selvaggi G, Weppler D, Nishida S, Moon J, Levi D, Kato T et al. Ten-year experience in porto-

caval hemitransposition for liver transplantation in the presence of portal vein thrombosis.

Am J Transplant 2007;7:454-60.

100. Sheil AG, Stephen MS, Chui AK, Ling J, Bookallil MJ. A liver transplantation technique in a

patient with a thrombosed portal vein and a functioning renal-lieno shunt. Clin Transplant

1997;11: 71-3.

101. Kato T, Levi DM, DeFaria W, Nishida S, Tzakis AG. Liver transplantation with renoportal

anastomosis after distal splenorenal shunt. Arch Surg 2000; 135: 1401-4.

102. Paskonis M, Jurgaitis J, Mehrabi A, Kashfi A, Fonouni H, Strupas K et al. Surgical strategies

for liver transplantation in the case of portal vein thrombosis--current role of cavoportal

hemitransposition and renoportal anastomosis. Clin Transplant 2006;20:551-62.

103. Troisi R, Kerremans I, Mortier E, Defreyne L, Hesse UJ, de Hemptinne B. Arterialization of the

portal vein in pediatric liver transplantation. A report of two cases. Transpl Int 1998;11:147-

51.

104. Charco R, Margarit C, Lopez-Talavera JC, Hidalgo E, Castells L, Allende H et al. Outcome

and hepatic hemodynamics in liver transplant patients with portal vein arterialization. Am J

Transplant 2001;1:146-51.

105. Settmacher U, Stange B, Schaser KD, Puhl G, Glanemann M, Steinmuller T et al. Primary

permanent arterialization of the portal vein in liver transplantation. Transpl Int 2003;16:430-

3.

106. Tzakis AG, Kato T, Levi DM, Defaria W, Selvaggi G, Weppler D et al. 100 multivisceral

transplants at a single center. Ann Surg 2005;242:480-90.

107. Bolognesi M, Sacerdoti D, Bombonato G, Merkel C, Sartori G, Merenda R et al. Change in

portal flow after liver transplantation: effect on hepatic arterial resistance indices and role

of spleen size. Hepatology 2002;35:601-8.

108. Kiuchi T, Tanaka K, Ito T, Oike F, Ogura Y, Fujimoto Y et al. Small-for-size graft in living donor

liver transplantation: how far should we go? Liver Transpl 2003;9:S29-35.

109. Troisi R, de Hemptinne B. Clinical relevance of adapting portal vein flow in living donor liver

transplantation in adult patients. Liver Transpl 2003;9:S36-41.

110. Merion RM, Burtch GD, Ham JM, Turcotte JG, Campbell DA. The hepatic artery in liver

transplantation. Transplantation 1989;48:438-43.

111. Quinones-Baldrich WJ, Memsic L, Ramming K, Hiatt J, Busuttil RW. Branch patch for

arterialization of hepatic grafts. Surg Gynecol Obstet 1986;162:488-90.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

42 | Chapter 2

112. Kalayoglu M, Belzer FO. A new technique for arterialization of the hepatic graft. Surg

Gynecol Obstet 1987;164:564-7.

113. Busuttil RW, Colonna JO 2nd, Hiatt JR, Brems JJ, el Khoury G, Goldstein LI et al. The first 100

liver transplants at UCLA. Ann Surg 1987;206:387-402.

114. Meneu-Diaz JC, Moreno-Gonzalez E, Garcia Garcia I, Jimenez Romero C, Loinaz Segurola

C, Gomez Sanz R et al. Hepatic allograft arterialization by means of the gastroduodenal

bifurcation (branch patch) as a prognostic factor. Transplantation 2004;77:1513-7.

115. Starzl TE, Halgrimson CG, Koep LJ, Weil R 3rd, Taylor PD. Vascular homografts from cadaveric

organ donors. Surg Gynecol Obstet. 1979;149:737.

116. Shaw BW Jr, Iwatsuki S, Starzl TE. Alternative methods of arterialization of the hepatic graft.

Surg Gynecol Obstet 1984;159:490-3.

117. Goldstein RM, Secrest CL, Klintmalm GB, Husberg BS. Problematic vascular reconstruction

in liver transplantation. Part I. Arterial. Surgery 1990;107:540-3.

118. Stewart MT, Millikan WJ Jr, Henderson JM, Galloway JR, Dodson TF. Proximal abdominal

graft for arterialization during hepatic transplantation. Surg Gynecol Obstet 1989;169:261-

2.

119. Shaked AA, Takiff H, Busuttil RW. The use of the supraceliac aorta for hepatic arterial

revascularization in transplantation of the liver. Surg Gynecol Obstet 1991;173:198-202.

120. Garcia-Valdecasas JC, Grande L, Rimola A, Fuster J, Lacy A, Visa J. The use of the saphenous

vein for arterial reconstruction in orthotopic liver transplant. Transplant Proc 1990;22:2376-

7.

121. Rogers J, Chavin KD, Kratz JM, Mohamed HK, Lin A, Baillie GM et al. Use of autologous

radial artery for revascularization of hepatic artery thrombosis after orthotopic liver

transplantation: case report and review of indications and options for urgent hepatic artery

reconstruction. Liver Transpl 2001;7:913-7.

122. Muiesan P, Rela M, Heaton ND. Use of cadaveric superior mesenteric artery as interpositional

vascular graft in orthotopic liver transplantation. Br J Surg 2001;88:70-2.

123. Kuang AA, Renz JF, Ferrell LD, Ring EJ, Rosenthal P, Lim RC et al. Failure patterns of

cryopreserved vein grafts in liver transplantation. Transplantation. 1996;62:742-7.

124. Cappadonna CR, Johnson LB, Lu AD, Kuo PC. Outcome of extra-anatomic vascular

reconstruction in orthotopic liver transplantation. Am J Surg 2001;182:147-50

125. Zamboni F, Franchello A, Ricchiuti A, Fop F, Rizzetto M, Salizzoni M. Use of arterial conduit as

an alternative technique in arterial revascularization during orthotopic liver transplantation.

Dig Liver Dis 2002;34:122-6.

The evolution of surgical techniques in liver transplantation - a review | 43

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

126. Muralidharan V, Imber C, Leelaudomlipi S, Gunson BK, Buckels JA, Mirza DF et al. Arterial

conduits for hepatic artery revascularisation in adult liver transplantation. Transpl Int

2004;7:163-8.

127. Del Gaudio M, Grazi GL, Ercolani G, Ravaioli M, Varotti G, Cescon M et al. Outcome of

hepatic artery reconstruction in liver transplantation with an iliac arterial interposition graft.

Clin Transplant 2005;19:399-405.

128. Stange BJ, Glanemann M, Nuessler NC, Settmacher U, Steinmuller T, Neuhaus P. Hepatic

artery thrombosis after adult liver transplantation. Liver Transpl 2003;9:612-20.

129. Silva MA, Jambulingam PS, Gunson BK, Mayer D, Buckels JA, Mirza DF et al. Hepatic artery

thrombosis following orthotopic liver transplantation: a 10-year experience from a single

centre in the United Kingdom. Liver Transpl 2006;12:146-51.

130. Cherqui D, Riff Y, Rotman N, Julien M, Fagniez PL. The recipient splenic artery for

arterialization in orthotopic liver transplantation. Am J Surg. 1994; 167: 327-30.

131. Figueras J, Pares D, Aranda H, Rafecas A, Fabregat J, Torras J et al. Results of using the

recipient’s splenic artery for arterial reconstruction in liver transplantation in 23 patients.

Transplantation 1997;64:655-8.

132. Settmacher U, Haase R, Heise M, Bechstein WO, Neuhaus P. Variations of surgical

reconstruction in liver transplantation depending on vasculature. Langenbecks Arch Surg

1999;384: 378-83.

133. Fukuzawa K, Schwartz ME, Katz E, Mor E, Emre S, Acarli K et al. The arcuate ligament

syndrome in liver transplantation. Transplantation 1993;56:223-4.

134. Inomoto T, Nishizawa F, Sasaki H, Terajima H, Shirakata Y, Miyamoto S et al. Experiences

of 120 microsurgical reconstructions of hepatic artery in living related liver transplantation.

Surgery 1996;119:20-6.

135. Testa G, Losanoff JE, Gangemi A, Benedetti E. Excellent outcome using an alternative

technique for arterial reconstruction in living-related liver transplant: sphenoid anastomosis.

Transpl Int 2007; 20:392-4.

136. Okazaki M, Asato H, Takushima A, Nakatsuka T, Sarukawa S, Inoue K, Harii K, Sugawara Y,

Makuuchi M. Hepatic artery reconstruction with double-needle microsuture in living-donor

liver transplantation. Liver Transpl 2006; 12: 46-50.

137. Hiatt JR, Gabbay J, Busuttil RW. Surgical anatomy of the hepatic arteries in 1000 cases. Ann

Surg 1994;220:50-2.

138. Gruttadauria S, Foglieni CS, Doria C, Luca A, Lauro A, Marino IR. The hepatic artery in liver

transplantation and surgery: vascular anomalies in 701 cases. Clin Transplant 2001;15:359-

63.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

44 | Chapter 2

139. Nijkamp DM, Slooff MJ, van der Hilst CS, Ijtsma AJ, de Jong KP, Peeters PM et al. Surgical

injuries of postmortem donor livers: incidence and impact on outcome after adult liver

transplantation. Liver Transpl 2006;12:1365-70.

140. Gordon RD, Shaw BW Jr, Iwatsuki S, Todo S, Starzl TE. A simplified technique for

revascularization of homografts of the liver with a variant right hepatic artery from the

superior mesenteric artery. Surg Gynecol Obstet 1985;160:474-6.

141. Ikegami T, Kawasaki S, Matsunami H, Hashikura Y, Nakazawa Y, Miyagawa S et al. Should

all hepatic arterial branches be reconstructed in living-related liver transplantation? Surgery

1996;119:431-6.

142. Takatsuki M, Chiang YC, Lin TS, Wang CC, Concejero A, Lin CC et al. Anatomical and

technical aspects of hepatic artery reconstruction in living donor liver transplantation.

Surgery 2006;140:824-8.

143. Marcos A, Killackey M, Orloff MS, Mieles L, Bozorgzadeh A, Tan HP. Hepatic arterial

reconstruction in 95 adult right lobe living donor liver transplants: evolution of anastomotic

technique. Liver Transpl 2003;9:570-4.

144. Mori K, Nagata I, Yamagata S, Sasaki H, Nishizawa F, Takada Y et al. The introduction of

microvascular surgery to hepatic artery reconstruction in living-donor liver transplantation-

-its surgical advantages compared with conventional procedures. Transplantation

1992;54:263-8.

145. Shackleton CR, Goss JA, Swenson K, Colquhoun SD, Seu P, Kinkhabwala MM et al. The impact

of microsurgical hepatic arterial reconstruction on the outcome of liver transplantation for

congenital biliary atresia. Am J Surg 1997; 173: 431-5.

146. Stratta RJ, Wood RP, Langnas AN, Hollins RR, Bruder KJ, Donovan JP et al. Diagnosis and

treatment of biliary tract complications after orthotopic liver transplantation. Surgery

1989;106:675-83.

147. Greif F, Bronsther OL, Van Thiel DH, Casavilla A, Iwatsuki S, Tzakis A et al. The incidence,

timing, and management of biliary tract complications after orthotopic liver transplantation.

Ann Surg 1994;219:40-5.

148. Lerut JP, Gordon RD, Iwatsuki S, Starzl TE. Human orthotopic liver transplantation: surgical

aspects in 393 consecutive grafts. Transplant Proc 1988;20(1 Suppl 1):603-6.

149. Klein AS, Savader S, Burdick JF, Fair J, Mitchell M, Colombani P et al. Reduction of morbidity

and mortality from biliary complications after liver transplantation. Hepatology 1991;

14:818-23.

150. Heffron TG, Emond JC, Whitington PF, Thistlethwaite JR Jr, Stevens L, Piper J et al. Biliary

complications in pediatric liver transplantation. A comparison of reduced-size and whole

grafts. Transplantation 1992;53:391-5.

The evolution of surgical techniques in liver transplantation - a review | 45

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

151. Neuhaus P, Blumhardt G, Bechstein WO, Steffen R, Platz KP, Keck H. Technique and results

of biliary reconstruction using side-to-side choledochocholedochostomy in 300 orthotopic

liver transplants. Ann Surg 1994;219:426-34.

152. Chardot C, Candinas D, Mirza D, Gunson B, Davison S, Murphy MS et al. Biliary complications

after paediatric liver transplantation: Birmingham’s experience. Transpl Int 1995;8:133-40.

153. Cronin DC 2nd, Alonso EM, Piper JB, Newell KA, Bruce DS, Woodle ES et al. Biliary

complications in living donor liver transplantation. Transplant Proc 1997;29:419-20.

154. Egawa H, Uemoto S, Inomata Y, Shapiro AM, Asonuma K, Kiuchi T et al. Biliary complications

in pediatric living related liver transplantation. Surgery 1998;124:901-10.

155. Buis CI, Verdonk RC, Van der Jagt EJ, van der Hilst CS, Slooff MJ, Haagsma EB et al.

Nonanastomotic biliary strictures after liver transplantation, part 1: Radiological features

and risk factors for early vs. late presentation. Liver Transpl 2007;13:708-18.

156. Colonna JO 2nd, Shaked A, Gomes AS, Colquhoun SD, Jurim O, McDiarmid SV et al. Biliary

strictures complicating liver transplantation. Incidence, pathogenesis, management, and

outcome. Ann Surg 1992;216:344-50.

157. Calne RY. A new technique for biliary drainage in orthotopic liver transplantation utilizing

the gall bladder as a pedicle graft conduit between the donor and recipient common bile

ducts. Ann Surg 1976;184:605-9.

158. Calne RY, McMaster P, Portmann B, Wall WJ, Williams R. Observations on preservation, bile

drainage and rejection in 64 human orthotopic liver allografts. Ann Surg 1977;186:282-

90.

159. Halff G, Todo S, Hall R, Starzl TE. Late complications with gallbladder conduit biliary

reconstruction after liver transplantation. Transplantation 1989;48:537-9.

160. Terblanche J, Allison HF, Northover JM. An ischemic basis for biliary strictures. Surgery 1983;

94:52-7.

161. Krom RA, Kingma LM, Haagsma EB, Wesenhagen H, Slooff MJ, Gips CH.

Choledochocholedochostomy, a relatively safe procedure in orthotopic liver transplantation.

Surgery 1985;97:552-6.

162. Rabkin JM, Orloff SL, Reed MH, Wheeler LJ, Corless CL, Benner KG et al. Biliary tract

complications of side-to-side without T tube versus end-to-end with or without T tube

choledochocholedochostomy in liver transplant recipients. Transplantation 1998;65:193-9.

163. Davidson BR, Rai R, Kurzawinski TR, Selves L, Farouk M, Dooley JS et al. Prospective

randomized trial of end-to-end versus side-to-side biliary reconstruction after orthotopic

liver transplantation. Br J Surg 1999;86:447-52.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

46 | Chapter 2

164. Castaldo ET, Pinson CW, Feurer ID, Wright JK, Gorden DL, Kelly BS et al. Continuous versus

interrupted suture for end-to-end biliary anastomosis during liver transplantation gives

equal results. Liver Transpl 2007;13:234-8.

165. Thomas KT, Gorden DL, Chari RS, Wright JK, Feurer ID, Pinson CW. Biliary reconstruction

using non-penetrating, tissue everting clips versus conventional sewn biliary anastomosis in

liver transplantation. HPB 2006;8:132-136.

166. Northover J, Terblanche J. Bile duct blood supply. Its importance in human liver

transplantation. Transplantation 1978;26:67-9.

167. Terblanche J, Worthley CS, Spence RA, Krige JE. High or low hepaticojejunostomy for bile

duct strictures? Surgery 1990;108:828-34.

168. O’Connor TP, Lewis WD, Jenkins RL. Biliary tract complications after liver transplantation.

Arch Surg 1995;130:312-7.

169. Sossenheimer M, Slivka A, Carr-Locke D. Management of extrahepatic biliary disease after

orthotopic liver transplantation: review of the literature and results of a multicenter survey.

Endoscopy 1996;28:565-71.

170. Schmitz V, Neumann UP, Puhl G, Tran ZV, Neuhaus P, Langrehr JM. Surgical complications

and long-term outcome of different biliary reconstructions in liver transplantation for

primary sclerosing cholangitis-choledochoduodenostomy versus choledochojejunostomy.

Am J Transplant 2006;6:379-85.

171. Ben-Ari Z, Neville L, Davidson B, Rolles K, Burroughs AK. Infection rates with and without

T-tube splintage of common bile duct anastomosis in liver transplantation. Transpl Int

1998;11:123-6.

172. Sawyer RG, Punch JD. Incidence and management of biliary complications after 291 liver

transplants following the introduction of transcystic stenting. Transplantation 1998;66:1201-

7.

173. Johnson MW, Thompson P, Meehan A, Odell P, Salm MJ, Gerber DA et al. Internal biliary

stenting in orthotopic liver transplantation. Liver Transpl 2000;6:356-61.

174. Bawa SM, Mathew A, Krishnan H, Minford E, Talbot D, Mirza DF et al. Biliary reconstruction

with or without an internal biliary stent in orthotopic liver transplantation: a prospective

randomised trial. Transpl Int 1998;11 Suppl 1: S245-7.

175. Goodwin SC, Bittner CA, Patel MC, Noronha MA, Chao K, Sayre JW. Technique for

reduction of bile peritonitis after T-tube removal in liver transplant patients. J Vasc Interv

Radiol 1998;9:986-90.

176. Urbani L, Campatelli A, Romagnoli J, Catalano G, Sartoni G, Costa A et al. T-tube removal

after liver transplantation: a new technique that reduces biliary complications. Transplantation

2002;74:410-3.

The evolution of surgical techniques in liver transplantation - a review | 47

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

177. Scatton O, Meunier B, Cherqui D, Boillot O, Sauvanet A, Boudjema K et al. Randomized trial

of choledochocholedochostomy with or without a T tube in orthotopic liver transplantation.

Ann Surg 2001;233:432-7.

178. Vougas V, Rela M, Gane E, Muiesan P, Melendez HV, Williams R et al. A prospective

randomised trial of bile duct reconstruction at liver transplantation: T tube or no T tube?

Transpl Int 1996;9:392-5.

179. Shimoda M, Saab S, Morrisey M, Ghobrial RM, Farmer DG, Chen P et al. A cost-effectiveness

analysis of biliary anastomosis with or without T-tube after orthotopic liver transplantation.

Am J Transplant 2001;1:157-61.

180. Amador A, Charco R, Marti J, Navasa M, Rimola A, Calatayud D et al. Clinical trial on

the cost-effectiveness of T-tube use in an established deceased donor liver transplantation

program. Clin Transplant 2007;21:548-53.

181. Starzl Te. Demetris AJ: Liver transplantation: A 31 year perspective. Chicago, Year Book,

1990: 38-41.

182. Bismuth H, Houssin D. Reduced-sized orthotopic liver graft in hepatic transplantation in

children. Surgery 1984;95:367-70.

183. Broelsch CE, Neuhaus P, Burdelski M, et al. Orthotope transplantation von lebersegmenten

bei kleinkindern mit gallengangsatresien. Orthotopic transplantation of hepatic segments

in infants with biliary atresia. In Koslowski L, ed. Chirurgisches Forun ‘84 F. Experim U.

Klimische Forschung Hrsg. Berlin/Heidelberg: Springer, 1984.

184. Otte JB, de Ville de Goyet J, Sokal E, Alberti D, Moulin D, de Hemptinne B et al. Size

reduction of the donor liver is a safe way to alleviate the shortage of size-matched organs

in pediatric liver transplantation. Ann Surg 1990;211:146-57.

185. Emond JC, Whitington PF, Thistlethwaite JR, Alonso EM, Broelsch CE. Reduced-size

orthotopic liver transplantation: use in the management of children with chronic liver

disease. Hepatology 1989;10:867-72.

186. de Ville de Goyet J, Hausleithner V, Reding R, Lerut J, Janssen M, Otte JB. Impact of innovative

techniques on the waiting list and results in pediatric liver transplantation. Transplantation

1993;56:1130-6.

187. Langnas AN, Marujo WC, Inagaki M, Stratta RJ, Wood RP, Shaw BW Jr. The results of

reduced-size liver transplantation, including split livers, in patients with end-stage liver

disease. Transplantation 1992;53:387-91.

188. Slooff MJ. Reduced size liver transplantation, split liver transplantation, and living related

liver transplantation in relation to the donor organ shortage. Transpl Int 1995;8:65-8.

189. Pichlmayr R, Ringe B, Gubernatis G, Hauss J, Bunzendahl H. Transplantation of a donor

liver to 2 recipients (splitting transplantation)- a new method in the further development of

segmental liver transplantation. Langenbecks Arch Chir 1988;373:127-30.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

48 | Chapter 2

190. Bismuth H, Morino M, Castaing D, Gillon MC, Descorps Declere A, Saliba F et al. Emergency

orthotopic liver transplantation in two patients using one donor liver. Br J Surg 1989;76:722-

4.

191. Broelsch CE, Emond JC, Whitington PF, Thistlethwaite JR, Baker AL, Lichtor JL. Application

of reduced-size liver transplants as split grafts, auxiliary orthotopic grafts, and living related

segmental transplants. Ann Surg 1990;212:368-75.

192. Azoulay D, Astarcioglu I, Bismuth H, Castaing D, Majno P, Adam R et al. Split-liver

transplantation. The Paul Brousse policy. Ann Surg 1996;224:737-46.

193. Kalayoglu M, D’Alessandro AM, Knechtle SJ, Hoffmann RM, Pirsch JD, Judd RH et al.

Preliminary experience with split liver transplantation. J Am Coll Surg 1996;182:381-7.

194. Rela M, Vougas V, Muiesan P, Vilca-Melendez H, Smyrniotis V, Gibbs P et al. Split liver

transplantation: King’s College Hospital experience. Ann Surg 1998;227:282-8.

195. Sieders E, Peeters PM, TenVergert EM, Bijleveld CM, de Jong KP, Zwaveling JH et al. Analysis

of survival and morbidity after pediatric liver transplantation with full-size and technical-

variant grafts. Transplantation 1999;68:540-5.

196. de Ville de Goyet J : Technique for ex situ cadaveric liver division. In : Rogiers X, Bismuth H,

Busuttil RW, Broering DC, Azoulay D. eds. Split Liver Transplantation. Darmstadt : Springer;

2002:75-87.

197. Wilms C, Walter J, Kaptein M, Mueller L, Lenk C, Sterneck M et al.. Long-term outcome of

split liver transplantation using right extended grafts in adulthood: A matched pair analysis.

Ann Surg 2006;244:865-72.

198. Cardillo M, De Fazio N, Pedotti P, De Feo T, Fassati LR, Mazzaferro V et al. Split and whole

liver transplantation outcomes: a comparative cohort study. Liver Transpl 2006;12:402-10.

199. Rogiers X, Malago M, Gawad K, Jauch KW, Olausson M, Knoefel WT et al. In situ splitting

of cadaveric livers. The ultimate expansion of a limited donor pool. Ann Surg 1996;224:331-

9.

200. Noujaim HM, Gunson B, Mayer DA, Mirza DF, Buckels JA, Candinas D et al. Worth continuing

doing ex situ liver graft splitting? A single-center analysis. Am J Transplant 2003;3:318-23.

201. Goss JA, Yersiz H, Shackleton CR, Seu P, Smith CV, Markowitz JS et al. In situ splitting of the

cadaveric liver for transplantation. Transplantation 1997;64:871-7.

202. Otte JB, de Ville de Goyet J, Alberti D, Balladur P, de Hemptinne B. The concept and

technique of the split liver in clinical transplantation. Surgery 1990;107:605-12.

203. Rogiers X, Malago M, Habib N, Knoefel WT, Pothmann W, Burdelski M et al. In situ splitting

of the liver in the heart-beating cadaveric organ donor for transplantation in two recipients.

Transplantation 1995;59:1081-3.

The evolution of surgical techniques in liver transplantation - a review | 49

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

204. Yersiz H, Renz JF, Farmer DG, Hisatake GM, McDiarmid SV, Busuttil RW. One hundred in situ

split-liver transplantations: a single-center experience. Ann Surg 2003;238:496-505.

205. Reyes J, Gerber D, Mazariegos GV, Casavilla A, Sindhi R, Bueno J et al. Split-liver

transplantation: a comparison of ex vivo and in situ techniques. J Pediatr Surg 2000;35:283-

9.

206. Cescon M, Spada M, Colledan M, Torre G, Andorno E, Valente U et al. Feasibility and limits

of split liver transplantation from pediatric donors: an italian multicenter experience. Ann

Surg 2006;244:805-14.

207. Muiesan P, Jassem W, Girlanda R, Steinberg R, Vilca-Melendez H, Mieli-Vergani G et al.

Segmental liver transplantation from non-heart beating donors--an early experience with

implications for the future. Am J Transplant 2006;6:1012-6.

208. Azoulay D, Castaing D, Adam R, Mimoz O, Bismuth H. Transplantation of three adult

patients with one cadaveric graft: wait or innovate. Liver Transpl 2000;6:239-40.

209. Sommacale D, Farges O, Ettorre GM, Lebigot P, Sauvanet A, Marty J et al. In situ split liver

transplantation for two adult recipients. Transplantation 2000;69:1005-7.

210. Humar A, Ramcharan T, Sielaff TD, Kandaswamy R, Gruessner RW, Lake JR et al. Split liver

transplantation for two adult recipients: an initial experience. Am J Transplant 2001;1:366-

72.

211. Kilic M, Seu P, Stribling RJ, Ghalib R, Goss JA. In situ splitting of the cadaveric liver for two

adult recipients. Transplantation 2001;72:1853-8.

212. Yersiz H, Renz JF, Hisatake G, Reichert PR, Feduska NJ Jr, Lerner S et al. Technical and

logistical considerations of in situ split-liver transplantation for two adults: Part I. Creation

of left segment II, III, IV and right segment I, V-VIII grafts. Liver Transpl 2001;7:1077-80.

213. Azoulay D, Castaing D, Adam R, Savier E, Delvart V, Karam V et al. Split-liver transplantation

for two adult recipients: feasibility and long-term outcomes. Ann Surg 2001;233:565-74.

214. Colledan M, Segalin A, Andorno E, Corno V, Lucianetti A, Spada M et al. Modified splitting

technique for liver transplantation in adult-sized recipients. Technique and preliminary

results. Acta Chir Belg 2000;100:289-91.

215. Strong R, Lynch S, Yamanaka J, Kawamoto S, Pillay P, Ong TH. Monosegmental liver

transplantation. Surgery 1995;118:904-6.

216. Mentha G, Belli D, Berner M, Rouge JC, Bugmann P, Morel P et al. Monosegmental liver

transplantation from an adult to an infant. Transplantation 1996;62:1176-8.

217. Srinivasan P, Vilca-Melendez H, Muiesan P, Prachalias A, Heaton ND, Rela M. Liver

transplantation with monosegments. Surgery 1999;126:10-2.

218. Raia S, Nery JR, Mies S. Liver transplantation from live donors. Lancet 1989;2:497.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

50 | Chapter 2

219. Strong RW, Lynch SV, Ong TH, Matsunami H, Koido Y, Balderson GA. Successful liver

transplantation from a living donor to her son. N Engl J Med 1990;322:1505-7.

220. Tanaka K, Uemoto S, Tokunaga Y, Fujita S, Sano K, Nishizawa T et al. Surgical techniques

and innovations in living related liver transplantation. Ann Surg 1993;217:82-91.

221. Emond JC, Heffron TG, Kortz EO, Gonzalez-Vallina R, Contis JC, Black DD et al. Improved

results of living-related liver transplantation with routine application in a pediatric program.

Transplantation 1993;55:835-40.

222. Miller CM, Gondolesi GE, Florman S, Matsumoto C, Muñoz L, Yoshizumi T et al. One

hundred nine living donor liver transplants in adults and children: a single-center experience.

Ann Surg 2001;234:301-11

223. Roberts JP, Hulbert-Shearon TE, Merion RM, Wolfe RA, Port FK. Influence of graft type on

outcomes after pediatric liver transplantation. Am J Transplant 2004;4:373-7.

224. Bourdeaux C, Darwish A, Jamart J, Tri TT, Janssen M, Lerut J et al. Living-related versus

deceased donor pediatric liver transplantation: a multivariate analysis of technical and

immunological complications in 235 recipients. Am J Transplant 2007;7:440-7.

225. Grewal HP, Thistlewaite JR Jr, Loss GE, Fisher JS, Cronin DC, Siegel CT et al. Complications

in 100 living-liver donors. Ann Surg 1998;228:214-9.

226. Fujita S, Kim ID, Uryuhara K, Asonuma K, Egawa H, Kiuchi T et al. Hepatic grafts from live

donors: donor morbidity for 470 cases of live donation. Transpl Int 2000;13:333-9.

227. Lo CM.Complications and long-term outcome of living liver donors: a survey of 1,508 cases

in five Asian centers. Transplantation 2003;75(3 Suppl):S12-5.

228. Kawasaki S, Makuuchi M, Matsunami H, Hashikura Y, Ikegami T, Nakazawa Y et al. Living

related liver transplantation in adults. Ann Surg 1998; 227: 269-74.

229. Soejima Y, Shimada M, Suehiro T, Hiroshige S, Ninomiya M, Shiotani S, Harada N, Hideki I,

Yonemura Y, Maehara Y. Outcome analysis in adult-to-adult living donor liver transplantation

using the left lobe. Liver Transpl 2003;9:581-6.

230. Lee S, Hwang S, Park K, Lee Y, Choi D, Ahn C et al. An adult-to-adult living donor liver

transplant using dual left lobe grafts. Surgery 2001;129:647-50.

231. Hwang S, Lee SG, Lee YJ, Sung KB, Park KM, Kim KH et al. Lessons learned from 1,000

living donor liver transplantations in a single center: how to make living donations safe. Liver

Transpl 2006;12:920-7.

232. Lo CM, Fan ST, Liu CL, Wei WI, Lo RJ, Lai CL, Chan JK, Ng IO, Fung A, Wong J. Adult-to-

adult living donor liver transplantation using extended right lobe grafts. Ann Surg 1997;

226: 261-9.

The evolution of surgical techniques in liver transplantation - a review | 51

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

233. Thuluvath PJ, Yoo HY. Graft and patient survival after adult live donor liver transplantation

compared to a matched cohort who received a deceased donor transplantation. Liver

Transpl 2004;10:1263-8.

234. Olthoff KM, Merion RM, Ghobrial RM, Abecassis MM, Fair JH, Fisher RA et al. Outcomes

of 385 adult-to-adult living donor liver transplant recipients: a report from the A2ALL

Consortium. Ann Surg 2005;242:314-23.

235. Morioka D, Egawa H, Kasahara M, Ito T, Haga H, Takada Y et al. Outcomes of adult-to-adult

living donor liver transplantation: a single institution’s experience with 335 consecutive

cases. Ann Surg 2007;245:315-25.

236. Ito T, Kiuchi T, Egawa H, Kaihara S, Oike F, Ogura Y et al. Surgery-related morbidity in

living donors of right-lobe liver graft: lessons from the first 200 cases. Transplantation

2003;76:158-63.

237. Marcos A, Orloff M, Mieles L, Olzinski AT, Renz JF, Sitzmann JV. Functional venous anatomy

for right-lobe grafting and techniques to optimize outflow. Liver Transpl 2001;7:845-52.

238. Sugawara Y, Makuuchi M, Sano K, Imamura H, Kaneko J, Ohkubo T et al. Vein reconstruction

in modified right liver graft for living donor liver transplantation. Ann Surg 2003;237:180-

5.

239. Lee SG, Park KM, Hwang S, Kim KH, Choi DN, Joo SH et al. Modified right liver graft from

a living donor to prevent congestion. Transplantation 2002; 74: 54-9

240. Lo CM, Fan ST, Liu CL, Yong BH, Wong Y, Lau GK, Lai CL, Ng IO, Wong J. Lessons learned

from one hundred right lobe living donor liver transplants. Ann Surg 2004;240:151-8.

241. Egawa H, Inomata Y, Uemoto S, Asonuma K, Kiuchi T, Fujita S et al. Biliary anastomotic

complications in 400 living related liver transplantations. World J Surg 2001;25:1300-7.

242. Liu CL, Lo CM, Chan SC, Fan ST. Safety of duct-to-duct biliary reconstruction in right-lobe

live-donor liver transplantation without biliary drainage. Transplantation 2004;77:726-32.

243. Shah SA, Grant DR, McGilvray ID, Greig PD, Selzner M, Lilly LB et al. Biliary strictures in 130

consecutive right lobe living donor liver transplant recipients: results of a Western center.

Am J Transplant 2007;7:161-7.

244. Kasahara M, Egawa H, Takada Y, Oike F, Sakamoto S, Kiuchi T et al. Biliary reconstruction

in right lobe living-donor liver transplantation: Comparison of different techniques in 321

recipients. Ann Surg 2006;243:559-66.

245. Fan ST, Lo CM, Liu CL, Tso WK, Wong J. Biliary reconstruction and complications of right

lobe live donor liver transplantation. Ann Surg 2002;236:676-83.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

52 | Chapter 2

246. Ericzon BG, Larsson M, Herlenius G, Wilczek HE; Familial Amyloidotic Polyneuropathy World

Transplant Registry. Report from the Familial Amyloidotic Polyneuropathy World Transplant

Registry (FAPWTR) and the Domino Liver Transplant Registry (DLTR). Amyloid 2003;10 Suppl

1:67-76.

247. Furtado A, Tome L, Oliveira FJ, Furtado E, Viana J, Perdigoto R. Sequential liver transplantation.

Transplant Proc 1997; 29: 467-8.

248. Khanna A, Hart M, Nyhan WL, Hassanein T, Panyard-Davis J, Barshop BA. Domino liver

transplantation in maple syrup urine disease. Liver Transpl. 2006;12:876-82.

249. Monteiro E, Perdigoto R, Furtado AL. Liver transplantation for familial amyloid polyneuropathy.

Hepatogastroenterology 1998;45:1375-80.

250. Furtado AJ. Domino liver transplantation using FAP grafts. HUC experience -hopes and

realities. Amyloid 2003;10 Suppl 1:84-7.

251. Yamamoto S, Wilczek HE, Nowak G, Larsson M, Oksanen A, Iwata T et al. Liver transplantation

for familial amyloidotic polyneuropathy (FAP): a single-center experience over 16 years. Am

J Transplant 2007;7:2597-604.

252. Azoulay D, Samuel D, Castaing D, Adam R, Adams D, Said G et al. Domino liver transplants

for metabolic disorders: experience with familial amyloidotic polyneuropathy. J Am Coll

Surg 1999;189:584-93.

253. Pacheco-Moreira LF, de Oliveira ME, Balbi E, da Silva AC, Miecznikowski R, de Faria LJ et al.

A new technical option for domino liver transplantation. Liver Transpl 2003;9:632-3.

254. Inomata Y, Zeledon ME, Asonuma K, Okajima H, Takeichi T, Ishiko T et al. Whole-liver

graft without the retrohepatic inferior vena cava for sequential (domino) living donor liver

transplantation. Am J Transplant 2007;7:1629-32.

255. Jabbour N, Gagandeep S, Genyk Y, Selby R, Mateo R. Caval preservation with reconstruction of

the hepatic veins using caval-common iliac bifurcation graft for domino liver transplantation.

Liver Transpl 2006;12:324-5.

256. Mergental H, Gouw AS, Slooff MJ, de Jong KP. Venous outflow reconstruction with

surgically reopened obliterated umbilical vein in domino liver transplantation. Liver Transpl

2007;13:769-72.

257. Azoulay D, Castaing D, Adam R, Mimoz O, Bismuth H. Transplantation of three adult

patients with one cadaveric graft: wait or innovate. Liver Transpl 2000;6:239-40.

258. Absolon KB, Hagihara PF, Griffen WO Jr, Lillehei RC. Experimental and clinical heterotopic

liver homotransplantation. Rev Int Hepatol. 1965;15:1481-90.

259. Fortner JG, Beattie EJ Jr, Shiu MH, Kawano N, Howland WS. Orthotopic and heterotopic

liver homografts in man. Ann Surg 1970;172:23-32.

The evolution of surgical techniques in liver transplantation - a review | 53

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

260. de Rave S, Hansen BE, Groenland TH, Kazemier G, de Man RA, Metselaar HJ et al. Heterotopic

vs. orthotopic liver transplantation for chronic liver disease: a case-control comparison of

short-term and long-term outcomes. Liver Transpl 2005;11:396-401.

261. Terpstra OT, Metselaar HJ, Hesselink EJ, de Rave S, Groenland TH, Stibbe J et al..

Auxiliary partial liver transplantation for acute and chronic liver disease. Transplant Proc

1990;22:1564.

262. Ringers J, Dubbeld J, Baranski AG, Coenraad M, Sarton E, Schaapherder AF et al. A novel

technique for auxiliary partial liver transplantation with reno-portal anastomosis and

avoidance of the hepatoduodenal ligament. Am J Transplant 2006;6:2802-8.

263. Gubernatis G, Pichlmayr R, Kemnitz J, Gratz K. Auxiliary partial orthotopic liver transplantation

(APOLT) for fulminant hepatic failure: first successful case report. World J Surg 1991;15:660-

5.

264. van Hoek B, de Boer J, Boudjema K, Williams R, Corsmit O, Terpstra OT. Auxiliary versus

orthotopic liver transplantation for acute liver failure. EURALT Study Group. European

Auxiliary Liver Transplant Registry. J Hepatol 1999;30:699-705.

265. Durand F, Belghiti J, Handra-Luca A, Francoz C, Sauvanet A, Marcellin P et al. Auxiliary liver

transplantation for fulminant hepatitis B: results from a series of six patients with special

emphasis on regeneration and recurrence of hepatitis B. Liver Transpl 2002;8:701-7.

266. Chenard-Neu MP, Boudjema K, Bernuau J, Degott C, Belghiti J, Cherqui D et al. Auxiliary

liver transplantation: regeneration of the native liver and outcome in 30 patients with

fulminant hepatic failure--a multicenter European study. Hepatology 1996;23:1119-27.

267. Bismuth H, Azoulay D, Samuel D, Reynes M, Grimon G, Majno P et al. Auxiliary partial

orthotopic liver transplantation for fulminant hepatitis. The Paul Brousse experience. Ann

Surg 1996;224:712-24.

268. Sudan DL, Shaw BW Jr, Fox IJ, Langnas AN. Long-term follow-up of auxiliary orthotopic liver

transplantation for the treatment of fulminant hepatic failure. Surgery 1997;122:771-7.

269. Azoulay D, Samuel D, Ichai P, Castaing D, Saliba F, Adam R et al. Auxiliary partial orthotopic

versus standard orthotopic whole liver transplantation for acute liver failure: a reappraisal

from a single center by a case-control study. Ann Surg 2001;234:723-31.

270. Jaeck D, Boudjema K, Audet M, Chenard-Neu MP, Simeoni U et al. Auxiliary partial orthotopic

liver transplantation (APOLT) in the treatment of acute liver failure. J Gastroenterol 2002;37

Suppl 13:88-91.

271. Boudjema K, Bachellier P, Wolf P, Tempe JD, Jaeck D. Auxiliary liver transplantation and

bioartificial bridging procedures in treatment of acute liver failure. World J Surg 2002;26:264-

74.

regel 1

regel 2

regel 3

regel 4

regel 5

regel 6

regel 7

regel 8

regel 9

regel 10

regel 11

regel 12

regel 13

regel 14

regel 15

regel 16

regel 17

regel 18

regel 19

regel 20

regel 21

regel 22

regel 23

regel 24

regel 25

regel 26

regel 27

regel 28

regel 29

regel 30

regel 31

regel 32

regel 33

regel 34

regel 35

regel 36

regel 37

regel 38

regel 39

54 | Chapter 2

272. Kasahara M, Takada Y, Egawa H, Fujimoto Y, Ogura Y, Ogawa K et al. Auxiliary partial

orthotopic living donor liver transplantation: Kyoto University experience. Am J Transplant

2005;5: 558-65.

273. Rela M, Muiesan P, Vilca-Melendez H, Dhawan A, Baker A, Mieli-Vergani G, Heaton ND.

Auxiliary partial orthotopic liver transplantation for Crigler-Najjar syndrome type I. Ann Surg

1999; 229:565-9.

274. Inomata Y, Kiuchi T, Kim I, Uemoto S, Egawa H, Asonuma K, Fujita S, Hayashi M, Tanaka

K. Auxiliary partial orthotopic living donor liver transplantation as an aid for small-for-size

grafts in larger recipients. Transplantation 1999;67:1314-9.