Open access peer-reviewed chapter

Assessment and Optimization of the Future Liver Remnant

Written By

Mandivavarira Maundura and Jonathan B Koea

Submitted: 12 April 2016 Reviewed: 04 October 2016 Published: 05 April 2017

DOI: 10.5772/66139

From the Edited Volume

Updates in Liver Cancer

Edited by Hesham Mohamed Abdeldayem

Chapter metrics overview

2,373 Chapter Downloads

View Full Metrics

Abstract

Safe liver resection is a vital element in the management of primary and secondary hepatic malignancies. The indications for resection have evolved Over time, and this has in part been due to the ability to improve the future liver remnant (FLR). This chapter reviews the current and future methods used for assessing the future liver remnant volume and function in order to minimize the risk of post-hepatectomy liver failure (PHLF). Current and evolving methods used in augmenting the future liver remnant are also considered. Since its introduction in the 1990s, portal venous embolization (PVE) has become the most widely used method of augmenting the FLR. The factors that affect hypertrophy following embolization as well as techniques used in portal venous embolization will be reviewed. Other methods of augmentation discussed include portal vein ligation (PVL) and the emerging method of associating liver partition and portal vein ligation for staged hepatectomy (ALPPS). The chapter also considers the various methods in the context of limiting tumour progression in the future liver remnant and attempts to integrate newer techniques such as ALPPS into current treatment algorithms.

Keywords

  • future liver remnant
  • volume
  • portal venous embolization
  • hypertrophy
  • liver function measurement
  • resectability

1. Introduction

Safe liver resection is a vital element in the curative management of primary and secondary hepatic malignancies. The ability to perform major liver resections relies on the capacity of the future liver remnant (FLR) to maintain normal liver function. The quality of the FLR may also be influenced by pre-existing liver disease and/or prior chemotherapy, thereby limiting the size of resection possible. Various methods have been utilized to assess the size and functionality of the future liver remnant to avoid post-hepatectomy liver failure and, when major liver resections are being considered, techniques are available to increase the volume of the FLR.

The commonest indication worldwide for hepatic resection is to treat colorectal cancer metastatic to the liver, and overtime the criteria for surgical resectability of colorectal liver metastases has evolved (Table 1). Initially, surgically resectable colorectal liver metastases included low volume, unilobar disease of 1–3 metastases which could be resected with a 1 cm macroscopic margin and no evidence of extrahepatic disease [1]. More recently, the number or bilaterality of metastases is not in itself a contraindication provided they can be resected with a macroscopic margin with an adequate FLR [2], and increasingly the presence of localized extrahepatic disease [2] is not an absolute contraindication to resection. However, the response of metastases to chemotherapy has emerged as an important prognostic factor for disease-free survival [3], and consequently, most patients now receive neoadjuvant treatment prior to resection. These factors and a globally more aggressive policy of resection have increased the numbers of patients eligible for potentially curative therapy but placed new emphasis on the importance of accurate assessment of the volume and function of the FLR.

CharacteristicsHistorical indicationsCurrent indications
Tumour number<4 lesionsAny
Lobes involvedUnilobarBilobar or unilobar
Size of tumour<5 cmAny
Extrahepatic diseaseNoneTreatable extrahepatic disease
Functional Liver RemnantAdequateAdequate or amenable to augmentation
Lymph node involvementNo hepatic pedicle nodesNo coeliac node involvement
SynchronicityMetachronousMetachronous or synchronous
Venous involvementNo vena caval or hepatic venous involvementVenous resection or reconstruction

Table  1.

Indications for surgical resection of liver metastases.

Adapted from Sherman and Mahvi [2].


The aim of this chapter is to review currently available methods to assess the quality and volume of the FLR pre-operatively as well as summarizing the methods used to improve the FLR including pre-operative portal venous embolization (PVE) and associating liver partition and portal vein ligation for staged hepatectomy (ALPPS).

Advertisement

2. Pre-operative assessment of liver function

Whether the indication for liver resection is a primary or secondary liver malignancy in order for surgery to be successful, the patient must be able to tolerate the physical and psychological challenges of surgery and the FLR must be able to sustain liver function. A thorough history and examination should identify presence and extent of comorbidities [4]. The assessment should also include assessment of liver function tests, coagulation status, full blood count and platelet count, relevant tumour markers and cross-sectional radiology [4]. All radiology and clinical information should be reviewed in a multidisciplinary meeting and early input obtained from specialist services such as hepatology, interventional radiology and medical oncology.

2.1. Assessing liver function

2.1.1. Liver function tests

The assessment of liver function is complex and largely reliant on surrogate markers. Initial clinical assessment involves assessment for signs of overt liver disease such as jaundice, spider naevi and palmar erythema. An initial set of liver function tests including measurement of plasma bilirubin, transaminases, γ-glutamyl transferase and alkaline phosphatase as well as albumin and prothrombin time should be performed [5]. Two commonly used scoring systems have been developed using these parameters to assess liver function and associated surgical risk.

2.1.2. Scoring systems

The Child-Pugh (CP) and Model for End-Stage Liver Disease (MELD) scores are the most widely used stratification scores used in making decisions regarding surgery in cirrhotic patient (Tables 2 and 3).

Factor1 point2 points3 points
Bilirubin (μmol/L)<3434–50>50
Albumin (g/L)>3528–35<28
INR<1.71.7–2.2>2.2
AscitesNoneDiuretic controlledRefractory
EncephalopathyNoneGrade I-II (medication controlled)Grade III-IV (refractory)

Table 2.

Child-Pugh score.

Class A = 5–6 points, Class B = 7–9 points, Class C = 10–15 points.


INR; international normalized ratio.


Modified from Hanje and Patel [10].


MELD = 3.78 × ln[serum bilirubin (mg/dL)] + 11.2 × ln[INR] + 9.57 × ln[serum creatinine (mg/dL)] + 6.43
Predicts development post-operative liver failure post-hepatectomy for hepatocellular carcinoma where a score >11 is predictive of worse outcome. Maximum creatinine is 4.0 mg/dL. Patients dialysing twice within the last week are assigned the maximum creatinine.
Adapted from Hanje and Patel 2007 [10] and Cha 2012 [39].

Table 3.

MELD score.

The CP score has been in clinical use for several decades and is based on the patient’s albumin, bilirubin, coagulation studies, severity of ascites and encephalopathy [6]. Individuals are stratified to Child A, B and C, and these correspond to increasing risk of perioperative mortality as well as post-operative complications such as bleeding, infection, ascites, renal failure and hepatic failure [6] (Figure 1).

Figure 1.

Relative risk of perioperative complication or death with increasing Child-Pugh and MELD scores. Adapted from Hanje and Patel [10].

The MELD score originally used to predict mortality following transjugular intrahepatic portosystemic shunt (TIPS) has since been extrapolated to stratifying liver transplant patients as well predicting perioperative mortality [6] (Figure 1).

2.2. Dynamic tests of liver function

These tests are based on complete hepatic clearance or metabolism of a substrate following intravenous administration and include indocyanine green (ICG) clearance as well as nuclear medicine techniques.

2.2.1. Indocyanine green clearance

ICG is the most widely discussed pre-operative test to assess liver function. Historically, the test entails intravenous administration of ICG with multiple blood samples taken at 15-min intervals to determine plasma clearance but has become easier to perform with the availability of non-invasive bedside monitors [7]. ICG is a water soluble, inert tricarbocyanine with a hepatic extraction rate above 70%, and is almost completely excreted in its unchanged form by the liver [7].

Test results are most commonly expressed as percentage of ICG retained after 15 min (ICG-R15), and however, they can also be reported as the plasma disappearance rate (ICG PDR) or as the ICG elimination rate constant [8] (Figure 2). The safety limit when expressed as ICG-R15 varies from 14 to 20% [8].

Figure 2.

Indocyanine green plasma clearance curve obtained from serial blood sampling or optical pulse spectrophotometry. Retention at 15 min (arrow) is commonly used to assess liver function. Modified from Cha et al. [39].

The use of ICG is limited in the presence of hyperbilirubinaemia since uptake is by the same hepatic transporters [8] and will therefore artificially decrease ICG clearance. The test is also dependent on overall liver blood flow and is less reliable in those with non-flow-dependent liver diseases [8].

2.2.2. Nuclear Medicine

Scintigraphy has been used to provide quantitative information on total and regional liver function using a variety of radiolabelled probes.

2.2.2.1. 99mTc-Mebrofenin

Mebrofenin is the iminodiacetic acid (IDA) analogue with the highest specificity for hepatocytes [8, 9]. It is absorbed by hepatocytes and eliminated in the bile without biotransformation in a similar fashion to bilirubin [8, 9]. The rate of hepatocyte uptake of technicium-labelled mebrofenin can be quantitatively assessed using scintigraphy and rate of biliary excretion determined.

2.2.2.2. 99mTc-GSA

(99m)Tc-DTPA-galactosyl serum albumin (where DTPA is diethylenetriaminepentaacetic acid) binds to the asialoglycoprotein receptor found on the sinusoidal surface of the hepatocyte[8]. 99mTc-GSA is an asialoglycoprotein analogue that is taken up only in the liver [8]. The uptake of this agent is not affected by hyperbilirubinaemia and can therefore still be used for liver function assessment in the cholestatic patient [8]. Scintigraphy permits assessment of hepatic uptake as measure of function, and 99mTc-GSA remains trapped in the liver which permits further assessment of liver volume. However, this agent is not widely available outside of Japan.

2.2.3. 13C-Methacetin Breath Test (LiMax)

The 13C-methacetin breath test is based on activity of the cytochrome P450 1A2 (CYP1A2) enzyme system [8]. The system is distributed throughout the liver and is not affected by drugs or genetic variation [8]. 13C-methacetin is exclusively metabolized by CYP1A2 into paracetamol and 13CO2 [10]. The test is performed by measuring 13CO2/12CO2 ratio in expired breath before and after administration of 13C-methacetin. The result is expressed in μg/kg/h and gives total liver function. If combined with computed tomographic (CT) scan, it may be used to approximate the function of a section of liver, and however, this assumes uniform distribution of hepatic function, and it is known that this may vary between segments [8].

Advertisement

3. Radiological measurement of liver volume

Multiple cross-sectional imaging modalities are available for imaging the liver and include ultrasound, CT and magnetic resonance imaging (MRI). Data obtained with these investigations can be used to volumetrically assess the FLR as well as define the presence and position of hepatic tumours and the presence of chronic liver disease.

3.1. Ultrasound

Transabdominal ultrasound is widely available, non-invasive and low cost. However, it is operator independent, and its accuracy may be affected by body habitus, the presence of ileus or ascites as well as the presence of diffuse hepatic disease and steatosis which may be seen following chemotherapy [11].

In patients with colorectal liver metastases, the sensitivity of lesion-by-lesion analysis ranges from 60.9 to 64.9%. The specificity ranges from 50 to 60%, and the range increases from 76.7 to 83.3% with the use of contrast [11]. The increased sensitivity of contrast-enhanced US (80–90%) makes it useful in guiding the percutaneous biopsies of lesions [11].

Three-dimensional ultrasound probes are available, but the use of transabdominal ultrasound in hepatic volumetry assessment remains limited by the previously stated problems of body habitus and operator expertise [12]. Ultrasound is also routinely used intraoperatively where it may identify occult liver metastases denoting unresectable disease in up to 25% of patients [13] but currently has no role in assessing FLR volume.

3.2. Computed tomography (CT)

CT has become widely available and is relatively inexpensive. It offers the ability not only to detect lesions, but also to detect accurately localize lesions as well as their vascular and biliary relations [11]. It does involve exposure to ionizing radiation and the risk of allergic reactions to iodinated contrast [11]. The lesion-by-lesion sensitivity is up to 75% [11] although the rate of detection decreases with size of the lesion with a 16% detection rate for lesions smaller than 10mm in diameter [11]. The ability to construct three-dimensional (3D) models from the images allows for more accurate planning of surgical resection and appreciation of intrahepatic vascular anatomy prior to resection [14].

CT scan is also commonly used to estimate the volume of the FLR by directly quantifying the volume from scan acquired data. The FLR volume is measured by CT and then standardized to the total estimated liver volume (TELV) [15].

TELV(cm3)=794.41+1267.28×BSA(m2)E1

The ratio of the CT measured FLR volume to the TELV is known as the standardized FLR (sFLR) that allows a uniform comparison of FLR volume before and after PVE [15]. More commonly, total liver volume can be measured using data acquired in the CT scan, and comparison is made with the directly measured volume of the FLR (Figure 3).

Figure 3.

A: Axial CT scan showing solitary colorectal metastasis in segment 8 (arrow) with a congenitally small left lobe. B: Three-dimensional reconstruction and total liver volume (1531.20 ml) measured 6 weeks following right portal vein embolization. C: Three-dimensional reconstruction and total remnant volume (314.67 ml) measured 6 weeks following right portal vein embolization. The left-sided remnant now constitutes 20.5% of the total liver volume and the patient proceeded to right hepatic lobectomy.

3.3. Magnetic resonance imaging (MRI)

MRI is the most accurate of the available modalities in detecting colorectal metastases as well as many other malignancies. It does not make use of ionizing radiation and gadolinium-based extracellular agents or hepatocyte-specific contrast agents such as gadoxetic acid may be used as contrast [11]. Overall, the sensitivity of contrast-enhanced MRI is 94% for colorectal metastases[11] and is superior to CT scan in the detection of small lesions as well as lesions in steatotic livers [11]. MRI is not routinely used clinically to assess hepatic volumetry.

3.4. PET CT

Fluorodeoxyglucose positron emission tomography (18FDG PET) provides metabolic information which when combined with CT provides a metabolic map of glucose uptake [16] that is highly specific for cancer. PET CT is less sensitive in the detection of hepatic lesions than CT or MRI but is more specific and is able to accurately define the presence of extrahepatic disease [16]. PET CT is not currently used to assess hepatic volumetry.

Advertisement

4. Combined imaging and dynamic tests

As previously described, available imaging and liver function tests have a number of shortcomings with regard to estimation of function of the future liver remnant. This is important since hepatic volume and hepatic function do not have a linear correlation. A number of groups have attempted to improve predictability of FLR volume and function by combining modalities (Table 4).

GroupModalitiesOutcome
Chapelle et al. [25]99mTc-mebrofenin/FLRVPredicts future liver function after resection (eFLRF). Cut-off of 2.3%/min/m2 for eFLRF would have prevented all mortalities related to PHLF
Hwang et al. [40]FRL-kICG (derived ICG and CT volumetry)Appeared to predict PHLF risk quantitatively
De Graaf et al. [41]99mTc-mebrofenin/SPECTNo difference between actual FLR and predicted FLR

Table 4.

Examples of combined modalities in FLR estimation.

PHLF, post-hepatectomy liver failure. FRL-kICG is the ICG clearance rate constant (ICG-K) fraction of future remnant liver to total liver volume.


Advertisement

5. Definition of an adequate future liver remnant

With regard to the future liver remnant, an FLR ≤20% of total hepatic volume is the strongest predictor of hepatic insufficiency and is thus set as the minimum FLR volume for a healthy non-cirrhotic liver [17]. It has generally been regarded that those who have received chemotherapy for longer than 12 weeks should have an FLR >30% of total hepatic volume and those with fibrosis or cirrhosis an FLR >40–50% of total hepatic volume [17]. However, it must be emphasized that patients with cirrhotic livers, even where FLR is adequate, remain at increased risk of wound breakdown, infection, ascites and fluid retention, as they would be for any major surgery.

The increased FLR requirement for patients who have received chemotherapy is based on the premise that pre-operative chemotherapy may cause liver damage or increase the risk of post-operative complications [18]. Treatment with irinotecan is associated with rates of steatohepatitis as high as 20.2% compared to 4.4% in those not on chemotherapy [19]. Treatment with oxaliplatin is associated with hepatic sinusoidal injury that can result in venoocclusive disease and nodular regenerative hyperplasia [19]. There is greater morbidity following hepatectomy in those with evidence of sinusoidal obstruction syndrome and a greater risk of perioperative blood transfusion [19].

However, the majority of investigations have shown (Table 5) that liver injury in the setting of neoadjuvant therapy does not appear to have significant clinical consequences if chemotherapy is maintained until a response is observed and disease is then resected as early as is feasible [20].

AuthorInterventionNo.ComparisonFLR effect
Goéré et al. [42]PVE20≥1 -month interval vs no intervalNone
Ribero et al. [43]PVE112Chemo vs no chemoNone
Gruenberger et al. [44]Hepatectomy52None
Covey et al. [45]PVE100Chemo vs no chemoNone
Aussilhou et al. [46]PVE40Chemo+bevacizumab/chemo without bevacizumabImpaired FLR/none
Tanaka et al. [47]PVE/Hepatectomy60Chemo vs no chemoNone
Sturesson et al. [48]PVE26Chemo vs no chemoImpaired FLR
Sturesson et al. [49]Hepatectomy74Chemo vs no chemoImpaired FLR
Beal et al. [50]Hepatectomy72Chemo vs no chemo
>6 cycles vs ≤6 cycles
None
Dello et al. [51]Hepatectomy72Chemo vs no chemo
>6 cycles vs ≤6 cycles
None
Fischer et al. [52]PVE64Chemo vs no chemoNone

Table 5.

Effect of chemotherapy on liver hypertrophy.

PVE, portal vein embolization; FLR, future liver remnant.


Adapted from Simoneau et al. [27].


Advertisement

6. Augmentation of the future liver remnant

With advances in surgical technique and radiological imaging, more extensive liver resections have become feasible and the challenge remains the ability to maintain liver function post-operatively. Methods have been developed aiming to increase the size of the future liver remnant. These include portal venous embolization, portal vein ligation (PVL) and associating liver partition and portal vein ligation for staged hepatectomy (ALPPS). Augmentation is recommended where FLR is anticipated to be ≤20% for normal, ≤30% for those with chemotherapy associated steatohepatitis and ≤40–50% in those with cirrhosis [21].

6.1. Portal venous embolization

Portal venous embolization was developed to improve the size and function of the FLR in tumour bearing liver by occluding the ipsilateral portal vein, the non-tumour bearing contralateral side, which is to be the FLR, increases in volume by a combination of hypertrophy and hyperplasia [22]. The capacity for regeneration in the otherwise healthy liver is significant, and PVE results in increased FLR volume in roughly 60% of patients with the average increase in volume being 12% [17]. The response is variable, and the size of the FLR prior to PVE may predict the degree of hypertrophy [23]. There is evidence to suggest that the degree of hypertrophy is inversely proportional to the FLR ratio before PVE such that a smaller FLR will have a larger hypertrophy [24]. PVE is contraindicated in the presence of ipsilateral portal vein tumour thrombus or occlusion and in patients with severe portal hypertension [22].

PVE may be performed under general anaesthesia or local anaesthesia with sedation, and the approach may be contralateral or ipsilateral [24]. The procedure is commonly used to occlude the right portal vein and induce left lobe hypertrophy. In the contralateral approach, the left portal vein is punctured to give access for embolization of the right portal vein [24]. The contralateral approach is less technically demanding, and however, it does risk potential injury to the FLR [24]. Where extended right hepatectomy is considered, embolization of portal vein branches to segment IVa and IVb can be undertaken. A short segment (1 cm) of unembolized right portal vein may be left to allow for surgical ligation during resection [25].

Various agents have been used for embolization, and most are associated with adequate hypertrophy rates and acceptable complication profiles (Table 6).

Embolic agentAuthorsNo. of patientsIncrease FRL %
Gelatin spongeFuji et al. [53]
Kusaka et al. [54]
Kazikawa et al. [55]
Nanashima et al. [56]
30
18
14
30
17.8
21.2
23.8
29.4
Polyvinyl alcohol + coils or plugsCovey et al. [44]
Van den Esschert et al. [57]
Libicher et al. [58]
100
10
10
24.3
26.1
26.4
N-butyl cyanoacrylateDe Baere et al. [59]
Giraudo et al. [60]
Elias et al. [61]
Broering et al. [62]
107
146
68
17
57.8
41.7
59.1
69.4
Fibrin glueNagino et al. [63]
Liem et al. [64]
105
15
27.4
31.4

Table 6.

Hypertrophic response to embolic agents.

Modified from Loffroy et al. [24].


The degree of FLR hypertrophy is also influenced by the health of the underlying liver. Non-cirrhotic liver hypertrophies at a rate of about 12–21 cm3/day at 2 weeks compared to just 9 cm3/day for a cirrhotic liver [22] and the growth rate can be used to predict the probability of liver failure and major complications [23] (Figure 4).

Figure 4.

Nonparametric regression of measured future liver remnant growth rate to predict probability of liver failure. Minimal or negative remnant growth rate following portal vein embolization was associated with higher rates of post-resection liver failure. Reproduced from Leung et al. [23] with permission.

While PVE is used for its effect on the contralateral lobe of liver, it has also been associated with tumour progression. Liver growth is regulated by a number of growth factors and cytokines the up-regulation of which is known to be involved in multiple tumour pathways [26]. Other factors thought to contribute to tumour progression are the compensatory increased inflow via the hepatic artery and the cellular host response [26]. There are currently no specific therapies available aimed at limiting the effects of these growth factors and cytokines on tumour progression. However, two-stage hepatectomy or ablation can be used where disease in the FLR is resected or ablated prior to PVE or PVL [14].

The timing of definitive resectional surgery following PVE is not formally prescribed, and however, most investigations report repeating imaging (usually CT scan) at 4–6 weeks following PVE and, if sufficient FLR volume has been achieved, undertaking resection soon afterward. Simoneau et al. demonstrated an increase of 1-day post-PVE increased the risk of tumour progression by 1% [27]. It has been suggested that earlier surgery such as 2 weeks after PVE may reduce the risk of tumour progression [26].

6.2. Portal vein ligation

PVL is undertaken operatively and often in the setting of staged hepatectomies where small tumours in the FLR are removed or ablated and open ligation of the right portal vein is performed. Pandanaboyana et al. in their meta-analysis found that PVE and PVL had a mean percentage increase in FLR volume of 39% and 27%, respectively; however, this did not reach statistical significance [28]. They proposed that this may be explained the observation that the later formation of portoportal collaterals does not impact on liver hypertrophy as this is induced early after portal occlusion [28].

6.3. Associated liver partition and portal vein ligation

ALPPS was first performed in 2007 and was noted to result in significant hepatic hypertrophy with increased resectability in those with large tumours [29]. The procedure was performed during an exploration for hilar cholangiocarcinoma. A left hepaticojejunostomy was performed to reduce cholestasis to the FLR, the liver was divided along the falciform ligament, and the right portal vein was ligated [29]. A CT scan performed 8 days later demonstrated a 94% increase in FLR, and the resection was successfully completed the following day [29].

The classical ALPPS procedure for a large right-sided tumours involves right portal vein ligation, ligation and division of the segment IV portal branches as well as transection of the liver parenchyma along the falciform ligament [29]. Any tumour deposits within the future liver remnant can also be resected or ablated at this time.

Associating liver tourniquet and portal ligation for staged hepatectomy (ALTPS) is a variation in which rather than dividing the liver parenchyma and ligating the portal vein, the effect of occluding portal flow and transection is achieved by use of tourniquet [29] and radiofrequency or microwave ablation may also be combined with portal vein ligation [29]. In an attempt to reduce surgical complications following the initial surgical procedure, a partial ALPPS in which parenchymal transection is not complete has also been described [29].

The advantage ALPPS over two-stage hepatectomy is that it may improve the feasibility of resecting previously unresectable tumours owing to the very high FLR gains seen. The short time between the procedures makes tumour progression unlikely [30].

In 2012, the International ALPPS registry was formed to systematically collect data from multiple centres performing the procedure, and the first analysis was published in 2014 [31]. This analysis which included 202 patients, 70% of whom had an underlying diagnosis colorectal liver metastases, and reported a 90-day mortality of 9% [31] (Table 7). Independent risk factors for in hospital severe complications included patient age >60 years, tumours other than colorectal liver metastases as well as 2 markers of complex liver resection (stage 1 ALPPS procedure >5 h and/or the need for intraoperative blood transfusions) [31].

AuthorNumber of patientsSimultaneous colorectal resectionFLR hypertrophy %Mean LOSMorbidity%Mortality %Follow-up daysDFS %OS%
Schnitzbauer et al. [65]25074nr681260-7768086
Alvarez et al. [66]15378.41953018–41073100
Li et al. [67]9nr87.2nr10022nrnrnr
Dokmak and Belghiti [68]8nr704290nrnrnrnr
Machado et al. [69]8nr88nrnr0nrnrnr
Donati et al. [70]8nr66–200nrnrnrnrnrnr
Adriani et al. [71]2nrnrnrnr0912–14600100
Govil [72]1nrnrnrnrnrnrnrnr
Oldhafer et al. [73]10nrnrnr0nrnrnr
Conrad et al. [74]1nr45nr00nrnrnr
Cavaness et al. [75]1010013nr0nrnrnr

Table 7.

Overview of hypertrophy, mortality and morbidity following ALPPS for various studies.

Adapted from Alvarez et al. [76].


Nr, not reported; DFS, disease-free survival; OS, overall survival.


The data in Table 7 confirm that ALPPS is a physiologically demanding procedure, and there is a paucity of data concerning long-term outcomes. Buac et al. [32] recently conducted a survey of 66% of the surgeons contributing to the International ALPPS registry and noted that there was significant variability in the indications for surgery as well as how it is performed. Currently, PVE is widely used while ALPPS is still under investigation. Schadde et al. [31] published a head to head comparison of the two procedures (Table 8).

Reason for failurePVE/PVL (n = 83)ALPPS (n = 48)
Perioperative death (90 days) %6%15%
No stage 2 due to tumour progression %16%0%
No stage 2 due to failure to grow%7%0%
R1 resection %5%2%
Failure to reach primary endpoint %34%17%

Table 8.

Mortality and outcomes of ALPPS v PVE/PVL. Modified from Schadde et al. 2014 [31].

6.4. Assessment of liver remnant volume using ICG clearance intraoperatively during vascular exclusion (ALIIVE)

This is a newly reported intraoperative procedure, which may have the advantage of some planned ALPPS procedures occurring as a single-step hepatectomy as well as identifying the need for an ALPPS procedure where one had not been planned [33]. The technique involves non-invasive measurement of ICG PDR at 5 points during resectional surgery. Measurement occurs before anaesthetic induction (ICG 1), following mobilization of FLR (ICG 2), during inflow occlusion of the resection lobe (ICG 3), following parenchymal transection with inflow occlusion (ICG 4) and finally during inflow as well as outflow occlusion following parenchymal transection (ICG 5/ALIIVE) [33].

The aim of this test is to replicate the post-resection state intraoperatively. Lau et al. published their initial experience and while their series was too small to deduce an ICG PDR cut-off level, they suggested that as the post-hepatectomy state was replicated, it was likely that previously demonstrated cut-off levels could be applied to the procedure [33]. Previous studies have suggested a PDR >9%/min would likely be safe, while a PDR <7%/min would confer a high risk of insufficiency [33]. Interestingly, the only mortality of the 10 patients had an ALIIVE ICG of 7.1%/min [33].

This procedure will require further validation studies, but could certainly spare some patients a second procedure or allow others the opportunity of an ALPPS procedure if not already planned pre-operatively.

6.5. Transarterial embolization (TAE)

Transarterial chemoembolization (TACE) is based on the concept that blood supply to tumour is generally derived from the hepatic artery [34]. Following portal venous embolization, the hepatic arterial flow is increased and this is thought to preserve viability of the embolized lobe [35]. However, there may still not be adequate hypertrophy of the FLR, and it has been suggested that further interruption of the vascular inflow (arterial occlusion) may result in further hypertrophy [35]. However, the near complete occlusion that occurs with PVE followed by TAE may induce parenchymal infarction, and this sequence currently has few applications [35].

Conversely, TAE followed by PVE has demonstrated safety in case of hepatocellular carcinoma with inadequate FLR [35]. Unfortunately, this has not been useful in management of colorectal metastases as these tumours are generally not fed by an artery [35].

6.6. Radioembolization

Selective internal radiation therapy (SIRT) with Y-90 has generally been used as treatment for locally advanced liver tumours. A transarterial catheter is introduced, and Y-90 microspheres are infused to lodge at the tumour arteriolar level. The radioactive microspheres result in reduced blood flow to the tumour but also deliver Y-90 brachytherapy [36]. The reported rates of response are 42–70% for colorectal liver metastases [36] and, in addition to the local control of tumour, unilateral treatment has been noted to result in hypertrophy of the contralateral liver lobe [37]. The theoretical advantage of SIRT over PVE would then be that tumour progression might not continue unabated while awaiting hypertrophy of the FLR if SIRT were selectively administered. Teo et al. [36] in a systematic review found that, while the degree of hypertrophy from SIRT was comparable to that of PVE (26–47% vs 10–46%), the time interval over which growth occurred was much slower than that of PVE (44 days to 9 months vs 2–8 weeks) making it less likely to be clinically useful.

6.7. Associating portal embolization and artery ligation (APEAL)

This procedure combines portal vein embolization and arterial ligation [38]. At the first stage of the procedure, the FLR is surgically mobilized as in the ALPPS procedure. The right portal vein is embolized before being ligated and divided [38]. A right sectoral hepatic artery ligation is performed (either the artery for segments V/VIII or segments VI/VII), and the segment IVb inflow is also interrupted [38]. There is no parenchymal transection. The second, resection stage of the procedure is undertaken 1–2 months later.

Dupre et al. [38] published their series of 10 patients who had required two-stage extended right hemihepatectomy for bilobar colorectal metastases. All the patients included had a low FLR volume and/or prolonged pre-operative chemotherapy and the procedure resulted in FLR hypertrophy of over 100% at day 7 [38]. There were no complications related to hepatic necrosis, and the authors suggest that the avoidance of parenchymal dissection reduces the risk of bile leak and infection. The interval of 1–2 months between stages was chosen to allow for post-operative recovery and to identify those with rapidly progressive disease. Initial results suggest morbidity and mortality rates comparable to ALPPS and PVE, and however, more long-term results and further validation studies are required [38].

Advertisement

7. Conclusion

The indications for liver resection continue to evolve as do the improvements in radiological ability to assess disease extent and accurately measure FLR volume. This information enables surgical teams to precisely calculate perioperative risk and determine resectability—almost to the millimetre. There is an evolving use biochemical markers which, when combined with imaging, may improve the safety of surgery further by allowing not only for estimating the volume but also the function of the future liver remnant.

The development of surgical techniques such as ALPPS, ALIIVE as well as adjuncts to surgery such as PVE/PVL and perhaps SIRT are increasing the number of patients who can be considered to have resectable disease. This would not be possible in the absence of oncological advancements as well as improvement in perioperative care. As our imaging and functional assessment technology improves, current management algorithms will also evolve (Figure 5).

Figure 5.

Example of future algorithm. ALIIVE technique could be utilized at the time of first stage hepatectomy or ALPPS to determine whether resection can be completed at first stage.

References

  1. 1. Dunne DFJ, Parks RW, Jones RP, Adam R, Poston G. Colorectal liver metastases. In: Garden OJ A Companion to Specialist Surgical Practice: Hepatobiliary and Pancreatic Surgery 5th ed. Elsevier New York; 2014. p. 109–131.
  2. 2. Sherman K, Mahvi D. Liver metastases. In: Niederhuber JE, Armitage JO, Doroshow JH. Karstan MB, Tepper JE. Abeloff’s Clinical Oncology 5th ed. Saunders; Philadelphia 2014. p778–793.
  3. 3. Brouquet A, Nordlinger B. Neoadjuvant and adjuvant chemotherapy in relation to surgery for colorectal liver metastases. Scandinavian Journal of Gastroenterology. New York 2012;47:286–295.
  4. 4. Koea J. Cancer of the Bile Ducts: Intrahepatic Cholangiocarcinoma. In: Jamagin W, Blumgart L editors, Blumgart’s Surgery of the Liver, Biliary Tract and Pancreas 5th ed. Saunders Philadelphia 2012; p760–770.
  5. 5. Wigmore SJ, Parks RW, Stutchfield BM, Forbes SJ. Liver function and failure. In: Garden OJ editor, A Companion to Specialist Surgical Practice: Hepatobiliary and Pancreatic Surgery 5th ed. Elsevier, New York; 2104. p.1–16.
  6. 6. Friedman L. Surgery in the patient with liver disease. Transactions of the American Clinical and Climatological Association. 2010;121:192–205.
  7. 7. De Gasperi A, Mazza E, Prosperi M. Indocyanine green kinetics to assess liver function: ready for a clinical dynamic assessment in major liver surgery? World Journal of Hepatology. 2016;8:355–367. doi: 10.4254/wjh.v8.i7.355
  8. 8. Cieslak KP, Runge JH, Heger M, Stoker J, Bennink RJ, Van Gulik TM. New perspectives in the assessment of future remnant liver. Digestive Surgery. 2014;31:255–268. doi:10.1159/000364836
  9. 9. Geisel D, Lüdemann L, Hamm B, Denecke T. Imaging-based liver function tests—past, present and future. Fortschr Röntgenstr. 2015;187:863–871.
  10. 10. Hanje AJ, Patel T. Nature Clinical Practice Gastroenterology and Hepatology. Preoperative evaluation of patients with liver disease. 2007;4:266–276.
  11. 11. Matos AP, Altun E, Ramalho M, Velloni F, Alobaidy M, Semelka RC. An overview of imaging techniques for liver metastases management. Expert Review of Gastroenterology and Hepatology. 2015;9:1561–1576.
  12. 12. D’Onofrio M, De Robertis R, Demozzi E, Crosara S, Canestrini S, Mucelli RP. Liver volumetry: is imaging reliable? Personal experience and review of the literature. World Journal of Radiology. 2014;6:62–71. doi:10.4329/wjr.v6.i4.62
  13. 13. Simoneau E, Alanazi R, Alshenaifi J, Molla N, Aljiffry M, Medkhali A, Boucher LM, Asselah J, Metrakos P, Hassanain M. Neoadjuvant chemotherapy does not impair liver regeneration following hepatectomy or portal vein embolisation for colorectal liver metastases. Journal of Surgical Oncology. 2016;113:449–455.
  14. 14. Lamade W, Glombitza G, Fischer L. The impact of 3-dimensional reconstruction on operative planning in liver surgery. Archives of Surgery. 2000;135:1256–1261.
  15. 15. Ribero D, Chun YS, Vauthey JN. Surgery for colorectal metastases. In: Vauthey JN, Hoff PMG, Audisio RA, Poston GJ, Editors. Liver Metastases. 1st ed. Springer. London; 2009. p25–38.
  16. 16. Mattar RE, Al-Alem F, Simoneau E, Hassanain M. Preoperative selection of patients with colorectal cancer liver metastasis for hepatic resection. World Journal of Gastroenterology. 2016;22:567–581. doi:10.3748/wjg.v22.i2.567
  17. 17. Ethun C, Maithel S. Determination of resectability. Surgical Clinics of North America. 2016;96:163–181. doi:10.1016/j.suc.2015.12.002
  18. 18. Brudvik KW, Passot G, Vauthey JN. Colorectal liver metastases: a changing treatment landscape. Journal of Oncology Practice. 2016;12:40–41.
  19. 19. Maor Y, Malnick S. Liver injury by anticancer chemotherapy and radiation therapy. International Journal of Hepatology. 2013; Article ID 815105, 8 pages. Published online 2013, July 17. Doi:10.1155/2013/815105
  20. 20. Nordlinger B, Vauthey JN, Poston G, Benoist S, Rougier P, Van Custem E. The timing of chemotherapy and surgery for the treatment of colorectal liver metastases. Clinical Colorectal Cancer. 2010;9:212–218.
  21. 21. Orcutt ST, Kobayashi K, Sultenfuss M, Hailey BS, Sparks A, Satpathy B, Anaya DA. Portal vein embolization as an oncosurgical strategy prior to major hepatic resection: anatomic, surgical, and technical considerations. Frontiers in Surgery. 2016;3:14. Published online 2016, March 11. doi:10.3389/fsurg.2016.00014
  22. 22. May BJ, Madoff DC. Portal vein embolization: rationale, technique, and current application. Seminars in Interventional Radiology. 2012;29:81–89.
  23. 23. Leung U, Simpson AL, Arajuo RLC, Gönen M, McAuliffe C, Miga MI, Parada EP, Allen PJ, D’Angelica MI, Kingham TP, DeMatteo RP, Fong Y, Jarnagin WR. Remnant growth rate after portal vein embolization is a good early predictor of post-hepatectomy liver failure. Journal of the American College of Surgeons. 2014;219:620–630. doi:10.1016/j.jamcollsurg.2014.04.022
  24. 24. Loffroy R, Favelier S, Chevallier O, Estivalet L, Genson PY, Pottecher P, Gehin S, Krausé D, Cercueil JP. Preoperative portal vein embolization in liver cancer: indications, techniques and outcomes. Quantitative Imaging in Medicine and Surgery. 2015;5:730–739. doi:10.3978/j.issn.2223-4292.2015.10.04
  25. 25. Chapelle T, Op De Beeck B, Huyghe I, Driessen A, Roeyen G, Ysebaert D, De Greef K. Future remnant liver function estimated by combining liver volumetry on magnetic resonance imaging with total liver function on (99m)Tc-mebrofenin hepatobiliary scintigraphy: can this tool predict post-hepatectomy liver failure? HPB. 2016;18:494–503.
  26. 26. Al-Sharif E, Simoneau E, Hassanain M. Portal vein embolization effect on colorectal cancer liver metastasis progression: lessons learned. World Journal of Clinical Oncology. 2015;6:142–146. doi:10.5306/wjco.v6.i5.142
  27. 27. Simoneau E, Hassanain M, Shaheen M, Aljiffry M, Molla N, Chaudhury P, Anil S, Khashper A, Valenti D, Metrakos P. Portal vein embolization and its effect on tumour progression of colorectal cancer liver metastases. British Journal of Surgery. 2015;102:1240–1249.
  28. 28. Pandanaboyana S, Bell R, Hidalgo E, Toogood G, Prasad KJ, Bartlett A, Lodge JP. A systematic review and meta-analysis of portal vein ligation versus portal vein embolization for elective liver resection. Surgery. 2015;157:690–698. doi:10.1016/j.surg.2014.12.009
  29. 29. Li J, Ewald F, Gulati A, Nashan B. Associating liver partition and portal vein ligation for staged hepatectomy: from technical evolution to oncological benefit. World Journal of Gastrointestinal Surgery. 2016;8:124–133. doi:10.4240/wjgs.v8.i2.12
  30. 30. Vennarecci G, Grazi GL, Sperduti I, Rizzi EB, Felli E, Antonini M, D’Offizi, Ettorre GM. ALPPS for primary and secondary liver tumors. International Journal of Surgery. 2016;30:38–44.
  31. 31. Schadde E, Ardiles V, Slankamenac K, Tschuor C, Sergeant G, Amacker N, Baumgart J, Croome K, Hernandez-Alejandro R, Lang H, de Santibanes E, Clavien PA. ALPPS offers a better chance of complete resection in patients with primarily unresectable liver tumors compared with conventional-staged hepatectomies: results of a multicenter analysis. World Journal of Surgery. 2014;38:1510–1519. doi:10.1007/s00268-014-2513-3
  32. 32. Buac S, Schadde E, Schnitzbauer AA, Vogt K, Hernandez-Alejandro R. The many faces of ALPPS: surgical indications and techniques among surgeons collaborating in the international registry. HPB. 2016;18:442–448.
  33. 33. Lau L, Christophi C, Nikfarjam M, Starkey G, Goodwin M, Weinberg L, Ho Loretta Muralidharan V. Assessment of liver remnant using ICG clearance intraoperatively during vascular exclusion: early experience with the ALIIVE technique. HPB Surgery. 2015;757052. doi:10.1155/2015/757052
  34. 34. Riemsma RP, Bala MM, Wolff R, Kleijnen J. Transarterial (chemo)embolisation versus no intervention or placebo intervention for liver metastases. Cochrane Database of Systematic Reviews.2013;http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD009498.pub3/abstract. Accessed online Dec 2, 2016
  35. 35. Yokoyama Y, Nagino M. Sequential PVE and TAE for biliary tract cancer and liver metastases. In: Madoff DC, Makuuchi M, Mizuno T, Vauthey JN, editors, Venous Embolization of the Liver. Springer-Verlag London 2011. p241–248.
  36. 36. Teo JY, Allen JC, Ng DC, Choo SP, Tai DWM, Chang JPE, Cheah FK, Chow PKH, Goh BKP. A systematic review of contralateral liver lobe hypertrophy after unilobar selective internal radiation therapy with Y90. HPB. 2016;18:7–12. doi:10.1016/j.hpb.2015.07.002
  37. 37. Bester L, Meteling B, Boshell D, Chua TC, Morris DL. Transarterial chemoembolization and radioembolisation for the treatment of primary liver cancer and secondary liver cancer: a review of the literature. Journal of Medical Imaging and Radiation Oncology. 2014;58:341–352.
  38. 38. Dupre A, Hitier M, Peyrat P, Chen Y, Meeus P, Rivoire M. Associating portal embolization and artery ligation to induce rapid liver regeneration in staged hepatectomy. The British Journal of Surgery. 2015;102:1541–1550. doi:10.1002/bjs.9900
  39. 39. Cha C. Assessment of hepatic function. In: Jamagin W, Blumgart L editors, Blumgart’s Surgery of the Liver, Biliary Tract and Pancreas 5th ed. Saunders Philadelphia 2012; p58–64.
  40. 40. Hwang S, Ha TY, Song GW, Jung DH, Ahn CS, Moon DB, Kim KH, Lee YJ, Lee SG. Quantified risk assessment for major hepatectomy via the indocyanine green clearance rate and liver volumetry combined with standard liver volume. Journal of Gastrointestinal Surgery. 2015;19:1305–1314.
  41. 41. De Graaf W, van Lienden KP, van Gulik TM, Bennink RJ. 99mTc-mebrofenin hepatobiliary scintigraphy with SPECT for the assessment of hepatic function and liver functional volume before partial hepatectomy. Journal of Nuclear Medicine. 2010;51:229–236.
  42. 42. Goéré D, Farges O, Leporrier J, Sauvanet A, Vilgrain V, Belghiti J. Chemotherapy does not impair hypertrophy of the left liver after right portal vein obstruction. Journal of Gastrointestinal Surgery. 2006;10:365–370.
  43. 43. Ribero D, Abdalla EK, Madoff DC, Donaldon M, Loyer EM, Vauthey JN. Portal vein embolization before major hepatectomy and its effects on regeneration, resectability and outcome. British Journal of Surgery. 2007;94:1386–1394.
  44. 44. Gruenberger B, Tamandl D, Schueller J, Scheithauer W, Zielinski C, Herbst F, Gruenberger T. Bevacizumab, capecitabine, and oxaliplatin as neoadjuvant therapy for patients with potentially curable metastatic colorectal cancer. Journal of Clinical Oncology. 2008;26:1830–1835.
  45. 45. Covey AM, Brown KT, Jarnagin WR, Brody LA, Schwartz L, Tuorto S, Sofocleous CT, D’Angelica M, Getrajdman GI, DeMatteo R, Kemeny NE, Fong Y. Combined portal vein embolization and neoadjuvant chemotherapy as a treatment strategy for resectable hepatic colorectal metastases. Annals of Surgery. 2008;247:451–455.
  46. 46. Aussilhou B, Dokmak S, Faivre S, Paradis V, Vilgrain V, Belghiti J. Preoperative liver hypertrophy induced by portal flow occlusion before major hepatic resection for colorectal metastases can be impaired by bevacizumab. Annals of Surgical Oncology. 2009;16:1553–1559.
  47. 47. Tanaka K, Kumamoto T, Matsuyama R, Takeda K, Nagano Y, Endo I. Influence of chemotherapy on liver regeneration induced by portal vein embolization or first hepatectomy of a staged procedure for colorectal liver metastases. Journal of Gastrointestinal Surgery. 2010;14:359–368.
  48. 48. Sturesson C, Keussen I, Tranberg KG. Prolonged chemotherapy impairs liver regeneration after portal vein occlusion—an audit of 26 patients. European Journal of Surgical Oncology. 2010;36:358–364.
  49. 49. Sturesson C, Nilsson J, Eriksson S, Spelt L, Andersson R. Limiting factors for liver regeneration after a major hepatic resection for colorectal cancer metastases. HPB (Oxford). 2013;15:646–652.
  50. 50. Beal IK, Anthony S, Papadopoulou A, Hutchins R, Fusai G, Begent R, Davies N, Tibballs J, Davidson B. Portal vein embolisation prior to hepatic resection for colorectal liver metastases and the effects of periprocedure chemotherapy. The British Journal of Radiology. 2006;79:473–478.
  51. 51. Dello SAWG, Kele PGS, Porte RJ, van Dam RM, Klaase JM, Verhoef C, van Gulik T, Molenaar Q, Bosscha K, van der Jagt EJ, Dejong CHC, de Boer MT. Influence of preoperative chemotherapy on CT volumetric liver regeneration following right hemihepatectomy. World Journal of Surgery. 2014;38:497–504.
  52. 52. Fischer C, Melstrom LG, Arnaoutakis D, Jarnagin W, Brown K, D’Angelica M, Covey A, DeMatteo R, Allen P, Kingham TP, Tuorto S, Kemeny N, Fong Y. Chemotherapy after portal vein embolization to protect against tumor growth during liver hypertrophy before hepatectomy. JAMA Surgery. 2013;148:1103–1108.
  53. 53. Fujii Y, Shimada H, Endo I, Kamiyama M, Kamimukai N, Tanaka K, Kunisaki C, Sekido H, Togo S, Nagashima Y. Changes in clinicopathological findings after portal vein embolization. Hepatogastroenterology. 2000;47:1560–1563.
  54. 54. Kusaka K, Imamura H, Tomiya T, Makuuchi M. Factors affecting liver regeneration after right portal vein embolization. Hepatogastroenterology. 2004;51:532–535.
  55. 55. Kakizawa H, Toyota N, Arihiro K, Naito A, Fujimura Y, Hieda M, Hirai N, Tachikake T, Matsuura N, Murakami Y, Itamoto T, Ito K. Preoperative portal vein embolization with a mixture of gelatin sponge and iodized oil: efficacy and safety. Acta Radiologica. 2006;47:1022–1028.
  56. 56. Nanashima A, Sumida Y, Abo T, Nonaka T, Takeshita H, Hidaka S, Sawai T, Yasutake T, Sakamoto I, Nagayasu T. Clinical significance of portal vein embolization before right hepatectomy. Hepatogastroenterology. 2009;56:773–777.
  57. 57. Van den Esschert JW, de Graaf W, van Lienden KP, Busch OR, Heger M, van Delden OM, Gouma DJ, Bennink RJ, Laméris JS, van Gulik TM. Volumetric and functional recovery of the remnant liver after major liver resection with prior portal vein embolization. Journal of Gastrointestinal Surgery. 2009;13:1464–1469.
  58. 58. Libicher M, Herbrik M, Stippel D, Poggenborg J, Bovenschulte H, Schwabe H. Portal vein embolization using the amplatz vascular plug II: preliminary results. Rofo. 2010;182:501–506.
  59. 59. de Baere T, Teriitehau C, Deschamps F, Catherine L, Rao P, Hakime A, Auperin A, Goere D, Elias D, Hechelhammer L. Predictive factors for hypertrophy of the future remnant liver after selective portal vein embolization. Annals of Surgical Oncology. 2010;17:2081–2089.
  60. 60. Giraudo G, Greget M, Oussoultzoglou E, Rosso E, Bachellier P, Jaeck D. Preoperative contralateral portal vein embolization before major hepatic resection is a safe and efficient procedure: a large single institution experience. Surgery. 2008;143:476–482.
  61. 61. Elias D, Ouellet JF, De Baère T, Lasser P, Roche A. Preoperative selective portal vein embolization before hepatectomy for liver metastases: long-term results and impact on survival. Surgery. 2002;131:294–299.
  62. 62. Broering DC, Hillert C, Krupski G, Fischer L, Mueller L, Achilles EG, Schulte am Esch J, Rogiers X. Portal vein embolization vs. portal vein ligation for induction of hypertrophy of the future liver remnant. Journal of Gastrointestinal Surgery. 2002;6:905–913; discussion 913.
  63. 63. Nagino M, Kamiya J, Nishio H, Ebata T, Arai T, Nimura Y. Two hundred forty consecutive portal vein embolizations before extended hepatectomy for biliary cancer: surgical outcome and long-term follow-up. Annals of Surgery. 2006;243:364–372.
  64. 64. Liem MS, Liu CL, Tso WK, Lo CM, Fan ST, Wong J. Portal vein embolisation prior to extended right-sided hepatic resection. Hong Kong Medical Journal. 2005;11:366–372.
  65. 65. Schnitzbauer AA, Lang SA, Goessmann H, Nadalin S, Baumgart J, Farkas SA, Fichtner-Feigl S, Lorf T, Goralcyk A, Hörbelt R, Kroemer A, Loss M, Rümmele P, Scherer MN, Padberg W, Königsrainer A, Lang H, Obed A, Schlitt HJ. Right portal vein ligation combined with in situ splitting induces rapid left lateral liver lobe hypertrophy enabling two-staged extended right hepatic resection in small-for-size settings. Annals of Surgery. 2012;255:405–414.
  66. 66. Alvarez FA, Ardiles V, Sanchez Claria R, Pekolj J, de Santibañes E. Associating liver partition and portal vein ligation for staged hepatectomy (ALPPS): tips and tricks. Journal of Gastrointestinal Surgery. 2013;17:814–821.
  67. 67. Li J, Girotti P, Königsrainer I, Ladurner R, Königsrainer A, Nadalin S. ALPPS in right trisectionectomy: a safe procedure to avoid postoperative liver failure? Journal of Gastrointestinal Surgery. 2013;17:956–961.
  68. 68. Dokmak S, Belghiti J. Which limits to the “ALPPS” approach? Annals of Surgery. 2012;256:e6. (e16–17).
  69. 69. Machado MA, Makdissi FF, Surjan RC. Totally laparoscopic ALPPS is feasible and may be worthwhile. Annals of Surgery. 2012;256:e13. (e16–19) doi:10.1097/SLA.Ob013e318265ff2e
  70. 70. Donati M, Stavrou GA, Basile F, Gruttadauria S, Niehaus KJ, Oldhafer KJ. Combination of in situ split and portal ligation: lights and shadows of a new surgical procedure. Annals of Surgery. 2012;256:e11–12. (e16–9).
  71. 71. Andriani OC. Long-term results with associating liver partition and portal vein ligation for staged hepatectomy (ALPPS). Annals of Surgery. 2012;256:e5 (e16–9).
  72. 72. Govil S. Rapid improvement in liver volume induced by portal vein ligation and staged hepatectomy: the ALPPS procedure. HPB (Oxford). 2012;14:874 Doi:10.1111/j.1477-2574.2012.00573.x.
  73. 73. Oldhafer KJ, Donati M, Maghsoudi T, Ojdanić D, Stavrou GA. Integration of 3D volumetry, portal vein transection and in situ split procedure: a new surgical strategy for inoperable liver metastasis. Journal of Gastrointestinal Surgery. 2012;16:415–416.
  74. 74. Conrad C, Shivathirthan N, Camerlo A, Strauss C, Gayet B. Laparoscopic portal vein ligation with in situ liver split for failed portal vein embolization. Annals of Surgery. 2012;256:e14–5 (e16–7).
  75. 75. Cavaness KM, Doyle MB, Lin Y, Maynard E, Chapman WC. Using ALPPS to induce rapid liver hypertrophy in a patient with hepatic fibrosis and portal vein thrombosis. Journal of Gastrointestinal Surgery. 2013;17:207–212.
  76. 76. Alvarez FA, Ardiles V, de Santibanes E. The ALPPS approach for the management of colorectal carcinoma liver metastases. Current Colorectal Cancer Reports. 2013. Doi:10.1007/s11888-013-0159-4

Written By

Mandivavarira Maundura and Jonathan B Koea

Submitted: 12 April 2016 Reviewed: 04 October 2016 Published: 05 April 2017