You are on page 1of 20

Current Oncology Reports (2020) 22: 59

https://doi.org/10.1007/s11912-020-00922-x

INTERVENTIONAL ONCOLOGY (DC MADOFF, SECTION EDITOR)

Improving the Safety of Major Resection for Hepatobiliary


Malignancy: Portal Vein Embolization and Recent Innovations in Liver
Regeneration Strategies
David C. Madoff 1 & Bruno C. Odisio 2 & Erik Schadde 3,4,5 & Ron C. Gaba 6 & Roelof J. Bennink 7 & Thomas M. van Gulik 8 &
Boris Guiu 9

Published online: 16 May 2020


# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Purpose of review For three decades, portal vein embolization (PVE) has been the “gold-standard” strategy to hypertrophy the
anticipated future liver remnant (FLR) in advance of major hepatectomy. During this time, CT volumetry was the most common
method to preoperatively assess FLR quality and function and used to determine which patients are appropriate surgical
candidates. This review provides the most up-to-date methods for preoperatively assessing the anticipated FLR and summarizes
data from the currently available strategies used to induce FLR hypertrophy before surgery for hepatobiliary malignancy.
Recent findings Functional and physiological imaging is increasingly replacing standard CT volumetry as the method of choice
for preoperative FLR assessment. PVE, associating liver partition and portal vein ligation, radiation lobectomy, and liver venous
deprivation are all currently available techniques to hypertrophy the FLR. Each strategy has pros and cons based on tumor type,
extent of resection, presence or absence of underlying liver disease, age, performance status, complication rates, and other factors.
Summary Numerous strategies can lead to FLR hypertrophy and improve the safety of major hepatectomy. Which is best has yet
to be determined.

Keywords Hepatic regeneration . Future liver remnant . Portal vein embolization . ALPPS . Liver venous deprivation . Radiation
lobectomy

Introduction resected if indicated. However, major hepatic resection places


patients at risk for developing complications related to liver
Hepatic resection remains a potentially curative treatment op- insufficiency, particularly in the perioperative period, before
tion for many types of hepatobiliary malignancy. Owing to its the liver regeneration occurs [1, 2]. Although the causes of
unique ability to regenerate, a large part of the liver can be postoperative liver insufficiency are multifactorial, the volume

This article is part of the Topical Collection on Interventional Oncology

* David C. Madoff 5
Institute of Physiology, Center for Integrative Human Physiology,
david.madoff@yale.edu University of Zurich, Zurich, Switzerland

6
1
Department of Radiology, Interventional Radiology Section,
Department of Radiology and Biomedical Imaging, Section of University of Illinois Hospital, Chicago, IL, USA
Interventional Radiology, Yale School of Medicine, New Haven, CT,
USA 7
Department of Radiology and Nuclear Medicine, Amsterdam UMC,
2 University of Amsterdam, Amsterdam, The Netherlands
Department of Interventional Radiology, The University of Texas
MD Anderson Cancer Center, Houston, TX, USA
8
3 Department of Surgery, Amsterdam UMC, University of
Department of Surgery, Rush University Medical Center, Amsterdam, Amsterdam, The Netherlands
Chicago, IL, USA
4 9
Department of Surgery, Cantonal Hospital Winterthur, Department of Radiology, St-Eloi University Hospital-Montpellier,
Zurich, Switzerland Montpellier, France
59 Page 2 of 20 Curr Oncol Rep (2020) 22: 59

of the anticipated future liver remnant (FLR) (i.e., volume of from regenerative liver surgery prior to extensive resection, as
liver that remains after surgery) has been shown to be a strong well as to decide at which point sufficient functional increase
independent predictor of postoperative complications [3, 4]. has been achieved in the follow-up of the augmentation
Over the past three decades, various strategies have been de- procedure.
veloped to increase the volume and function of the anticipated
FLR in advance of the definitive resection. The original tech-
nique, portal vein embolization (PVE), was first described by Volumetric Assessment Based on Imaging Studies
Makuuchi et al. in 1990 [5] to induce left liver hypertrophy
before right hepatectomy in thirteen patients with hilar chol- Gold standard in the assessment of FLR volume is computed
angiocarcinoma. Since publication of this inaugural paper, tomography (CT)–based volumetry by which whole liver vol-
PVE has become the standard of care worldwide for preparing ume, tumor volume, and FLR volume are calculated [3]. An
patients for major hepatic resection with an inadequate FLR FLR volume of 20–30% has been considered sufficient in
[6]. Its early adoption was based on data showing that it re- patients without underlying parenchymal disease; however,
sulted in the reduction of perioperative complications (includ- in patients with compromised liver (e.g., cirrhosis, bile duct
ing liver failure and death) and an extension of resectability for obstruction), the requirement has been increased to at least
patients initially considered unresectable due to insufficient 40% [4, 16]. For regenerative liver interventions, CT
anticipated FLR volume [2, 3, 7]. volumetry is sequentially applied to monitor volume increase
Despite the success of PVE as a preoperative strategy, it until the target volume has been attained. However, CT
is not without limitations. These limitations include (a) volumetry is error-sensitive due to varying size and number
insufficient FLR hypertrophy in some patients, especially of tumors, the presence of dilated bile ducts, variable vascu-
those with hepatocellular carcinoma in the setting of cir- larity, and quality of the liver parenchyma. Although CT
rhosis [8], (b) the lack of treatment to the tumor with the volumetry remains widely used, its role has been reconsidered
potential for tumor growth during the waiting period be- [17]. To better predict postoperative outcomes, especially in
tween PVE and surgery [9–11], and (c) the need for the setting of liver regeneration inducing modalities, combi-
assessing tumor biology using a “test of time” approach, nation of CT volumetry with an additional, quantitative liver
which may be viewed to either delay definitive treatment function test is now recommended by many expert centers.
or negate the performance of futile surgery [12]. To address To overcome some shortcomings of CT volumetric assess-
these concerns, newer preoperative treatment strategies ment of the FLR, a modified method of total liver volume
have recently been advocated, including associating liver estimation was proposed, based on Western patients’ character-
partition and portal vein ligation (ALPPS), radiation lobec- istics and body surface area (BSA): estimated total liver volume
tomy (RL), and liver venous deprivation (LVD) [13–15]. (eTLV) [cc] = −794.41 + 1267.28 x BSA [18] (Figure 1). This
This article will review how to assess the FLR in advance formula has been compared with other formulas and validated
of surgery and each of the currently available methods for in several reports [19, 20]. The ratio of FLR volume measured
inducing selective hepatic hypertrophy with particular at- by CT volumetry and estimated TLV is referred to as the stan-
tention to rationale, patient selection/indications, contrain- dardized FLR (sFLR) volume. It is presumed to better predict
dications, technique, available data supporting or criticiz- liver function than measured FLR volume in %, although no
ing its use, potential complications, and future directions of empiric proof has been presented. Of, course, discrepancies
the field of regenerative liver surgery. between measured and estimated volumetric results have been
reported [17]. In an attempt to measure overall FLR volume
growth before and after regenerative liver interventions, the
Preoperative Assessment of the FLR degree of hypertrophy (DH) (i.e., FLR-post–FLR-pre) is used.
Kinetic growth rate (KGR) is a metric for FLR growth over
Application of liver enhancing techniques largely rely on an time and is calculated by dividing the DH by the number of
adequate assessment of the regenerative response of the FLR. weeks after the intervention. KGR may be better predictor of
Volumetric studies based on CT or MRI imaging are common- perioperative outcomes after extended resections than FLR size
ly used to evaluate the preoperative anticipated liver remnant in % or sFLR [21].
as well as to determine if sufficient increase of the FLR oc- As an alternative, liver-to-body-weight-ratio (LBWR) has
curred in order to determine if a safe resection can be under- also been proposed to assess the FLR, consisting of the ratio of
taken. However, volumetric assessment of the whole liver or FRL-volume measured by CT volumetry and body weight
part of it only provides indirect information on the actual [22], expressed in %. Patients with healthy liver and a
functional capacity of the liver, while underlying parenchymal LBWR < 0.5% as well as cirrhotic patients with LBWR <
disease remains unknown. Direct assessment of liver function 1.4% [23] have been shown to be at risk for post-
is therefore crucial to select those patients who will benefit hepatectomy liver failure.
Curr Oncol Rep (2020) 22: 59 Page 3 of 20 59

b c d

Fig. 1 Assessment of liver hypertrophy following right and segment 4 remnant (FLR), namely segments I, II, III provided an absolute FLR
portal vein embolization using the estimated the total liver volume volume of 276.1 cc, which represents 20.3% of the eTLV; c Post-PVE
(eTLV) formula based on the body surface area (BSA) on a 62-year-old showing adequate embolization of the right portal and segment IV
female patient with colorectal liver metastasis planned to undergo a right branches; d Post-PVE computed-tomography based volumes of the
extended hepatectomy (right hepatic lobe + segment IV resection). a future liver remnant (FLR) performed 4 weeks after the PVE showing
eTLV on a patient with a BSA of 1.7 resulted in a eTLV of 1359.96 cc; increase of the absolute FLR volume to 430.8 cc, which represents 31.6%
b Pre-PVE computed-tomography based volumes of the future liver of the eTLV. Patient underwent an uneventful right extended hepatectomy

Functional Assessment Based on Dynamic Studies Scintigraphic Assessment of Liver Function

Quantitative liver function tests are based on the capacity Nuclear imaging studies are increasingly applied in protocols
of the liver to clear a substance that is mostly or exclu- to evaluate liver functional increase after induction of liver
sively metabolized by the liver. The indocyanine green regeneration [27]. Scintigraphy provides simultaneous mor-
(ICG) clearance test is worldwide the most commonly phologic (visual) and physiologic (quantitative functional) in-
used quantitative liver function test in liver surgery and formation of the liver, especially when SPECT-CT cameras
was initially introduced as a modality for the measure- are used. Regional (segmental) differentiation allows func-
ment of blood flow [24]. ICG is removed from the blood tional assessment of specifically, the FLR. Technetium-99 m
exclusively by the liver and excreted into the bile with- (99mTc)-galactosyl serum albumin (GSA) scintigraphy and
99m
out intrahepatic conjugation. The ICG elimination rate or Tc-mebrofenin hepatobiliary scintigraphy (HBS) have
the percentage of retained ICG of all ICG given at been used to identify patients potentially at risk for PRLF.
15 min after administration (ICG-R15) are used to ex- The use of 99m Tc-GSA is limited in that this radio-
press the results of this test. The ICG clearance test is a pharmaceutical agent has not been approved by regulatory
global test that represents function of the whole liver. agencies in several countries, while 99mTc-mebrofenin is. An
Function of specifically the FLR can be calculated by increasing number of centers worldwide are using 99mTc-
multiplying ICG-R15 by the proportion of the FRL vol- mebrofenin HBS with SPECT-CT to assess function of the
ume as determined by CT volumetry. This has been FLR in both assessment and monitoring of FLR function in
shown to be predictive of post-hepatectomy liver failure regenerative liver surgery [28–30]. 99mTc-mebrofenin is an
(PRLF). However, the results rest on the assumption that HIDA (hepatobiliary iminodiacetic acid) agent most suitable
liver function is homogeneously distributed [25]. For this for functional assessment because of its high hepatic uptake,
reason, the ICG clearance test is not very useful to assess low displacement by bilirubin, and low urinary excretion.
the increase in FLR function after liver enhancement as Camera-based measurement of the hepatic 99mTc-mebrofenin
an increase in function of the regenerated lobe is accom- uptake ratio allows segmental definition of function. The up-
panied by a decreased function of the atrophied lobe take ratio is divided by the body surface area (BSA) and
resulting in no net effect on total liver function [26]. expressed as %/min/m2 in order to compensate for differences
59 Page 4 of 20 Curr Oncol Rep (2020) 22: 59

in individual metabolic requirements based on body size. correlate with function, interstage CT volumetry may not re-
Using a cut-off value of 2.7/min/m2, the test enables identifi- liably predict FRL function after ALPPS stage 1. A caveat of
cation of patients who are at increased risk of liver failure after the use of HBS is the decreased uptake of 99mTc-mebrofenin
major resection irrespective of the quality of liver parenchyma in the presence of cholestasis due to competitive uptake of
[31•]. bilirubin and mebrofenin by the same cellular transporter sys-
tems, reflecting a true but mostly reversible diminished liver
Scintigraphic Assessment of Changes in Liver function possibly at the segmental level. Especially in patients
Function with bile duct tumors requiring extensive liver resection, com-
plete decompression of the (FLR) biliary system with a reduc-
Using scintigraphic techniques, it was found that after PVE, tion of hypoerbilirubinemia justifies subsequent use of HBS
the increase in FLR function exceeded the increase in FLR [36].
volume [32–34] suggesting that the time interval between
PVE and liver resection may be shorter than determined on Novel Techniques for Functional Assessment
the basis of volumetric parameters (Fig. 2). A novel interesting
application of HBS may be the selection of most appropriate LiMAx is a commercially available carbon-13 ( 13 C)-
regenerative procedure, PVE, ALPPS, or LVD in patient with methacetin breath test offered as a non-invasive method for
low FLR volume. FLR-function assessed with HBS may be the assessment of global liver function in surgical patients.
used as a predictor of future insufficient functional hypertro- Regional function of the liver, similar to ICG, can only be
phy after PVE, selecting patients who may be candidates for calculated as a function of CT-based volume and the test re-
upfront ALPPS or LVD [35]. Sequential real time assessment sult. In patients undergoing PVE [37], this test was used to
of function may allow choosing the earliest time point for visualize the changes in FLR function after PVE, showing an
resection. As laid out in the ALPPS section, the timing of increase in FLR function albeit that function of the FLR post-
stage 2 resection in ALPPS, the removal of the deportalized resection was lower in comparison with the preoperatively
liver lobe appears to be important to avoid perioperative mor- calculated function. Similarly to the ICG clearance test, the
bidity. As volume of regenerating liver tissue does not major limitation of the 13C-breath tests is that read-outs

Fig. 2 A 66-year-old man with a


large hepatocellular carcinoma in
the right liver and segment 4 (a)
on CT requiring extended right-
sided hemihepatectomy. FLR-
volume (segments 2–4) was 32%
(b). FRL-function was 2.9%/min/
m2 (35% of total liver function)
and considered sufficient (c). 43-
year-old-female patient with
multiple liver adenomas on the
right side (d, e) requiring a right-
sided hemihepatectomy. FLR-
volume was 27%. FLR-function
(f) was 2.3%/min/m2 (26% of
total liver function) and
considered insufficient.
Hepatobiliary scintigraphy
3 weeks after right PVE (g)
showed increase of FLR-volume
to 47% (h) and FLR-function to
3.6%/min/m2 (58% of total liver
function) (i) considered sufficient
for safe resection
Curr Oncol Rep (2020) 22: 59 Page 5 of 20 59

represent total liver function, whereas after liver enhancement, For cirrhotic patients, a sFLR of < 40% of the TLV should
the distribution of function in the regenerated FLR and the warrant the need of PVE [6, 44]. Not offering preoperative
atrophic parts of the liver greatly differ. To determine function PVE for patients with FLR volumes below such thresholds
of the FLR according to the share of FLR as part of total liver carries a significant risk of post-operative liver insufficiency
volume, therefore, seems less reliable. and should be discouraged. On the other hand, offering PVE
Contrast-enhanced MRI is part of the standard preoperative in patients with FLR above these thresholds may be unneces-
work-up in patients with liver tumors considered for resection sary overuse of PVE [45, 46].
and has recently also been introduced as a potential technique The only absolute contraindication to PVE is overt clinical
for preoperative assessment of liver function [38, 39]. portal hypertension, which itself contraindicates major hepatic
Functional imaging with MRI-Gd-EOB-DTPA facilitates as- resection. Relative contraindications include portal vein tumor
sessment of total and regional liver function in a similar way invasion, uncorrectable coagulopathy, tumor precluding safe
as scintigraphic methods. An additional feature of MRI is transhepatic access, non-corrected biliary dilation and chole-
regional assessment of parenchymal steatosis or fibrosis, stasis, and renal failure. Bilobar tumor extension is no longer
which potentially supports functional findings [40]. considered a contraindication to PVE since aggressive local
Application of MRI as a “one-stop-shop” modality for both approaches such as two-stage hepatectomy [47, 48] and com-
diagnostic and functional assessment of the FLR in patients bined resection and thermal ablation [49, 50] allow curative
undergoing liver enhancing techniques is promising but still resection.
faces substantial technical challenges concerning the amount
of tracer needed for visualization and quantification of func- Technique
tion [40].
The two most commonly utilized percutaneous access routes
to the portal venous system are the ipsilateral and contralateral
Techniques Used for Preoperative Liver approaches. Given the similar safety profile for these two ac-
Regeneration cesses [4, 51, 52], its choice is mainly dictated by operator’s
experience and preference.
Portal Vein Embolization In the ipsilateral approach [53], the portal vein is accessed
via the tumor-bearing liver. Its main advantage is to avoid
Rationale puncture and excessive instrumentation of the FLR. When
performing this approach, the risk of puncturing through tu-
Portal vein embolization (PVE) promotes proliferation of the mor tissue and, consequently, promote tumor seeding should
surviving hepatocytes with consequent hypertrophy and func- be recognized [6]. Also, care should be taken during catheter
tional shift towards the non-embolized liver segments. manipulation to avoid dislodging the surrounding embolic
Although not completely elucidated at the present time, this material. Finally, when right PVE is combined with segment
pathophysiological process is thought to be triggered by 4 embolization, segment 4 should be embolized first in order
injury-induced growth-factor stimulation, which occurs once to avoid potential embolic material dislodgment from the right
> 10% of the liver is injured [41, 42]. Preoperative PVE is portal vein to the left portal vein and its branches [6, 52].
considered a major advance on modern hepatobiliary surgery Additionally, having the right portal vein patent would main-
as it allows curative resection in patients with limited antici- tain portal venous flow to the liver in the event of inadvertent
pated future liver volume while providing reduced rates of left portal vein thrombosis during segment 4 embolization.
postoperative liver failure [43]. In the contralateral approach [54], care should be taken to
limit the number of punctures in the FLR. For this reason, this
Patient Selection access requires ultrasonography guidance. The main advan-
tage of this approach is that it allows for straight catheteriza-
In addition to surgical candidacy evaluation, factors associated tion of the right portal vein branches, obviating the potential
with poor liver regeneration should also be thoroughly need of using a reverse curve catheter. In fact, segment 4
assessed and, whenever possible, addressed before branch catheterization can be challenging when using a con-
performing PVE. Those include: presence of underlying he- tralateral approach via the left portal vein. To overcome this
patic disease, chronic ethanol consumption, malnutrition, bil- issue, an adequate distance should exist between the segment
iary obstruction, diabetes, and infection. There is broad con- 4 branches and left portal entry point. This can be achieved by
sensus that patients without any underlying liver disease obtaining percutaneous access within a peripheral segment 3
should undergo PVE if the sFLR < 20% of the TLV. For pa- portal vein branch [44]. The main drawback of the contralat-
tients with hepatic fibrosis or post-chemotherapy hepatic inju- eral approach is the instrumentation of the FLR parenchyma,
ry, PVE should be performed if the sFLR < 30% of the TLV. which could potentially make it more prone to complications
59 Page 6 of 20 Curr Oncol Rep (2020) 22: 59

such as bleeding and thrombosis [55]. If this were to occur, Nagino et al. reported their experience with PVE in 494
surgical resection could be made more challenging or in the patients with primary biliary cancers [61]. PVE-related com-
worst-case scenario, impossible. plications requiring interventions occurred in 0.6% of the pa-
Several embolic materials have been used for PVE tients. Also, on a single-institution analysis of 358 patients,
[56–59]. To date, there are no large clinical studies comparing Shindoh et al. reported PVE complication rate of 3.9% [43],
the effects of different embolic materials in the hypertrophy which included portal vein thrombosis (2.2%), coil misplace-
response. Operator experience, cost, and availability dictate ment (0.8%), subcapsular hematoma (0.6%), and esophageal
the embolic agent of choice. N-butyl cyanoacrylate (NBCA), bleeding (0.3%).
a relatively low-cost embolic agent, provides fast and reliable
FLR hypertrophy in the hands of experienced operators. Future Directions
NBCA is thought to result in greater FLR growth when com-
pared with gelatin, sponge, fibrin glue, and PVA based on Currently, the true impact of PVE on tumor growth is unclear
recent systematic analysis [56]. Nevertheless, NBCA induces [11, 62, 63]. The sequential use of arterial and portal emboli-
a periportal inflammation that may make hepatectomy more zation relies on the local oncological effects of the transarterial
challenging. Absolute ethanol is another effective embolic embolization, which is critical to prevent tumor growth stim-
agent, but its use is associated with significant changes in ulation associated with the hepatic artery buffer response fol-
the hepatic function and poor patient tolerability due to pain. lowing PVE [64]. In patients with HCC, transarterial emboli-
The combination of spherical particles and coils is a safe and zation might also help to occlude arterio-portal shunts, which
effective embolic method, with the former promoting distal are thought to negatively impact the FLR growth [65]. Yoo
embolization, which limits the development of collateral cir- et al. described their experience on 135 patients with HCC
culation, while the coils are placed more proximal to reduce submitted to sequential TACE followed by PVE (n = 71 pa-
chances of recanalization. The high costs associated with this tients) or to PVE-only (n = 64). The TACE + PVE group,
combination is the main limitation of its use. when compared with the PVE-only group, had a significant
increase in the FLR (7.3% vs 5.8%, respectively; P = 0.035),
Supporting Data as well lower rates of hepatic failure following hepatectomy
(4% vs 12%, respectively; P = 0.185). The combination of
In a meta-analysis, Abulkhir et al. reported the outcomes of TACE and PVE was associated with improved overall (P =
1088 patients submitted to PVE using both percutaneous and 0.028) and recurrence-free (P = 0.001) survival rates on a
transilieocolic approaches. Procedures were successful in > comparison using log-rank test. On a recent systematic review
95% of the patients. The FLR volume increase was greater in on the use of sequential TACE and PVE on patients with
patients using a percutaneous approach (11.9%), compared HCC, Piardi et al. [66] included 171 patients from 4 selected
with those patients using the transileocolic approach (9.7%) publications. An increase on the FLR volume was noticed
[60]. A recent systematic review of 47 articles included at total when the sequential TACE + PVE approach was utilized when
of 1791 patients [56]. The technical success rate was 99.3%, compared with the use of PVE-only (12% vs 8%, respective-
the mean FLR hypertrophy rate was 37.9 ± 0.1%, and the ly). Mortality rates ranged from 0 to 11%, and a 5-year over-
clinical success rate was 96.1%. The results of the largest all- (43–72%) and recurrence-free (37–61%) survivals were
cohort (level 2b) series published to date are summarized in recorded. Further refinements in imaging perfusion, along
Table 1. with recent improvements on understanding of tumor biology,
provide opportunities to evaluate the effects of portal and ar-
Potential Complications terial blood flow modulation in both the functional liver and
tumor growth.
Van Lieden et al. reported the complications described in 29 The delivery of hematopoietic cells on the portal system
studies (n = 1179 patients) [56]. Minor complications were has been suggested to stimulate hepatic regeneration [67, 68].
mostly related to post-embolization syndrome such as fever The combination of stem cell administration with PVE has
(36.9%), transaminase elevation (34.8%), abdominal been investigated as a potential driver for augmenting and
pain/discomfort (22.9%), nausea and vomiting (1.25), and il- accelerating FLR growth process. Furst et al. [69] prospective-
eus (1.2%). Major complications were portal thrombosis ly evaluated the combination of PVE with bone marrow stem
(0.8%), embolization of non-target vessels (0.6%), liver he- cell (BMSCs) administration to the non-embolized liver seg-
matoma (0.4%), and infection/abscess (0.4%). Importantly, ments in six patients (BMSCs + PVE group). Compared with
complications led to non-resectability in 0.4% of the patients. 7 other patients in whom only PVE was performed, a signif-
Two patients died after PVE due to lethal pulmonary embo- icant increase in the FLR volume was noted in the group
lism (n = 1) and cholangitis/septic shock (n = 1), resulting in receiving the BMSCs (81.2% ± 34.3, BMSCs + PVE vs
an overall mortality rate of 0.1%. 39.8% ± 20.3 m, PVE alone; P = 0.021). No complications
Curr Oncol Rep (2020) 22: 59 Page 7 of 20 59

Table 1 Summary of the largest portal vein embolization series to date

Article No. Underlying Procedural approach Embolic material used % Complications Complications Surgical
of Pts pathology Increase after PVE after surgery mortality
FLR*

Ebata 494 Biliary Percut. ipsilateral Fibrine glue OR NA 0.6%¥ NA 6.5%


cancer Ethanol + coils
Shindoh 358 Mixed Percut. ipsilateral Particles + Coils 10.2 3.9% 25.8% 3.8%
Giraudo 146 Mixed Percut. contralateral Isobutyl-2-cyanoacrylate 47.7 10 43% NA
De Baere 107 Mixed Percut. contralateral NBCA 69 NA 26.6 NA
Nagino 240 Biliary Percut. ipsilateral Fibrine glue 27.4 No major NA 8.8%
cancer
Di Stefano 188 Mixed Percut. contralateral NBCA 41 and 62 12.8% NA NA
Sakuhara 143 Mixed Percut. Ipsilateral Ethanol 33.6 6% NA 1.6%
(preferable) or
contralateral

FLR future liver remnant, PVE portal vein embolization, Percut percutaneous, NA not available, NBCA n-butyl cyanoacrylate
*Different methods for calculating % FLR among different studies
¥
Complications requiring treatment

were recorded in regard to BMSCs, PVE, and BMSCs deliv- by transection of the liver parenchyma 7 to 10 days prior to
ery in the portal system. The daily hepatic growth rate in the resection [14]. ALPPS accelerated liver regeneration to the
BMSCs + PVE group was significantly higher than that in the point that increases of sFLR and LBWR were achieved
PVE-only group (9.5 mL/d ± 4.3 vs 4.1 mL/d ± 1.9, P = 0.03), allowing resection after a median of 9 days. To date, the liter-
as well a short interval from PVE procedure to surgery. Later, ature had not supported such a short waiting period between
the same group published the outcomes of 11 patients submit- induction of liver hypertrophy and resection. The authors ar-
ted to BMSCs + PVE [70]. Again, a significant higher growth gued that their technique “enables curative resection of mar-
of the FLR was noted on the BMSCs + PVE group compared ginally resectable liver tumors or metastases in patients that
with the PVE-only group (138.66 mL ± 66.29 vs 62.95 mL ± might otherwise be regarded as palliative” [14]. Their ratio-
40.03; P = 0.004). Post hoc analysis also revealed improved nale was to expand resectability of all liver tumors, specifical-
survival for the BMSCs + PVE group compared with the ly those where PVE had been considered due to the small
PVE-only group. Further understanding on the mechanisms degree of hypertrophy induced with long waiting time. As
associated with regeneration of the damaged liver following spelled out in an accompanying editorial, ALPPS was devel-
BMSCs delivery is needed to improve patient selection and oped to find a solution for difficult intra-operative situations
subsequent outcomes. and as a salvage maneuver, when PVE was unsuccessful [72].
Lastly, for patients undergoing the two-stage hepatectomy Soon after its initial description, ALPPS was used by many
approach [47], the advent of hybrid IR/OR angiography surgeons worldwide to accelerate the growth of the FLR for all
rooms allows to combine PVE and first-stage hepatectomy types of tumors requiring extended resections or two-stage
in the same surgical event [71]. After FLR hypertrophy is hepatectomies [73]. A liberal patient selection and the high
achieved, the second-stage hepatectomy is then performed. physiological severity of the operation impacted perioperative
This approach minimizes the number of hospital encounters, outcomes [74, 75]. In the initial publications, 90-day mortality
reduces the time between the first- and second-hepatectomy of ALPPS was found to be 9–15%, too high even for complex
stages, and allows for early return to intended oncologic and small-FLR-volume liver surgery [14, 76–78].
treatment.
Technical Aspects

ALPPS ALPPS had barely reached reproducibility and maturity in


2014, when modifications were proposed to remedy its high
Rationale mortality and morbidity [76, 79]. In the original or “classic”
ALPPS description, the parenchyma is divided along a plane
In 2012, a novel approach was presented to increase the liver that completely separates the FLR from part of the liver to be
remnant prior to extended hepatectomies by occluding the removed and the portal vein branch into the part to be re-
portal vein flow to the side of the liver to be removed but also moved is surgically ligated (Fig. 3) [80]. The first technical
59 Page 8 of 20 Curr Oncol Rep (2020) 22: 59

Fig. 3 Schematic of ALPPS


procedure. The ALPPS procedure
is a two-stage hepatectomy. In
stage 1, the FLR is “cleaned” of
lesions if necessary, the liver
parenchyma is transected, and the
portal vein is surgically ligated. In
stage 2, after a short waiting time
of only 10 days and about 80%
hypertrophy of the FLR, the
deportalized part of the liver is
removed

modifications proposed alterations in the parenchymal tran- Data Supporting and/or Negating Its Adoption
section using the ALTPS technique (associating liver tourni-
quet ligation and portal vein ligation for staged hepatectomy), Despite its high morbidity and mortality rates, ALPPS in-
where the parenchyma is tied with a tourniquet instead of creased resectability rates when compared with PVE in a ret-
being transected (Table 2). Nevertheless, this technical modi- rospective cohort study (level 2a evidence) [78] as well as in
fication did not reduce the high morbidity and mortality [81]. the randomized Scandinavian LIGRO trial of ALPPS in pa-
In RALPPS (Radiofrequency assisted liver partition with por- tients with colorectal liver metastases (level 1a evidence)
tal vein ligation for staged hepatectomy), a precise 1-cm avas- (Table 3) [90••]. From this trial, 92% of patients underwent
cular ablation area was created across the liver instead of sur- tumor resection in the ALPPS arm compared with 57% of
gical transection [82]. The results of a randomized trial of patients after PVE. The mortality in the LIGRO trial, however,
RALPPS against PVE have been published [83]. The next remained 8.3% for ALPPS. Despite the extensive attention
modification replaced the ligation of the portal vein with in- directed towards ALPPS in the surgical literature [91], its
terventional radiology (IR)–based embolization and was use in daily practice is currently very limited. Even the
called “hybrid” ALPPS [84]. Comparative data on hybrid ver- Scandinavian centers where the LIGRO randomized trial
sus classic ALPPS are not yet available. In another modifica- was performed, now use ALPPS only as salvage when other
tion, intraoperative embolization through a catheter threaded methods to have failed (pers. communication, Per Sandström,
through a mesenteric vessel under fluoroscopy is performed Linköping, Sweden). A recent update on survival of patients
after transection and was called “mini”-ALPPS with no cur- in the LIGRO trial showed that the median survival of patients
rent comparative data [85]. Further, the formal description of a treated with ALPPS was 46 months compared with 26 months
limited instead of a complete transection was named “partial for two-stage hepatectomy [92•].
ALPPS” [86]. Limited transection was likely performed by The largest analysis of ALPPS worldwide is the interna-
many early ALPPS adopters, because of the concern of injury tional ALPPS registry, started at the University of Zürich in
to the biliary radicals emanating from the portal plate and the 2012. The first registry analysis showed an overall periopera-
danger of injury to the vena cava during transection. It has to tive mortality of 9%, but a 90-day mortality in hepatocellular
be assumed that a fair percentage of early ALPPS cases were carcinoma (HCC), intrahepatic cholangiocarcinoma (IHCC),
actually performed as “partial” ALPPS cases. Retrospective and perihilar cholangiocarcinoma (PHCC) of 12%, 13%, and
comparative reports published on partial ALPPS argue for its 27% respectively (level 2a evidence) [76]. The analysis of
general adoption due to a reduced morbidity and mortality complications was equally worrisome with a major complica-
[87]. Another commonly used modification is to simply wait tion rate of 25%, 43%, and 60% for HCC, IHCC, and PHCC,
longer than 7–10 days to perform the second stage. There is respectively [76]. A randomized trial for all tumor types by the
some experimental evidence that ALPPS hypertrophy is im- Zurich group had to be stopped due to mortalities after only 8
mature at 7–10 days [88] and a longer waiting period may gain patients were enrolled (data not published). Subsequent re-
in functional maturity [89]. All the technical modifications, ports of selected large volume centers within the ALPPS reg-
except for the last, have in common the reduction of the sur- istry showed a trend to use ALPPS mostly in colorectal liver
gical stage 1. metastases and an improvement in 90-day mortality to 4%
Curr Oncol Rep (2020) 22: 59 Page 9 of 20 59

Table 2 Main Ttchnical modifications of ALPPS

Modification Abbreviation Full description Grade of evidence Reference

Inaugural description Classic ALPPS Associating liver partition and IV Schnitzbauer et al.14
portal vein ligation for
staged hepatectomy
Tourniquet ligation instead of transection ALTPS Associating liver tourniquet IV Robles et al.56
ligation and portal vein Case series
ligation for staged hepatectomy
Radiofrequency ablation instead of transection RALPPS Radiofrequency assisted liver Ib Jiao et al.58
partition with portal vein RCT
ligation for staged hepatectomy (vs. PVE)
PVE instead of portal vein ligation Hybrid ALPPS Hybrid between ALPPS and portal IV Li et al.59
vein embolization Case series
Intraoperative PVE through a mesenteric Mini ALPPS Minimization of stage 1 ALPPS IV de Santibanes et al.60
catheter instead of portal vein ligation by endovascular embolization Case
series
Incomplete instead of complete transection Partial ALPPS Limited or partial transection of IV Petrowsky et al.61
the parenchymal instead Case series
of complete transection

[93]. However, the LIGRO trial for CRLM, despite higher part of liver tissue was removed; second, there was no corre-
resectability of CLRM, still showed an 8% 90-day mortality, lation between liver failure after stage 2 and the liver volumes
double as high claimed by the self-reported registry and clear- achieved by the induction of hypertrophy. The first finding
ly too high for elective liver surgery for colorectal metastases raised the question into the mechanism by which liver failure
[90••]. This is especially worrisome in the face of the known occurs after portal rerouting after the volume of the whole
low cure and high recurrence rate in primarily unresectable liver remains unchanged. The second finding raised the ques-
CRLM [94, 95]. Contemporary chemotherapy studies with tion of volume-function dissociation after rapid hypertrophy.
patients enrolled that could also be ALPPS candidates show The authors advised the use a rigid liver function assessment
a comparable overall survival between ALPPS and chemo- prior to resection in stage 2 beyond simply the assessment of
therapy alone, but without the postoperative mortality [96]. volume changes. Because of the non-availability of HIDA for
Even with widespread adoption of partial ALPPS for CRLM direct function assessment in many centers, the use of the
and a further reduction of the 90-mortality, the use of ALPPS MELD score prior to stage 2 was recommended. MELD, de-
for patients with extensive CRLM had to be viewed with rived from total bilirubin, INR, and creatinine, turned out to be
considerable skepticism from the perspective of oncological a reliable predictor of complications after completion hepatec-
effectiveness. tomy. The attempt to improve patient selection and improve
assessment of liver function lead to the development of a
Potential Complications variety of risk scores beyond MELD to improve patient selec-
tion for ALPPS [72, 97]. The use of mebrofenin scintigraphy
A thorough analysis of complications was performed in 320 demonstrated that the function of liver tissue hypertrophied by
patients submitted to ALPPS and enrolled in a registry in 2015 ALPPS and assessed by mebrofenin scintigraphy does not
(Table 4) [77]. Although ALPPS is a technically challenging increase in parallel to the volume observed [98]. This finding
procedure, technical complications like bile leaks were not the may explain the high mortality and morbidity of classic
predominant problem. Twenty-eight (8.7%) of 320 patients ALPPS [99]. It is possible that newer ALPPS modifications
had experienced a 90-day mortality, and 20 fulfilled clinical lead to a matching increase in function and volume within the
criteria for liver failure after stage 2, although sFLR volumes short period of 1 week to 10 days. A matching rapid increase
had grown above 30% in 23 of the 28. The study detailed for of volume and function has recently been demonstrated for the
all patients with mortalities, whether liver failure criteria were novel liver venous deprivation (LVD) technique by the
present (71%), what the direct cause of death was (septic Montpellier group (see below), which also induces rapid liver
shock in 68%) and what was the potential root cause of death hypertrophy like ALPPS [100••]. Immaturity of the rapidly
(poor patient selection in 50% and inaccurate liver function hypertrophied liver may therefore only apply to classic
assessment in 29%). There were two unexpected findings: ALPPS and not for alternative methods to induce rapid hyper-
first, it was noted that patients experienced liver failure after trophy. For all other ALPPS modifications, however, such a
stage 1, i.e. the part of the procedure where only a diminutive demonstration is currently pending [101]. Therefore, the use
59 Page 10 of 20 Curr Oncol Rep (2020) 22: 59

Table 3 Data supporting/negating the adoption of ALPPS

Year Title Main conclusion Oxford level of Reference


evidence

2012 “Right portal vein ligation combined with in suit Case series presenting the innovation of IV Schnitzbauer
splitting induced rapid left lateral liver ALPPS and the effect on hypertrophy et al.14
lobe hypertrophy…” (Technical feasibilty)
2014 “Early survival and safety of ALPPS: Overall 90-day mortality 9%, but IIc Schadde
first report of the International ALPPS registry.” prohibitive 90-day mortality in et al.51
HCC (12%), IHCC (13%) and IHCC (27%)
(Safety)
2015 “ALPPS offers a better chance of complete resection…” Feasibility of resection for all tumor IIIb Schadde
types 83% compared to 66% for et al.58
PVE/PVL in retrospective case
control study
(Short-term oncological outcomes)
2017 “Risk adjustment in ALPPS is associated Trend to use ALPPS mostly in CRLM and IIc Linecker
with a dramatic decrease in early mortality…” improvement in 90-day mortality to 4% et al.67
(Safety)
2017 “Survival after ALPPS for advanced Overall survival after ALPPS for IIc Olthof
colorectal liver metastases…” CRLM not better than chemotherapy alone et al.70
(Medium term oncological outcomes)
2018 “ALPPS improves resectability compared ALPPS feasibilty of resection for CRLM (92%) Ib Sandstrom
with conventional Two-stage hepatectomy better than for TSH/PVE (57%) et al.65
in patients with advances CRLM…” (Short-term oncological outcomes in CRLM)

of volume parameters alone to assess functional maturity of Future Directions


the FLR is discouraged in partial ALPPS or any of the other
ALPPS modifications. ALPPS has been added as a surgical option to promote rapid
liver hypertrophy by portal vein rerouting and has also led to
many clinical innovations and new insights into the mecha-
Contraindications nisms of liver regeneration. [91] The most promising future
direction of rapid hypertrophy are the reduced stage 1 varia-
To use ALPPS or any of its modifications as a first- tions like ALTPS, RALPPS, hybrid ALPPS, mini-ALPPS,
choice for promoting FLR regeneration requires strong and partial ALPPS. ALTPS, hybrid, and mini-ALPPS have
arguments due to its high procedural risk (Fig. 4) only been reported by one group each, while partial ALPPS
[102]. Therefore, the first question one should ask is is more common, at least in the few centers that still perform
whether or not ALPPS is appropriate for patients with ALPPS at all [93]. If two-stage procedures are going to remain
bi-lobar CRLM if the tumors are not resectable by a a true viable option to induce rapid liver regeneration, partial
parenchymal sparing resection instead [103]. If extended ALPPS may be the most promising variation, but there are
hepatectomy is really required, more than two-thirds of only limited retrospective data available. Overall, however,
patients should obtain a sufficient FLR volume increase the most promising way to induce rapid hypertrophy may well
by PVE alone because the FLR is rarely lower than 15%. turn out to be LVD, a completely endovascular approach to
In patients with extremely small FLR (sFLR < 15%), induce rapid liver hypertrophy, which will be discussed later
PVE with embolization of segment 4 may be attempted in this review.
and the volume changes as well as oncological adequacy
of extensive liver surgery evaluated over time. Salvage
ALPPS [104, 105] or additional hepatic vein emboliza-
tion [106, 107] may then be indicated as a salvage ma- Radiation Lobectomy
neuver for patients with suboptimal FLR growth, provid-
ed that there are no oncological concerns. Based on the Rationale
data available, there is a limited role for a two-stage
hepatectomy, like classic ALPPS, in PHCC [108], The hepatic regenerative strategy termed “radiation lobecto-
IHCC [76], or HCC [109]. ALPPS variations with a re- my” refers to the lobar transarterial administration of yttrium-
duced stage 1, as described above, may well play a role 90 (90Y) labeled microspheres with the intent of obliterating
in primary liver tumors in the future [110]. tumor-laden liver tissue while concurrently spurring
Curr Oncol Rep (2020) 22: 59 Page 11 of 20 59

Table 4 Key data about mortality


after ALPPS procedure according Number of Proportion Key figures in ALPPS mortality
to Schadde et al.52 patients (%)

320 100 Retrospective study population in 55 centers


28 of 320 9 90-day mortality in all ALPPS patients
20 of 28 71 Criteria for liver failure (ISGLS) present in patients with mortality
23 of 28 82 Appropriate liver volume (sFLR >30%) prior to resection
in patients with mortality
19 of 28 68 Direct cause of death in patients with mortality: septic shock
14 of 28 50 Root cause of death in patients with mortality: wrong patient selection
8 of 28 29 Root cause of death in patients with mortality: inaccurate liver function
assessment
2 90-day mortality of patients younger than 60 years
14 90-day mortality of patients older than 69 years
7 90-day mortality of patients without ISGLS-liver failure
5 days after stage 1
20 90-day mortality of patients with ISGLS-liver failure 5 days after stage 1
5 90-day mortality of patients with MELD < 10 prior to stage 2
22 90- day mortality of patients with MELD > 10 prior to stage 2

ALPPS associating liver partition and portal vein ligation for staged hepatectomy, ISGLS international study group
for liver surgery, sFLR standardized FLR according to Vauthey, MELD model for end stage liver disease

hypertrophy of the non-radiated contralateral liver lobe prior which relates to the induction of a liver atrophy-hypertrophy
to liver resection surgery [111]. In radiation lobectomy, a de- complex as a regenerative response following radiation in-
liberate lobar treatment approach is employed, rather than duced hepatocyte injury and loss [113]. As a neoadjuvant
more selective segmental treatment, in order to purposefully therapeutic modality, radiation lobectomy offers a three-fold
deliver radiotherapy to non-tumorous liver parenchyma in ad- theoretical benefit in patients with primary or secondary liver
dition to liver tumor [112]. This technical approach underlies cancer who may undergo future hepatectomy: [1] locoregional
the conceptual and mechanistic basis for radiation lobectomy, liver-directed treatment of cancer, inciting tumor and

Fig. 4 Algorithm for contraindications to ALPPS: A variety of FLR volumes are not extremely small and PVE with segment IV
contraindications limit the use of ALPPS: ALPPS in primary liver embolization may achieve sufficient FLR hypertrophy; if PVE with
tumors results in prohibitive morbidity and mortality and should be segment IV embolization does not result in sufficient hypertrophy,
avoided; a large number of extensive colorectal liver metastases can be salvage HVE or salvage ALPPS may be useful
resected using a parenchymal sparing approach; PVE may suffice when
59 Page 12 of 20 Curr Oncol Rep (2020) 22: 59

hepatocyte cell death and consequent atrophy of irradiated Tumor progression in the FLR after radiation lobectomy has
liver tissues; [2] compensatory hypertrophy of the contralater- been reported to range from 19 to 25% for hepatocellular
al, untreated liver lobe, which may serve to enlarge this lobe as carcinoma (HCC) and metastatic colorectal carcinoma, re-
a future liver remnant (FLR) for subsequent surgical resection; spectively [112]. Reports of surgical outcomes after radiation
[3] allowance of a biological “test-of-time” to ensure durable lobectomy remain somewhat limited; in one study of 31 HCC
tumor response and lack of cancer progression, which would patients who underwent liver resection after radiation lobec-
contraindicate surgery, as well as appropriate FLR hypertro- tomy, 67% of patients with preoperative FLR less than 40%
phy to allow safe hepatectomy and avoidance of post- achieved FLRs exceeding 40% after 90Y treatment [119••].
operative liver failure. Following surgery, the incidence of grade IIIA or higher
Clavien-Dindo adverse events was 16%, and 3-year overall
Technique survival was 86% [119••].

The technical aspects of radiation lobectomy follow standard Potential Complications


90
Y radioembolization methods, including planning arteriog-
raphy with arterial anatomic mapping, non-target vessel em- Potential complications of radiation lobectomy primarily relate
bolization, and technetium-99 m macroaggregated albumin to adverse events precipitated by 90Y radioembolization, in-
(99mTc-MAA) scanning for lung shunt fraction calculation, cluding gastrointestinal ulceration, biliary sequela (e.g. radia-
followed by 90Y dosimetry according to customary methods tion cholecystitis, biloma, and/or abscess), radiation pneumoni-
with ensuing 90Y labeled microsphere delivery [114]. Patients tis, vascular injury, and lymphopenia, all of which occur with
who are to undergo radiation lobectomy should be operative relatively low incidence [120]. Post-radioembolization
candidates, with normal bilirubin levels, no portal hyperten- syndrome—consisting of fatigue, nausea, vomiting, anorexia,
sion, and no significant medical comorbidities which preclude a n d f e v e r — i s a n a n t i c i p a t e d s i d e e ff e c t o f 9 0 Y
surgery. In selecting patients for radiation lobectomy, well- radioembolization that occurs in one-quarter to one-half of pa-
compensated liver disease (Child-Pugh A5 status) and a pre- tients after treatment and is managed conservatively [120].
treatment FLR less than 50% have been associated with max- Liver insufficiency is an uncommon occurrence following ra-
imal contralateral lobar hypertrophy exceeding 10% [115••]. diation lobectomy, as evidenced by lack of serious biochemical
From the standpoint of 90Y dosimetry, a healthy liver injected adverse events and no meaningful alterations in Child-Pugh or
dose exceeding 88 Gy has also been linked with maximal Model for End-stage Liver disease scores after treatment in
contralateral hemi-liver hypertrophy exceeding 10% [115••]. published series [111, 112, 115••,116,117,118,119••].
These selection parameters provide preliminary guidance as to
specific patients who may optimally benefit from the radiation Situations in Which to Avoid Radiation Lobectomy
lobectomy approach.
As a neoadjuvant therapy performed prior to potential hepa-
Supporting Data tectomy, radiation lobectomy should not be pursued in pa-
tients who are not operative candidates. Conversely, deliberate
Patient and tumor data, procedure technical parameters, and parenchymal irradiation should generally be averted in surgi-
liver volumetric changes of radiation lobectomy from repre- cally unresectable liver cancer patients in order to preserve
sentative publications with cohort sizes exceeding 10 are sum- functional liver and allow for safe repetition of locoregional
marized in the Table 5 [111, 112, 115••, 116,117,118,119••]. therapy should multiple tumor treatment sessions be required.
Overall, degrees of hypertrophy (DH)—defined as post- Otherwise, radiation lobectomy should also be avoided in sce-
treatment relative FLR volume (FLR volume divided by total narios in which 90Y radioembolization is contraindicated,
liver volume) minus pre-treatment relative FLR volume, namely in patients with markedly compromised liver function,
expressed as a percentage—ranged from 10 to 26%. Time poor functional status, active hepatic infection, significant ex-
dependent analyses indicate that FLR volumetric increases trahepatic metastases, uncorrectable extra-hepatic arterial
occur steadily over the course of several months after 90Y flow, which may result in non-target 90Y microsphere deposi-
radioembolization (Fig. 5), with small degrees of hypertrophy tion, and a high lung shunt fraction, which would result in a
(around 2–3%) evident by 1-month post-treatment [112, 117]. single-session lung dose exceeding 30 Gy [114].
In contrast to radiation lobectomy, published data indicates
that more selective radiation segmentectomy does not achieve Future Directions
as substantial FLR hypertrophy, with the degree of hypertro-
phy limited to 2% [119••]. Tumor response rates after radia- Though supported by published retrospective data, continued
tion lobectomy are high, typically approximating 70–90% ob- investigation of the radiation lobectomy concept is necessary
jective imaging response [111, 112, 115••,116,117,118,119••]. to solidify this procedure’s standing among liver regenerative
Curr Oncol Rep (2020) 22: 59 Page 13 of 20 59

Table 5 Patient and tumor data, procedure technical parameters, and liver volumetric changes of radiation lobectomy
90
Author Year Patients Tumor type(s) Y Treated RE sessions Cumulative target dose (Gy) Follow-up time (months) FLR
device lobe (median, (median, range or IQR or mean (median, range or mean ± DH1
range) ± SD) SD) (%)

Jakobs, et al 2008 10 Various 2 Glass Right 1 120 4.6 17


Gaba, et al 2009 20 HCC (17), Glass Right 1.7 (1–5) 175 (103–413) 18 (2–49) 26
CCA (3)
Vouche, et al 2013 83 HCC (67), Glass Right NS 112 (74–215) >9 11
CCA (8),
mCRC (8)
Theysohn, et al 2014 45 HCC Glass Right 1 112 (100–160) 12 22
Lewandowski, 2016 13 HCC (10), Glass Right 1.4 (1–2) 154 (111–298) 1 (1–6) 10
et al CCA (2),
mCRC (1)
Gabr, et al 2018 31 HCC Glass Right 1 (1–3) 128 (113–143) 3 (2–5) 10
Palard, et al 2018 73 HCC Glass Right 1 150 ± 44 5.9 ± 3.4 17

90
Y yttrium-90, RE radioembolization, IQR interquartile range, SD standard deviation, FLR future liver remnant, DH degree of hypertrophy, HCC
hepatocellular carcinoma, CCA cholangiocarcinoma, mCRC metastatic colorectal carcinoma, NS not specified
1
DH = post-treatment relative FLR volume (FLR volume divided by total liver volume) minus pre-treatment relative FLR volume, expressed as a
percentage
2
Including colon, pancreas, neuroendocrine, breast, esophageal, hemangiopericytoma, melanoma, and unknown primary tumors

strategies. First, further delineation of optimal 90Y dosimetry comparative effectiveness of the radiation lobectomy ap-
schemes to induce robust atrophy-hypertrophy complex proach for different tumor types, spanning both primary (i.e.,
formation—as well as continued definition of other prognostic HCC) and secondary (e.g. metastatic colorectal carcinoma)
factors for hepatic volumetric changes—is required to im- liver malignancies, as well as liver disease stages, need be
prove the objectiveness of the radiation lobectomy approach investigated. Fourth, the safety, feasibility, and clinical out-
and predictability of the hepatic morphologic response. comes of ancillary technical approaches used to further refine
Second, the optimal time from 90Y treatment to resection is and/or increase the potency of the radiation lobectomy proce-
unknown. This timing could be critical as disease control rates dure should be studied; for example, combined radiation
are better with a shorter time intervals (e.g., 3 months versus segmentectomy aimed at ablative tumor destruction plus con-
6 months), and therefore, resectability rates may decrease over current radiation lobectomy aimed at normal liver parenchy-
time. Future study in this area is required to better define the mal irradiation may optimize tumor obliteration and parenchy-
most appropriate time to resection, even in the presence of a mal injury but requires further exploration. Finally, direct pro-
smaller absolute anticipated FLR volume. Third, the spective comparison of the hepatic volumetric response and

Fig. 5 Representative example of hepatic volumetric response after 90Y CT scan b performed 24 months after 1 session of glass 90 Y
radioembolization. Pre-treatment contrast enhanced axial computed radioembolization (dose = 189 Gy) reveals complete radiologic
tomography (CT) scan a demonstrates hypervascular right hepatic lobe response in treated right hepatic lobe HCC (arrow); right hepatic lobe
HCC (arrow); right hepatic lobe (outlined by dashed white line) (outlined by dashed white line) volume = 736 mL, left hepatic lobe
volume = 1267 mL, left hepatic lobe (outlined by dashed black line) (outlined by dashed black line) volume = 1052 mL (FLR = 59%)
volume = 628 mL (FLR = 33%). Post-treatment contrast enhanced axial
59 Page 14 of 20 Curr Oncol Rep (2020) 22: 59

clinical outcomes of radiation lobectomy combined with other to assess whether any additional regeneration occurred follow-
liver regenerative techniques—such as portal vein ing the second procedure.
embolization—should be pursued. To reduce the time from PVE or sequential PVE and HVE
to resection, the concept of simultaneous PVE and HVE has
recently emerged. Guiu et al. reported the first series of 7
Liver Venous Deprivation patients prepared using the so-called “LVD technique [13],”
consisting of simultaneous PVE and HVE of right and acces-
General Rationale sory right HVs when present. Since the middle HV drains
approximately 2/3 of the right paramedian sector [123] and
Liver venous deprivation (LVD) refers to both portal and he- given the excellent tolerance of LVD, Guiu et al. expanded the
patic vein embolization (HVE) [13]. The goal of this strategy concept into extended LVD (eLVD) by embolizing not only
is to accelerate liver regeneration of the FLR, in order to limit the right (and accessory right when present) but also the mid-
patient’s drop-out from resection. HVE in addition to PVE is dle HVs in another series of 10 patients [100••].
supposed to, as compared with PVE alone, induce more dam- In summary, HV occlusion (a) indirectly reduces hepatic
age to the embolized liver and, thus, increase contralateral arterial inflow in PV embolized segments thereby causing
liver regeneration and/or to counteract persistent minute portal more damage herein and (b) reverses portal flow in non PV
inflow that may occur after PVE. The rationale relies on the embolized segments that might be helpful in case of challeng-
following: ing portal anatomy or too proximal PVE.

In the Absence of Persistent Minute Portal Inflow After PVE


Technical Aspects Associated with Venous
An increased hepatic arterial inflow is commonly observed Deprivation
after PVE owing to the hepatic arterial buffer response.
Occlusion of hepatic venous outflow in a context of PVE The (e)LVD technique was originally described as a complete-
decreases the hepatic arterial inflow because of the limited ly transhepatic procedure (Fig. 6). The HVs (right, accessory
outflow route [121], either through sinusoids and inter- right when present, ± middle) are first accessed under ultra-
lobular communications to reach a zone with preserved out- sound using a Chiba introducer needle and a micropuncture
flow and/or through intrahepatic arterio-arterial communica- set. After opacification, a 0.018-in. Cope Mandril guidewire is
tions between venous-deprived and venous-preserved areas left in place in each HV. PVE is then conducted using a mix-
[13]. Reduced rather than interrupted arterial inflow might ture of iodized oil and n-butyl-cyanoacrylate. Each HV access
be preferable to prevent the risk of severe liver ischemia and is used to introduce a 6 or 7F sheath in order to deploy an
liver abscess formation [122] and to safely increase damage to Amplatzer Vascular Plug (AVP) II (St-Jude Medical,
the embolized liver, in order to further increase the regenera- Plymouth, MN), with its distal part ~ 15 mm before the junc-
tion of the non-embolized liver. tion with the vena cava to facilitate HV surgical ligation using
a stapler. The plug must be oversized by 75–100% to prevent
In the Presence of Persistent Minute Portal Inflow After PVE device migration. Accurate AVP position should be confirmed
by ultrasound. As soon as plugs are occluded, embolization of
When embolization of portal vessels is incomplete (e.g. chal- distal branches and potential collaterals (inter-hepatic vein
lenging portal anatomy) or too proximal, PVE is not optimal communications when visible) is conducted with a mixture
with limitation of the atrophy/hypertrophy complex. of ethiodized oil and n-butyl-cyanoacrylate. Finally, tract em-
Interestingly, HV occlusion in areas with preserved portal in- bolization of all transhepatic accesses is performed using the
flow induces compensatory increase in hepatic arterial flow same mixture.
and arterio-portal regurgitation with PV acting as a draining Veno-venous collaterals can be observed both from the
vein with reversed flow [121]. Therefore, persistent minute proximal (i.e. near the vena cava) or the distal part of the
portal inflow would be decreased or suppressed, thereby HVs [124]. Existing intrahepatic venous collaterals have been
redirecting all portal inflow to the non-embolized liver. reported in 46% of normal livers (mainly in the right liver),
In 2009, Hwang et al. [107] reported on HVE performed in with a diameter ≤ 1 mm in 73% cases [125]. In LVD, whether
12 patients who had shown limited liver regeneration 2 weeks the proximal collaterals are occluded by the plug itself, liquid
after PVE. This approach was shown to be safe and effective agents and especially glue seem to be well-designed to oc-
in facilitating contralateral regeneration by inducing more se- clude the distal ones safely, provided low dilution and slow
vere liver damage than usually observed after PVE. However, injection of glue. A critical point is to prepare the mixture of
this sequential approach did not spare time to resection com- glue and ethiodized oil before deploying the plug and to start
pared with PVE alone, as additional waiting time was required embolization as soon as the plug is occluded, given the rapid
Curr Oncol Rep (2020) 22: 59 Page 15 of 20 59

Fig. 6 LVD technique in a 67-year-old patient bearing multiple liver oversizing) is deployed in right HV (white arrow), and slow injection of
metastases in the right liver. a 0.018 in. guidewire is inserted into the n-butyl cyanoacrylate and ethiodized oil (1:2) is performed to fill in distal
right HV and right PVE is performed using a mixture of n-butyl branches of the right HV
cyanoacrylate and ethiodized oil (1:6). b Then, AVP type 2 (75%

enlargement and development of these collaterals when out- first plug was released, the next was hooked to the first one to
flow is occluded (Fig. 7). limit the risk of migration. All but the first centimeters of the
The concept of “bi-embolization” or “double emboli- HV was embolized plug by plug.
zation” has also been introduced in the literature but
refers only to the deployment of one AVP at the HV Data Supporting and/or Negating Its Adoption
origin through a transjugular access [126]. By defini-
tion, this technique leaves open the distal collaterals, So far, LVD has 100% technical success in published
which may circumvent the AVP within several minutes case-series [13, 100, 127]. In the inaugural paper, LVD
after its deployment, as observed when injecting through increased FLR volume by 53% after 23 days (range: 13–
a transhepatic access. By embolizing both the proximal 30 days) in 7 patients [13]. In Klatskin tumors, LVD
(AVP) and distal part (glue) as well as potential collat- combined with biliary drainage increased FLR volume
erals of HVs, LVD should probably be preferred. by 63.4% after 23.5 days (15–29 days) [127]. In 10
Recently, a new technical variation has been proposed in patients, eLVD showed fast and great increase in FLR
Klatskin tumors to avoid direct puncture of undrained bile volume with + 53.4% at day 7, + 62.5% at day 14 and +
ducts [127]. Through a transjugular approach, the HV was 63.3% at day 21 [100••]. Kinetic growth rate (KGR)
embolized from its distal part using several AVPs. After the after eLVD (i.e. 25 cc/day) was far beyond published
KGR following PVE (i.e. 4 cm3/day) or even LVD (i.e.
9 cm3/day) [100••]. Only ALPPS (KGR: 25.4–32.7 cc/
day) can compete with eLVD in terms of volumetric
gain, but it has been shown that liver volume (strongly)
overestimates liver function in ALPPS [98]. To the con-
trary, after eLVD [100••], FLR function evaluated using
99 m-Tc mebrofenin scintigraphy increased the same way
as FLR volume (+ 64.3% at day 21). The same team
compared the perioperative impact of LVD vs. PVE in
28 patients undergoing right hemi-hepatectomy [128].
They showed no difference in terms of morbidity/mortal-
ity, intraoperative bleeding and red blood cell volume,
fluid volume administration, and operative time, thereby
suggesting that LVD does not cause any additional sur-
gical difficulty.

Potential Complications
Fig. 7 During LVD performed in a 55-year-old patient, opacification
through the 7F-sheath inserted in the right HV, 10 min after AVP
deployment (black arrow). “Spiderweb” appearance of distal veno- So far, only minor complications have been reported for LVD
venous collaterals (white arrows) from the right to the middle HV including moderate pain and fever in keeping with post-
59 Page 16 of 20 Curr Oncol Rep (2020) 22: 59

embolization syndrome, together with moderate transaminase prior to curative resection. Although the standard of care for
elevation. Grade 2–3 asthenia has been observed in the first the past two decades has been PVE, newer approaches have
eLVD patients together with strong depletion in blood pools of been used in an attempt to make hypertrophy faster, to enable
vitamins and ions, given the high energy demand for regener- patients on the fringe of resection to become surgical candi-
ation [100••]. Systematic multivitamin and phosphorus sup- dates and to apply minimally invasive therapies to treat the
plementation administered early after eLVD solved this tumor while giving the “test of time” to assess its biology
problem. before undergoing a potentially morbid major hepatectomy.
Potential complications that may occur with (e)LVD are Each of the techniques described herein has advantages and
migration of embolic material and liver hematoma, but none disadvantages. However, at the present time, only few pro-
of these have been reported so far. spective clinical trial has been published and none comparing
all of these different strategies for preoperative hepatic hyper-
Contraindications trophy prior to resection. Hopefully, in the coming years, com-
prehensive prospective clinical trials will be performed for
Very little data are available on (e)LVD in the context of cir- each tumor type to help elucidate which option is best.
rhosis since only two cirrhotic patients undergoing LVD be-
fore right hemi-hepatectomy have been reported so far [128]. Compliance with Ethical Standards
Occluding the outflow (in addition to PVE) in cirrhosis is
likely to increase portal hypertension. In the absence of safety Conflict of Interest David C. Madoff declares that he has no conflict of
interest. Bruno C. Odisio declares that he has no conflict of interest. Erik
data in that context, it should be avoided to prepare cirrhotic
Schadde declares that he has no conflict of interest. Ron C. Gaba has
patients with LVD outside clinical trials. received research funding from Guerbet USA LLC, the United States
Department of Defense, and the National Institutes of Health, and also
Future Directions has a service contract with Janssen Research & Development LLC.
Roelof J. Bennink declares that he has no conflict of interest. Thomas
M. van Gulik declares that he has no conflict of interest. Boris Guiu
The role of eLVD to increase hepatic regeneration of declares that he has no conflict of interest.
the anticipated FLR prior to resection must be further
investigated. Whether occluding right and middle HVs Human and Animal Rights and Informed Consent This article does not
in right hemi-hepatectomy extended to segment 4 is contain any studies with human or animal subjects performed by any of
the authors.
fully acceptable, eLVD in a context of right hemi-
hepatectomy is more questionable. By causing venous
ischemia in the part of segment 4 drained by the middle
HV, it might provide an attractive and simpler option References
than segment 4 PVE [100••], but this must be further
explored. Therefore, randomized controlled trials are Papers of particular interest, published recently, have been
highly needed to better define the place of LVD versus highlighted as:
PVE, with special attention not only to volumetric and • Of importance
functional gains but also to oncological outcomes. •• Of major importance
Indeed, accelerating liver regeneration might also accel-
erate tumor growth and increase the risk of recurrence. 1. Tsao JI, Loftus JP, Nagorney DM, Adson MA, Ilstrup DM. Trends
in morbidity and mortality of hepatic resection for malignancy. A
Provided the results are confirmed in randomized con- matched comparative analysis. Ann Surg. 1994;220(2):199–205.
trolled trials, (e)LVD would have the potential to become the 2. Shirabe K, Shimada M, Gion T, Hasegawa H, Takenaka K,
standard of care for liver preparation, but the decade-long Utsunomiya T, et al. Postoperative liver failure after major hepatic
history of great efficacy and safety of PVE could rather argue resection for hepatocellular carcinoma in the modern era with
special reference to remnant liver volume. J Am Coll Surg.
in favor of tailored liver preparation, which means keeping the 1999;188(3):304–9.
more complex procedure (i.e. (e)LVD) when high functional 3. Shoup M, Gonen M, D'Angelica M, Jarnagin WR, DeMatteo RP,
gain is needed in the FLR. To aid in the determination of Schwartz LH, et al. Volumetric analysis predicts hepatic dysfunc-
which patients will benefit from PVE or (e)LVD, future study tion in patients undergoing major liver resection. J Gastrointest
Surg. 2003;7(3):325–30.
will be necessary. 4. Ribero D, Abdalla EK, Madoff DC, Donadon M, Loyer EM,
Vauthey JN. Portal vein embolization before major hepatectomy
and its effects on regeneration, resectability and outcome. Br J
Conclusion Surg. 2007;94(11):1386–94.
5. Makuuchi M, Thai BL, Takayasu K, Takayama T, Kosuge T,
Gunven P, et al. Preoperative portal embolization to increase safe-
There are many different interventional radiological and sur- ty of major hepatectomy for hilar bile duct carcinoma: a prelimi-
gical options that can be used to promote hepatic hypertrophy nary report. Surgery. 1990;107(5):521–7.
Curr Oncol Rep (2020) 22: 59 Page 17 of 20 59

6. Madoff DC, Abdalla EK, Vauthey JN. Portal vein embolization in new cut-off to estimate postoperative risks after extended resection
preparation for major hepatic resection: evolution of a new stan- in noncirrhotic liver. J Am Coll Surg. 2007;204(1):22–33.
dard of care. J Vasc Interv Radiol. 2005;16(6):779–90. 23. Lin XJ, Yang J, Chen XB, Zhang M, Xu MQ. The critical value of
7. Abdalla EK, Barnett CC, Doherty D, Curley SA, Vauthey JN. remnant liver volume-to-body weight ratio to estimate
Extended hepatectomy in patients with hepatobiliary malignancies posthepatectomy liver failure in cirrhotic patients. J Surg Res.
with and without preoperative portal vein embolization. Arch 2014;188(2):489–95.
Surg. 2002;137(6):675–80 discussion 80-1. 24. Akita H, Sasaki Y, Yamada T, Gotoh K, Ohigashi H, Eguchi H,
8. Palavecino M, Chun YS, Madoff DC, Zorzi D, Kishi Y, Kaseb et al. Real-time intraoperative assessment of residual liver func-
AO, et al. Major hepatic resection for hepatocellular carcinoma tional reserve using pulse dye densitometry. World J Surg.
with or without portal vein embolization: perioperative outcome 2008;32(12):2668–74.
and survival. Surgery. 2009;145(4):399–405. 25. Yokoyama Y, Nishio H, Ebata T, Igami T, Sugawara G, Nagino M.
9. Elias D, De Baere T, Roche A, Mducreux LJ, Lasser P. During Value of indocyanine green clearance of the future liver remnant in
liver regeneration following right portal embolization the growth predicting outcome after resection for biliary cancer. Br J Surg.
rate of liver metastases is more rapid than that of the liver paren- 2010;97(8):1260–8.
chyma. Br J Surg. 1999;86(6):784–8. 26. Rassam F, Olthof PB, Bennink RJ, van Gulik TM. Current mo-
10. Kokudo N, Tada K, Seki M, Ohta H, Azekura K, Ueno M, et al. dalities for the assessment of future remnant liver function. Visc
Proliferative activity of intrahepatic colorectal metastases after Med. 2017;33(6):442–8.
preoperative hemihepatic portal vein embolization. Hepatology. 27. Cieslak KP, Runge JH, Heger M, Stoker J, Bennink RJ, van Gulik
2001;34(2):267–72. TM. New perspectives in the assessment of future remnant liver.
11. Simoneau E, Aljiffry M, Salman A, Abualhassan N, Cabrera T, Dig Surg. 2014;31(4–5):255–68.
Valenti D, et al. Portal vein embolization stimulates tumour growth 28. Chapelle T, Op De Beeck B, Huyghe I, Francque S, Driessen A,
in patients with colorectal cancer liver metastases. HPB (Oxford). Roeyen G, et al. Future remnant liver function estimated by com-
2012;14(7):461–8. bining liver volumetry on magnetic resonance imaging with total
12. Gabr A, Polineni P, Mouli SK, Riaz A, Lewandowski RJ, Salem liver function on (99m)Tc-mebrofenin hepatobiliary scintigraphy:
R. Neoadjuvant radiation lobectomy as an alternative to portal can this tool predict post-hepatectomy liver failure? HPB
vein embolization in hepatocellular carcinoma. Semin Nucl (Oxford). 2016;18(6):494–503.
Med. 2019;49(3):197–203. 29.•• Cieslak KP, Bennink RJ, de Graaf W, van Lienden KP, Besselink
MG, Busch OR, et al. Measurement of liver function using
13. Guiu B, Chevallier P, Denys A, Delhom E, Pierredon-Foulongne
hepatobiliary scintigraphy improves risk assessment in patients
MA, Rouanet P, et al. Simultaneous trans-hepatic portal and he-
undergoing major liver resection. HPB (Oxford). 2016;18(9):
patic vein embolization before major hepatectomy: the liver ve-
773–80 Study illustrates the rationale for performing func-
nous deprivation technique. Eur Radiol. 2016;26(12):4259–67.
tional imaging as a better tool for surgical risk assessment
14. Schnitzbauer AA, Lang SA, Goessmann H, Nadalin S, Baumgart
than standard volumetry.
J, Farkas SA, et al. Right portal vein ligation combined with in situ
30. Serenari M, Collaud C, Alvarez FA, de Santibanes M, Giunta D,
splitting induces rapid left lateral liver lobe hypertrophy enabling
Pekolj J, et al. Interstage assessment of remnant liver function in
2-staged extended right hepatic resection in small-for-size settings.
ALPPS using Hepatobiliary Scintigraphy: prediction of
Ann Surg. 2012;255(3):405–14.
Posthepatectomy liver failure and Introduction of the HIBA index.
15. Gulec SA, Pennington K, Hall M, Fong Y. Preoperative Y-90
Ann Surg. 2018;267(6):1141–7.
microsphere selective internal radiation treatment for tumor
31.• Rassam F, Olthof PB, Richardson H, van Gulik TM, Bennink RJ.
downsizing and future liver remnant recruitment: a novel ap-
Practical guidelines for the use of technetium-99m mebrofenin
proach to improving the safety of major hepatic resections.
hepatobiliary scintigraphy in the quantitative assessment of liver
World J Surg Oncol. 2009;7:6.
function. Nucl Med Commun. 2019;40(4):297–307 Article high-
16. Guglielmi A, Ruzzenente A, Conci S, Valdegamberi A, Iacono C. lights practical guidelines on how functional liver imaging
How much remnant is enough in liver resection? Dig Surg. should be performed.
2012;29(1):6–17. 32. de Graaf W, van Lienden KP, van den Esschert JW, Bennink RJ,
17. Martel G, Cieslak KP, Huang R, van Lienden KP, Wiggers JK, van Gulik TM. Increase in future remnant liver function after
Belblidia A, et al. Comparison of techniques for volumetric anal- preoperative portal vein embolization. Br J Surg. 2011;98(6):
ysis of the future liver remnant: implications for major hepatic 825–34.
resections. HPB (Oxford). 2015;17(12):1051–7. 33. de Graaf W, van Lienden KP, van Gulik TM, Bennink RJ.
18. Madoff DC, Hicks ME, Vauthey JN, Charnsangavej C, Morello (99m)Tc-mebrofenin hepatobiliary scintigraphy with SPECT for
FA Jr, Ahrar K, et al. Transhepatic portal vein embolization: anat- the assessment of hepatic function and liver functional volume
omy, indications, and technical considerations. Radiographics. before partial hepatectomy. J Nucl Med : Off Publ, Soc of Nucl
2002;22(5):1063–76. Med. 2010;51(2):229–36.
19. Johnson TN, Tucker GT, Tanner MS, Rostami-Hodjegan A. 34. Rassam F, Olthof PB, van Lienden KP, Bennink RJ, Besselink
Changes in liver volume from birth to adulthood: a meta-analysis. MG, Busch OR, et al. Functional and volumetric assessment of
Liver Transpl. 2005;11(12):1481–93. liver segments after portal vein embolization: differences in hy-
20. Olthof PB, van Dam R, Jovine E, Campos RR, de Santibanes E, pertrophy response. Surgery. 2019;165(4):686–95.
Oldhafer K, et al. Accuracy of estimated total liver volume formu- 35. Cieslak KP, Huisman F, Bais T, Bennink RJ, van Lienden KP,
las before liver resection. Surgery. 2019. Verheij J, et al. Future remnant liver function as predictive factor
21. Shindoh J, Truty MJ, Aloia TA, Curley SA, Zimmitti G, Huang for the hypertrophy response after portal vein embolization.
SY, et al. Kinetic growth rate after portal vein embolization pre- Surgery. 2017;162(1):37–47.
dicts posthepatectomy outcomes: toward zero liver-related mortal- 36. Olthof PB, Coelen RJS, Bennink RJ, Heger M, Lam MF,
ity in patients with colorectal liver metastases and small future Besselink MG, et al. (99m)Tc-mebrofenin hepatobiliary scintigra-
liver remnant. J Am Coll Surg. 2013;216(2):201–9. phy predicts liver failure following major liver resection for
22. Truant S, Oberlin O, Sergent G, Lebuffe G, Gambiez L, Ernst O, perihilar cholangiocarcinoma. HPB (Oxford). 2017;19(10):850–
et al. Remnant liver volume to body weight ratio > or =0.5%: a 8.
59 Page 18 of 20 Curr Oncol Rep (2020) 22: 59

37. Malinowski M, Lock JF, Seehofer D, Gebauer B, Schulz A, 54. Kinoshita H, Sakai K, Hirohashi K, Igawa S, Yamasaki O, Kubo
Demirel L, et al. Preliminary study on liver function changes after S. Preoperative portal vein embolization for hepatocellular carci-
trisectionectomy with versus without prior portal vein emboliza- noma. World J Surg. 1986;10(5):803–8.
tion. Surg Today. 2016;46(9):1053–61. 55. Denys A, Madoff DC, Doenz F, Schneider F, Gillet M, Vauthey
38. Nilsson H, Blomqvist L, Douglas L, Nordell A, Jacobsson H, JN, et al. Indications for and limitations of portal vein emboliza-
Hagen K, et al. Dynamic gadoxetate-enhanced MRI for the as- tion before major hepatic resection for hepatobiliary malignancy.
sessment of total and segmental liver function and volume in pri- Surg Oncol Clin N Am. 2002;11(4):955–68.
mary sclerosing cholangitis. J Magn Reson Imaging. 2014;39(4): 56. van Lienden KP, van den Esschert JW, de Graaf W, Bipat S,
879–86. Lameris JS, van Gulik TM, et al. Portal vein embolization before
39. Ryeom HK, Kim SH, Kim JY, Kim HJ, Lee JM, Chang YM, et al. liver resection: a systematic review. Cardiovasc Intervent Radiol.
Quantitative evaluation of liver function with MRI using Gd- 2013;36(1):25–34.
EOB-DTPA. Korean J Radiol. 2004;5(4):231–9. 57. Nagino M, Kamiya J, Nishio H, Ebata T, Arai T, Nimura Y. Two
40. Rassam F, Zhang T, Cieslak KP, Lavini C, Stoker J, Bennink RJ, hundred forty consecutive portal vein embolizations before ex-
et al. Comparison between dynamic gadoxetate-enhanced MRI tended hepatectomy for biliary cancer: surgical outcome and
and (99m)Tc-mebrofenin hepatobiliary scintigraphy with SPECT long-term follow-up. Ann Surg. 2006;243(3):364–72.
for quantitative assessment of liver function. Eur Radiol 2019. 58. de Baere T, Teriitehau C, Deschamps F, Catherine L, Rao P,
41. Yokoyama Y, Nagino M, Nimura Y. Mechanisms of hepatic re- Hakime A, et al. Predictive factors for hypertrophy of the future
generation following portal vein embolization and partial hepatec- remnant liver after selective portal vein embolization. Ann Surg
tomy: a review. World J Surg. 2007;31(2):367–74. Oncol. 2010;17(8):2081–9.
42. Bucher NL, Swaffield MN. The rate of incorporation of labeled 59. Bent CL, Low D, Matson MB, Renfrew I, Fotheringham T. Portal
thymidine into the deoxyribonucleic acid of regenerating rat liver vein embolization using a nitinol plug (Amplatzer vascular plug)
in relation to the amount of liver excised. Cancer Res. 1964;24: in combination with histoacryl glue and iodinized oil: adequate
1611–25. hypertrophy with a reduced risk of nontarget embolization.
43. Shindoh J, Tzeng CW, Aloia TA, Curley SA, Huang SY, Mahvash Cardiovasc Intervent Radiol. 2009;32(3):471–7.
A, et al. Safety and efficacy of portal vein embolization before 60. Abulkhir A, Limongelli P, Healey AJ, Damrah O, Tait P, Jackson
planned major or extended hepatectomy: an institutional experi- J, et al. Preoperative portal vein embolization for major liver re-
ence of 358 patients. J Gastrointest Surg : Off J Soc Surg section: a meta-analysis. Ann Surg. 2008;247(1):49–57.
Alimentary Tract. 2014;18(1):45–51. 61. Ebata T, Yokoyama Y, Igami T, Sugawara G, Takahashi Y, Nagino
M. Portal vein embolization before extended hepatectomy for bil-
44. Avritscher R, de Baere T, Murthy R, Deschamps F, Madoff DC.
iary cancer: current technique and review of 494 consecutive em-
Percutaneous transhepatic portal vein embolization: rationale,
bolizations. Dig Surg. 2012;29(1):23–9.
technique, and outcomes. Semin Interv Radiol. 2008;25(2):132–
62. Hoekstra LT, van Lienden KP, Doets A, Busch OR, Gouma DJ,
45.
van Gulik TM. Tumor progression after preoperative portal vein
45. Denys AL, De Baere T, Doenz F. Portal vein embolization: a plea
embolization. Ann Surg. 2012;256(5):812–7 discussion 7-8.
for strict patient selection. AJR Am J Roentgenol. 2006;187(1):
63. Azoulay D, Castaing D, Smail A, Adam R, Cailliez V, Laurent A,
W125 author reply 6.
et al. Resection of nonresectable liver metastases from colorectal
46. Nagino M, Nimura Y, Kamiya J, Kondo S, Uesaka K, Kin Y, et al.
cancer after percutaneous portal vein embolization. Ann Surg.
Right or left trisegment portal vein embolization before hepatic
2000;231(4):480–6.
trisegmentectomy for hilar bile duct carcinoma. Surgery.
64. Nagino M, Nimura Y, Kamiya J, Kanai M, Hayakawa N,
1995;117(6):677–81.
Yamamoto H. Immediate increase in arterial blood flow in
47. Adam R, Laurent A, Azoulay D, Castaing D, Bismuth H. Two- embolized hepatic segments after portal vein embolization: CT
stage hepatectomy: a planned strategy to treat irresectable liver demonstration. AJR Am J Roentgenol. 1998;171(4):1037–9.
tumors. Ann Surg. 2000;232(6):777–85. 65. Pawlik TM, Olino K, Gleisner AL, Torbenson M, Schulick R,
48. Jaeck D, Oussoultzoglou E, Rosso E, Greget M, Weber JC, Choti MA. Preoperative chemotherapy for colorectal liver metas-
Bachellier P. A two-stage hepatectomy procedure combined with tases: impact on hepatic histology and postoperative outcome. J
portal vein embolization to achieve curative resection for initially Gastrointest Surg. 2007;11(7):860–8.
unresectable multiple and bilobar colorectal liver metastases. Ann 66. Piardi T, Memeo R, Renard Y, Ammendola M, Bruno O,
Surg. 2004;240(6):1037–49 discussion 49-51. Habersetzer F, et al. Management of large hepatocellular carcino-
49. Narita M, Oussoultzoglou E, Jaeck D, Fuchschuber P, Rosso E, ma by sequential transarterial chemoembolization and portal vein
Pessaux P, et al. Two-stage hepatectomy for multiple bilobar co- embolization: a systematic review of the literature. Minerva Chir.
lorectal liver metastases. Br J Surg. 2011;98(10):1463–75. 2016;71(3):192–200.
50. Tanaka K, Shimada H, Nagano Y, Endo I, Sekido H, Togo S. 67. Li T, Zhu J, Ma K, Liu N, Feng K, Li X, et al. Autologous bone
Outcome after hepatic resection versus combined resection and marrow-derived mesenchymal stem cell transplantation promotes
microwave ablation for multiple bilobar colorectal metastases to liver regeneration after portal vein embolization in cirrhotic rats. J
the liver. Surgery. 2006;139(2):263–73. Surg Res. 2013;184(2):1161–73.
51. Di Stefano DR, de Baere T, Denys A, Hakime A, Gorin G, Gillet 68. Kaibori M, Adachi Y, Shimo T, Ishizaki M, Matsui K, Tanaka Y,
M, et al. Preoperative percutaneous portal vein embolization: eval- et al. Stimulation of liver regeneration after hepatectomy in mice
uation of adverse events in 188 patients. Radiology. 2005;234(2): by injection of bone marrow mesenchymal stem cells via the por-
625–30. tal vein. Transplant Proc. 2012;44(4):1107–9.
52. Kodama Y, Shimizu T, Endo H, Miyamoto N, Miyasaka K. 69. Furst G, Schulte am Esch J, Poll LW, Hosch SB, Fritz LB, Klein
Complications of percutaneous transhepatic portal vein emboliza- M, et al. Portal vein embolization and autologous CD133+ bone
tion. J Vasc Interv Radiol. 2002;13(12):1233–7. marrow stem cells for liver regeneration: initial experience.
53. Nagino M, Nimura Y, Kamiya J, Kondo S, Kanai M. Selective Radiology. 2007;243(1):171–9.
percutaneous transhepatic embolization of the portal vein in prep- 70. Am Esch JS, Schmelzle M, Furst G, Robson SC, Krieg A, Duhme
aration for extensive liver resection: the ipsilateral approach. C, et al. Infusion of CD133+ bone marrow-derived stem cells after
Radiology. 1996;200(2):559–63. selective portal vein embolization enhances functional hepatic
Curr Oncol Rep (2020) 22: 59 Page 19 of 20 59

reserves after extended right hepatectomy: a retrospective single- 88. Matsuo K, Hiroshima Y, Yamazaki K, Kasahara K, Kikuchi Y,
center study. Ann Surg. 2012;255(1):79–85. Kawaguchi D, et al. Immaturity of bile canalicular-ductule net-
71. Odisio BC, Kaseb AO. Anti-angiogenic therapy in the setting of works in the future liver remnant while associating liver partition
TACE: an elusive synergy? Lancet Gastroenterol Hepatol. and portal vein occlusion for staged hepatectomy (ALPPS). Ann
2018;3(1):5–6. Surg Oncol. 2017;24(9):2456–64.
72. Linecker M, Stavrou GA, Oldhafer KJ, Jenner RM, Seifert B, 89. Tanaka K, Matsuo K, Murakami T, Kawaguchi D, Hiroshima Y,
Lurje G, et al. The ALPPS risk score: avoiding futile use of Koda K, et al. Associating liver partition and portal vein ligation
ALPPS. Ann Surg. 2016;264(5):763–71. for staged hepatectomy (ALPPS): short-term outcome, functional
73. Olthof PB, Schadde E. The rush to novelty and high expectations changes in the future liver remnant, and tumor growth activity. Eur
in surgery: the case of ALPPS. J Clin Transl Res. 2016;2(3):79– J Surg Oncol. 2015;41(4):506–12.
83. 90.•• Sandstrom P, Rosok BI, Sparrelid E, Larsen PN, Larsson AL,
74. Aloia TA, Vauthey JN. Associating liver partition and portal vein Lindell G, et al. ALPPS improves resectability compared with
ligation for staged hepatectomy (ALPPS): what is gained and what conventional two-stage hepatectomy in patients with advanced
is lost? Ann Surg. 2012;256(3):e9 author reply e16–9. colorectal liver metastasis: results from a Scandinavian
75. Dokmak S, Belghiti J. Which limits to the "ALPPS" approach? Multicenter Randomized Controlled Trial (LIGRO Trial). Ann
Annals of surgery. 2012;256(3):e6; author reply e16–7. Surg. 2018;267(5):833–40 First randomized trial comparing
76. Schadde E, Ardiles V, Robles-Campos R, Malago M, Machado ALPPS to two-stage hepatectomy in patients with colorectal
M, Hernandez-Alejandro R, et al. Early survival and safety of liver metastases with small future liver remnants. Study found
ALPPS: first report of the international ALPPS Registry. Ann that the resection rate was 92% in the ALPPS arm and 57% in
Surg. 2014;260(5):829–36 discussion 36-8. the two-stage hepatectomy arm with the conclusion that
ALPPS improves resectability.
77. Schadde E, Raptis DA, Schnitzbauer AA, Ardiles V, Tschuor C,
Lesurtel M, et al. Prediction of mortality after ALPPS Stage-1: an 91. Olthof PB, Schnitzbauer AA, Schadde E. The HPB controversy of
analysis of 320 patients from the International ALPPS Registry. the decade: 2007-2017 - ten years of ALPPS. Eur J Surg Oncol.
Ann Surg. 2015;262(5):780–6. 2018;44(10):1624–7.
92.• Hasselgren K, Rosok BI, Larsen PN, Sparrelid E, Lindell G,
78. Schadde E, Ardiles V, Slankamenac K, Tschuor C, Sergeant G,
Schultz NA, et al. ALPPS improves survival compared with
Amacker N, et al. ALPPS offers a better chance of complete re-
TSH in patients affected of CRLM: survival analysis from the
section in patients with primarily unresectable liver tumors com-
randomized controlled trial LIGRO. Ann Surg. 2019. Survival
pared with conventional-staged hepatectomies: results of a multi-
analysis published 4 years after the conclusion of the
center analysis. World J Surg. 2014;38(6):1510–9.
LIGRO trial shows that median survival after ALPPS for
79. Schadde E, Schnitzbauer AA, Tschuor C, Raptis DA, Bechstein
colorectal liver metastases is 46 months compared to 26
WO, Clavien PA. Systematic review and meta-analysis of feasi-
months after two-stage hepatectomy.
bility, safety, and efficacy of a novel procedure: associating liver
93. Linecker M, Bjornsson B, Stavrou GA, Oldhafer KJ, Lurje G,
partition and portal vein ligation for staged hepatectomy. Ann
Neumann U, et al. Risk adjustment in ALPPS is associated with
Surg Oncol. 2015;22(9):3109–20.
a dramatic decrease in early mortality and morbidity. Ann Surg.
80. Alvarez FA, Ardiles V, Sanchez Claria R, Pekolj J, de Santibanes 2017;266(5):779–86.
E. Associating liver partition and portal vein ligation for staged 94. Fong Y, Fortner J, Sun RL, Brennan MF, Blumgart LH. Clinical
hepatectomy (ALPPS): tips and tricks. J Gastrointest Surg : Offl J score for predicting recurrence after hepatic resection for metasta-
Soc Surg Alimentary Tract. 2013;17(4):814–21. tic colorectal cancer: analysis of 1001 consecutive cases. Ann
81. Robles R, Parrilla P, Lopez-Conesa A, Brusadin R, de la Pena J, Surg. 1999;230(3):309–18 discussion 18-21.
Fuster M, et al. Tourniquet modification of the associating liver 95. Tomlinson JS, Jarnagin WR, DeMatteo RP, Fong Y, Kornprat P,
partition and portal ligation for staged hepatectomy procedure. Br Gonen M, et al. Actual 10-year survival after resection of colorec-
J Surg. 2014;101(9):1129–34 discussion 34. tal liver metastases defines cure. J Clin Oncol. 2007;25(29):4575–
82. Gall TM, Sodergren MH, Frampton AE, Fan R, Spalding DR, 80.
Habib NA, et al. Radio-frequency-assisted liver partition with por- 96. Olthof PB, Huiskens J, Wicherts DA, Huespe PE, Ardiles V,
tal vein ligation (RALPP) for liver regeneration. Ann Surg. Robles-Campos R, et al. Survival after associating liver partition
2015;261(2):e45–6. and portal vein ligation for staged hepatectomy (ALPPS) for ad-
83. Jiao LR, Fajardo Puerta AB, Gall TMH, Sodergren MH, Frampton vanced colorectal liver metastases: a case-matched comparison
AE, Pencavel T, et al. Rapid induction of liver regeneration for with palliative systemic therapy. Surgery. 2017;161(4):909–19.
major hepatectomy (REBIRTH): a randomized controlled trial of 97. Huiskens J, Schadde E, Lang H, Malago M, Petrowsky H, de
portal vein embolisation versus ALPPS assisted with radiofre- Santibanes E, et al. Avoiding postoperative mortality after
quency. Cancers (Basel). 2019;11(3). ALPPS-development of a tumor-specific risk score for colorectal
84. Li J, Kantas A, Ittrich H, Koops A, Achilles EG, Fischer L, et al. liver metastases. Oxford: HPB; 2019.
Avoid "all-touch" by hybrid ALPPS to achieve oncological effi- 98. Olthof PB, Tomassini F, Huespe PE, Truant S, Pruvot FR, Troisi
cacy. Ann Surg. 2016;263(1):e6–7. RI, et al. Hepatobiliary scintigraphy to evaluate liver function in
85. de Santibanes E, Alvarez FA, Ardiles V, Pekolj J, de Santibanes associating liver partition and portal vein ligation for staged hep-
M. Inverting the ALPPS paradigm by minimizing first stage im- atectomy: liver volume overestimates liver function. Surgery.
pact: the mini-ALPPS technique. Langenbeck's Arch Surg. 2017;162(4):775–83.
2016;401(4):557–63. 99. Kang D, Schadde E. Hypertrophy and liver function in ALPPS:
86. Petrowsky H, Gyori G, de Oliveira M, Lesurtel M, Clavien PA. Is correlation with morbidity and mortality. Visc Med. 2017;33(6):
partial-ALPPS safer than ALPPS? A single-center experience. 426–33.
Ann Surg. 2015;261(4):e90–2. 100.•• Guiu B, Quenet F, Escal L, Bibeau F, Piron L, Rouanet P, et al.
87. Linecker M, Kambakamba P, Reiner CS, Linh Nguyen-Kim TD, Extended liver venous deprivation before major hepatectomy in-
Stavrou GA, Jenner RM, et al. How much liver needs to be duces marked and very rapid increase in future liver remnant func-
transected in ALPPS? A translational study investigating the con- tion. Eur Radiol. 2017;27(8):3343–52 This study shows in 10
cept of less invasiveness. Surgery. 2017;161(2):453–64. patients that eLVD is safe and provides great increase in both
59 Page 20 of 20 Curr Oncol Rep (2020) 22: 59

FLR volume and function, unprecedented for an intervention- 116. Jakobs TF, Saleem S, Atassi B, Reda E, Lewandowski RJ,
al radiology procedure. Yaghmai V, et al. Fibrosis, portal hypertension, and hepatic vol-
101. Deshayes E, Schadde E, Piron L, Quenet F, Guiu B. Extended ume changes induced by intra-arterial radiotherapy with 90yttrium
liver venous deprivation leads to a higher increase in liver function microspheres. Dig Dis Sci. 2008;53(9):2556–63.
that ALPPS in early assessment: a comment to "Sparrelid, E. et al. 117. Theysohn JM, Ertle J, Muller S, Schlaak JF, Nensa F, Sipilae S,
Dynamic evaluation of liver volume and function in associating et al. Hepatic volume changes after lobar selective internal radia-
liver partition and portal vein ligation for staged hepatectomy. tion therapy (SIRT) of hepatocellular carcinoma. Clin Radiol.
Journal of gastrointestinal surgery (2017)". J Gastrointest Surg : 2014;69(2):172–8.
Off J Soc Surg Alimentary Tract. 2017;21(10):1754–5. 118. Lewandowski RJ, Donahue L, Chokechanachaisakul A, Kulik L,
102. Belghiti J, Dokmak S, Schadde E. ALPPS: innovation for inno- Mouli S, Caicedo J, et al. (90) Y radiation lobectomy: outcomes
vation's sake. Surgery. 2016;159(5):1287–8. following surgical resection in patients with hepatic tumors and
103. Vigano L, Costa G, Toso C, Cimino M, Andres A, Rubbia-Brandt small future liver remnant volumes. J Surg Oncol. 2016;114(1):
L, et al. "Precision surgery" for colorectal liver metastases: is the 99–105.
time ripe? Ann Surg. 2018. 119.•• Gabr A, Abouchaleh N, Ali R, Baker T, Caicedo J, Katariya N,
104. Tschuor C, Croome KP, Sergeant G, Cano V, Schadde E, Ardiles et al. Outcomes of surgical resection after radioembolization for
V, et al. Salvage parenchymal liver transection for patients with hepatocellular carcinoma. J Vasc Interv Radiol. 2018;29(11):
insufficient volume increase after portal vein occlusion – an ex- 1502–10 e1 Shows that radioembolization can facilitate safe
tension of the ALPPS approach. Eur J Surg Oncol. 2013;39(11): bridge to surgical resection by providing disease control and
1230–5. future liver remnant hypertrophy.
105. Enne M, Schadde E, Bjornsson B, Hernandez Alejandro R, 120. Riaz A, Lewandowski RJ, Kulik LM, Mulcahy MF, Sato KT, Ryu
Steinbruck K, Viana E, et al. ALPPS as a salvage procedure after RK, et al. Complications following radioembolization with
insufficient future liver remnant hypertrophy following portal vein yttrium-90 microspheres: a comprehensive literature review. J
occlusion. HPB (Oxford). 2017;19(12):1126–9. Vasc Interv Radiol. 2009;20(9):1121–30 quiz 31.
106. Balzan SM, Gava VG, Magalhaes MA, Dotto ML. Outflow mod-
121. Ko GY, Hwang S, Sung KB, Gwon DI, Lee SG. Interventional
ulation to target liver regeneration: something old, something new.
oncology: new options for interstitial treatments and intravascular
Eur J Surg Oncol. 2014;40(2):140–3.
approaches: right hepatic vein embolization after right portal vein
107. Hwang S, Lee SG, Ko GY, Kim BS, Sung KB, Kim MH, et al.
embolization for inducing hypertrophy of the future liver remnant.
Sequential preoperative ipsilateral hepatic vein embolization after
J Hepatobiliary Pancreat Sci. 2010;17(4):410–2.
portal vein embolization to induce further liver regeneration in
patients with hepatobiliary malignancy. Ann Surg. 2009;249(4): 122. Nagino M, Kanai M, Morioka A, Yamamoto H, Kawabata Y,
608–16. Hayakawa N, et al. Portal and arterial embolization before exten-
108. Olthof PB, Coelen RJS, Wiggers JK, Groot Koerkamp B, Malago sive liver resection in patients with markedly poor functional re-
M, Hernandez-Alejandro R, et al. High mortality after ALPPS for serve. J Vasc Interv Radiol. 2000;11(8):1063–8.
perihilar cholangiocarcinoma: case-control analysis including the 123. Akamatsu N, Sugawara Y, Kaneko J, Sano K, Imamura H,
first series from the international ALPPS registry. HPB (Oxford). Kokudo N, et al. Effects of middle hepatic vein reconstruction
2017;19(5):381–7. on right liver graft regeneration. Transplantation. 2003;76(5):
109. D'Haese JG, Neumann J, Weniger M, Pratschke S, Bjornsson B, 832–7.
Ardiles V, et al. Should ALPPS be used for liver resection in 124. Gai YH, Cai SF, Guo WB, Zhang CQ, Liang B, Jia T, et al.
intermediate-stage HCC? Ann Surg Oncol. 2016;23(4):1335–43. Sonographic classification of draining pathways of obstructed he-
110. Olthof PB, Jarnagin WR, van Gulik TM. Acknowledging the patic veins in Budd-Chiari syndrome. J Clin Ultrasound : JCU.
flaws to advance with the strengths of ALPPS. HPB (Oxford). 2014;42(3):134–42.
2017;19(5):473–4. 125. Hribernik M, Trotovsek B. Intrahepatic venous anastomoses with
111. Gaba RC, Lewandowski RJ, Kulik LM, Riaz A, Ibrahim SM, a focus on the middle hepatic vein anastomoses in normal human
Mulcahy MF, et al. Radiation lobectomy: preliminary findings of livers: anatomical study on liver corrosion casts. Surg Radiol Anat
hepatic volumetric response to lobar yttrium-90 : SRA. 2014;36(3):231–7.
radioembolization. Ann Surg Oncol. 2009;16(6):1587–96. 126. Le Roy B, Perrey A, Fontarensky M, Gagniere J, Abergel A,
112. Vouche M, Lewandowski RJ, Atassi R, Memon K, Gates VL, Ryu Pereira B, et al. Combined preoperative portal and hepatic vein
RK, et al. Radiation lobectomy: time-dependent analysis of future embolization (biembolization) to improve liver regeneration be-
liver remnant volume in unresectable liver cancer as a bridge to fore major liver resection: a preliminary report. World J Surg.
resection. J Hepatol. 2013;59(5):1029–36. 2017;41(7):1848–56.
113. Kim RD, Kim JS, Watanabe G, Mohuczy D, Behrns KE. Liver 127. Hocquelet A, Sotiriadis C, Duran R, Guiu B, Yamaguchi T, Halkic
regeneration and the atrophy-hypertrophy complex. Semin Interv N, et al. Preoperative portal vein embolization alone with biliary
Radiol. 2008;25(2):92–103. drainage compared to a combination of simultaneous portal vein,
114. Padia SA, Lewandowski RJ, Johnson GE, Sze DY, Ward TJ, Gaba right hepatic vein embolization and biliary drainage in Klatskin
RC, et al. Radioembolization of hepatic malignancies: back- tumor. Cardiovasc Intervent Radiol. 2018;41(12):1885–91.
ground, quality improvement guidelines, and future directions. J 128. Panaro F, Giannone F, Riviere B, Sgarbura O, Cusumano C,
Vasc Interv Radiol. 2017;28(1):1–15. Deshayes E, et al. Perioperative impact of liver venous deprivation
115.•• Palard X, Edeline J, Rolland Y, Le Sourd S, Pracht M, Laffont S, compared with portal venous embolization in patients undergoing
et al. Dosimetric parameters predicting contralateral liver hyper- right hepatectomy: preliminary results from the pioneer center.
trophy after unilobar radioembolization of hepatocellular carcino- HepatoBiliary Surg Nutr. 2019.
ma. Eur J Nucl Med Mol Imaging. 2018;45(3):392–401 Helps
define relationship between healthy injected liver dose and
Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
maximal hypertrophy of untreated liver lobe after
tional claims in published maps and institutional affiliations.
radioembolization.

You might also like