metricas
covid
Buscar en
Annals of Hepatology
Toda la web
Inicio Annals of Hepatology Argentinian clinical practice guideline for surveillance, diagnosis, staging and...
Información de la revista
Vol. 19. Núm. 5.
Páginas 546-569 (septiembre - octubre 2020)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
4232
Vol. 19. Núm. 5.
Páginas 546-569 (septiembre - octubre 2020)
Clinical Practice Guidelines
Open Access
Argentinian clinical practice guideline for surveillance, diagnosis, staging and treatment of hepatocellular carcinoma
Visitas
4232
Federico Piñeroa,
Autor para correspondencia
fpinerof@cas.austral.edu.ar

Corresponding author.
, Mario Tannob, Gabriel Aballay Soterasc, Matías Tisi Bañad, Melisa Dirchwolfe, Eduardo Fassiof, Andrés Rufe, Silvia Mengarellig, Silvia Borzih, Nora Fernándezi, Ezequiel Ridruejoa,j, Valeria Descalzik, Margarita Andersl, Guillermo Mazzolinia, Virginia Reggiardoc, Sebastián Marcianom, Florencia Perazzoi, Juan Carlos Spinam, Lucas McCormackl, Martín Maraschion..., Cecilia Laguesa, Adrián Gadanom, Federico Villamili, Marcelo Silvaa, Fernando Cairoi, Beatriz Ameigeiraso, the Argentinean Association for the Study of Liver Diseases (A.A.E.E.H) Ver más
a Hepatology and Liver Unit, Hospital Universitario Austral, School of Medicine, Austral University, B1629HJ Buenos Aires, Argentina
b Hospital Centenario de Rosario, Santa Fe, Argentina
c Hospital Argerich, Ciudad de Buenos Aires, Argentina
d Internal Medicine and Epidemiology Department, Hospital Universitario Austral, School of Medicine, Austral University, B1629HJ Buenos Aires, Argentina
e Hospital Privado de Rosario, Santa Fe, Argentina
f Hospital Alejandro Posadas, Buenos Aires, Argentina
g Hospital San Roque, Córdoba, Argentina
h Instituto Rossi, La Plata, Buenos Aires, Argentina
i Hospital Británico, Ciudad de Buenos Aires, Argentina
j Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Ciudad de Buenos Aires, Argentina
k Fundación Favaloro, Ciudad de Buenos Aires, Argentina
l Hospital Alemán, Ciudad de Buenos Aires, Argentina
m Hospital Italiano de Buenos Aires, Argentina
n Hospital Privado de Córdoba, Argentina
o Hospital Ramos Mejía, Ciudad de Buenos Aires, Argentina
Ver más
Este artículo ha recibido

Under a Creative Commons license
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (2)
Tablas (5)
Table 1. Quality of evidence and level of recommendations for HCC prevention, surveillance, diagnosis and staging.
Table 2. Quality of evidence and level of recommendations for HCC locoregional therapies, liver resection and liver transplantation.
Table 3. Quality of the evidence and level of recommendations for HCC endovascular and systemic therapies.
Table 4. Radiological tumor response assessment, management of adverse events and dose scheme for HCC systemic therapies.
Table 5. Comparison between A.A.E.E.H and other international guidelines.
Mostrar másMostrar menos
Material adicional (2)
Abstract

The A.A.E.E.H has developed this guideline for the best care of patients with hepatocellular carcinoma (HCC) from Argentina. It was done from May 2018 to March 2020. Specific clinical research questions were systematically searched. The quality of evidence and level of recommendations were organized according to GRADE. HCC surveillance is strongly recommended with abdominal ultrasound (US) every six months in the population at risk for HCC (cirrhosis, hepatitis B or hepatitis C); it is suggested to add alpha-feto protein (AFP) levels in case of inexeperienced sonographers. Imaging diagnosis in patients at risk for HCC has high specificity and tumor biopsy is not mandatory. The Barcelona Clinic Liver Cancer algorithm is strongly recommended for HCC staging and treatment-decision processes. Liver resection is strongly recommended for patients without portal hypertension and preserved liver function. Composite models are suggested for liver transplant selection criteria. Therapies for HCC with robust clinical evidence include transarterial chemoembolization (TACE) and first to second line systemic treatment options (sorafenib, lenvatinib, regorafenib, cabozantinib and ramucirumab). Immunotherapy with nivolumab and pembrolizumab has failed to show statistical benefit but the novel combination of atezolizumab plus bevacizumab has recently shown survival benefit over sorafenib in frontline.

Keywords:
Liver cancer
Practice guideline
Latin America
Argentina
Abbreviations
AFP

alpha-feto protein

AFP-L3

lens-culinaris AFP

ALT

alanine amino transferase

APHE

arterial phase hyperenhancement

AST

aspartate amino transferase

AUROC

area under the receiving operator curve

CI

confidence interval 95%

CR

complete response

CSPH

clinically significant portal hypertension

CT

computed axial tomography

DCP

desgamma carboxiprothrombin

DCR

disease control rate

DEB

drug-eluting beads

DFS

disease free survival

ECOG

Eastern Coperative Oncology Group

FDGF

fibroblast derived growth factor

HCC

hepatocellular carcinoma

HBV

hepatitis B virus

HCV

hepatitis C virus

HFSR

hand-foot skin reaction

HR

hazard ratio

irAEs

immune-related adverse events

LR

likelihood ratio

LR

liver resection

LSM

liver stiffness measure

LT

liver transplantation

MRI

magnetic resonance image

NAFLD

non-acoholic fatty liver disease

OR

odds ratio

ORR

objective response rate

PD

progressive disease

PDGF

platelet derived growth factor

PEI

percutaneous ethanol injection

PR

partial response

RR

relative risk

RRR

relative risk reduction

RA

absolute risk

RCT

randomized clinical trial

RECIST

Response Evaluation Criteria for Solid Tumors

RFA

radiofrequency ablation

RFS

recurrence free survival

Se

sensitivity

Sp

specificity

TACE

transarterial chemoembolization

TARE

transarterial radioembolización

TKI

tirosin kinase inhibitor

TTP

time to progression

US

ultrasound

VEGF

vascular endothelial growth factor

Texto completo
1Introduction

With the advent of new diagnostic and treatment tools for hepatocelluar carcinoma (HCC) during the last decade, the Argentinian Association for the Study of Liver Diseases (A.A.E.E.H) has developed this clinical practice guideline. The aim of the A.A.E.E.H was to develop a simple clinical tool, not a rigid dogma, applicable to national general physicians, hepatologists, gastroenterologists, oncologists, radiologist, and surgeons, for the best care of patients with HCC.

2Materials and methods

This guideline has been focused on epidemiology, surveillance, diagnosis, staging and treatment for patients with HCC. It has been carried out by a multidisciplinary board from May to December 2018 and up-dated in March 2020. Teams were invited to develop a synthesis of specific clinical research questions (Appendix 1), which were structured according to Population, Intervention/Exposure, Comparison and Outcomes (PICO). The evidence was systematically searched using Medline-Pubmed (MESH terms) and other databases (Trip-database, Lilacs) including original articles; editorial comments, personal opinions, reviews among others, were excluded.

The quality of the evidence and level of recommendations followed the Grade of Recommendation Assessment, Development and Evaluation (GRADE), including quality of evidence (High, Moderate, Low and Very Low) [1,2] using other guidelines for randomized clinical trials (RCT) [3], systematic reviews [4] and observational studies [5]. The strength of each recommendation followed a) balance between risks and benefits, b) precision of estimations (95% confidence intervals –CI), c) values and preferences of the patients and d) costs of interventions. For strong recommendations this guideline uses the term “it is recommended” and for conditional or weak ones “it is suggested”[1] (Appendix 1).

3Evidenced-based statements3.1Epidemiology

HCC is an increasing public health issue around the world, being the fifth most common cancer and the second leading cancer-related death [6,7]. In Asian countries, 60% of HCC cases are attributable to chronic hepatitis B virus (HBV) infection and vaccination with anti-HBV has dramatically reduced the incidence of HCC [8]. On the contrary, chronic hepatitis C virus (HCV) infection, followed by chronic alcohol consumption are the leading causes of HCC, with increasing HCC incidence and mortality rates in Western populations [9]. An increasing etiology of HCC during the last decade is non-alcoholic fatty liver disease (NAFLD) [10–15]. In Argentina, the HCC incidence rate is lower than 5.6 cases per 100.000 persons/year, equivalent to intermediate rates around the world [6].

This cancer originates in a cirrhotic liver or in the context of chronic liver injury in more than 90% of the cases [6,16]. In North America, HCV and alcoholic liver disease have been historically the most frequent causes of HCC but more recently, NAFLD has become a leading cause of HCC [10–12,17,18]. In Latin America, the most frequent etiology of HCC is HCV, followed by alcohol consumption [19,20]. Regional disparities have been reported, HBV in Brazil [21] and NAFLD in Chile. In Argentina, main etiologies are HCV in transplant centers and alcoholic liver disease in non-transplant centers [22,23]; whereas NAFLD has become an increasing etiology [23].

3.1.1Population at risk and HCC prevention

An estimated threshold of high risk for HCC was established in 1.5%/year [14,15]. Patients with cirrhosis of any etiology, or with chronic HBV or HCV infection are those with the highest risk of developing HCC [14,15]. Persistence of chronic infection [24–27], the degree of liver fibrosis and inflammation [28,29] and the presence of clinically significant portal hypertension (CSPH) are risk factors for HCC in HCV patients [30]. The risk of HCC decreases but it is not completely eliminated following HCV eradication, particularly in patients with advanced fibrosis (F3 or F4) [31–35] and CSPH [28,36].

Oncogenic pathways associated with HBV are independent from fibrosis grades. Presence of “e” antigen (Age), persistence of chronic inflammation, higher viral load in Age negative, pre-core mutants and the presence of advanced fibrosis have all been associated with higher risk [37]. On the contrary, asymptomatic carriers might be at lower risk, unless they present advanced fibrosis [38]. The REVEAL study cohort, including Age negative cohort, showed increasing HCC risk with higher viral loads [37]. Quantification of AgS levels might be another tool in Age- patients with lower viral load (<2000UI/ml) [39,40]. On the other hand, genotype C and pre-core mutants have been associated with higher risk of HCC [41]. More recently, values of liver stiffness measurement (LSM) have been associated with increasing Hazard Ratios (HR) of HCC, particularly with >18kPa HR 5.5 (CI 1.5;20.0) and >23kPa HR 6.6 (CI 1.8;23.8) [42]. Finally, some scoring models assessing the risk of HCC in treated patients with entecavir/tenofovir have been published, including the REACH (age, ALT levels, viral load and Age status) [43] and the PAGE score (age, gender and platelet count) [44]. A PAGE score of ≤9 points in non-cirrhotic HBV patients has a high negative predictive value for HCC [45].

The risk of HCC increases with alcohol consumption, particularly in the context of advanced fibrosis grades [46,47]. Moreover, it is a co-factor of HCC for other chronic liver diseases [48–50]. The risk of HCC in non-cirrhotic NAFLD is not clearly defined [10,12,17,23,51].

-Primary Prevention. The most effective measure is the universal vaccination of HBV [8]. Other unspecific measures include biosecurity of health procedures, sexual protective measures, avoidance of recreational drugs and alcohol consumption. Also, promotion of healthy lifestyle habits to avoid obesity and metabolic syndrome are recommended.

-Secondary Prevention. The most effective measures are viral eradication in chronic HBV [45,52–55] and HCV [24–26,31–34], stopping alcohol consumption [48,50,56,57] and promoting healthy lifestyle habits [58,59]. Direct-acting antivirals have shown decreasing incidence of HCC in those achieving HCV eradication [31,34,35,60]. Treatment of HBV has been associated with a RRR of 51% for HCC in a metanalysis [53]. This effect was observed even in patients with cirrhosis [45], although the risk was not completely abolished [55]. The rate in which the risk of HCC decreases in patients who present treatment-related fibrosis reversion is uncertain [52,53]. Some metanalyses have shown that coffee consumption may decrease the risk of HCC in patients with chronic liver disease [61,62]. However, the low quality of the evidence does not support any strong recommendation.

-Tertiary Prevention. In HBV, viral load has been associated with higher HCC recurrence after ablative therapies or LR [63,64]; treatment with interferon alfa 2b following resection did not reduce HCC recurrence rate [65] but opposite results were observed using nucleotide/nucleoside analogs [66]. In HCV, interferon-based regimens following ablation or liver resection did not reduce HCC recurrence in two RCT [67,68] and controversial data has been reported following direct-acting antivirals [69,70]. A higher tumor progression rate was not observed in HCV patients listed for liver transplantation (LT) treated with direct-acting antivirals [71,72] (Table 1).

Table 1.

Quality of evidence and level of recommendations for HCC prevention, surveillance, diagnosis and staging.

Topic  Statement  Grade of Recommendation  Quality of the evidence 
Primary Prevention  It is recommended universal vaccination for HBV, to reduce alcohol consumption, avoid illicit recreational drugs, to promote biosecurity and healthy life styles.  Strong  High 
Secondary PreventionIt is recommended to eradicate/treat HBV or HCV chronic infections, to stop alcohol abuse.  Strong  Moderate to high 
It is uncertain to recommend coffee consumption in patients with chronic liver disease.  Conditional  Low 
Tertiary PreventionIt is suggested to eradicate/treat HCV chronic infection after curative therapies.  Conditional  Low 
It is recommended to eradicate/treat HBV chronic infection after curative therapies.  Strong  Moderate 
SurveillanceIt is recommended for patients with cirrhosis and preserved liver function irrespective of etiology.  Strong  Low 
It is recommended for patients with decompensated cirrhosis listed for liver transplantation.  Strong  Low to very low 
It is recommended for patients with chronic HBV, previous family history of HCC1Strong  Moderate 
It is recommended for patients with chronic HCV and F>2.  Strong  Low 
It is suggested for any chronic liver disease with F>2.  Conditional  Very Low 
It is recommended after viral eradication of HBV or HCV in patients with F>2.  Strong  Low to moderate 
It is recommended to use abdominal ultrasound every 6 months; performed by expert sonographers.  Strong  Low to Moderate 
It is suggested to use alpha-feto protein (AFP) as additional tool for HCC surveillance (Fig. 1).  Weak  Low to very low 
DiagnosisIt is recommended to perform HCC diagnosis either with CT or MRI when specific hallmarks are observed in the population at risk (APHE plus washout in delay portal phase).  Strong  High 
A tumor biopsy is not recommended for HCC diagnosis confirmation in cases with imaging hallmarks.  Strong  High 
It is recommended to include LIRADS in radiological reports as well as detailed descriptions.  Strong  Very low 
AFP or other biomarkers are not recommended for HCC diagnosis.  Strong  High 
PET CT is not recommended for HCC diagnosis, staging or prognosis.  Strong  Low to moderate 
StagingThe Barcelona Clinic Liver Cancer staging is recommended for staging and therapeutic clinical-decision-making.  Strong  High 
It is recommended to perform clinical-decision-making in the context of multidisciplinary working teams.  Strong  Low 
It is not suggested to support BCLC-B sub-staging classification.  Conditional  Low to very low 

Notes: 1 – Among patient with chronic HBV, in the absence of fibrosis grade >2 or family history, it is suggested to assess risk of HCC and need for surveillance using the PAGE-B score. A PAGE-B score ≤9 points can exclude patients for surveillance.

Abbreviations: APHE: arterial phase hyper-enhancement; CT: computed axial tomography; F: fibrosis; MRI: magnetic resonance image.

3.2Surveillance for HCC3.2.1Rational and evidence supporting HCC surveillance

Surveillance is the sequencing or repetitive application of a cost-effective screening tool on a population at risk for a specific health problem, aiming at reducing its mortality rate avoiding the so-called lead and length-time bias. Surveillance failure is not the same as screening failure. Whereas the first one refers to the failure of any procedure associated with the surveillance program (e.g. non adherence), screening failure refers to false-negative tests associated with each screening tool.

Among HBV patients, the evidence comes from prospective observational studies [73,74] and three RCT with different risk of bias [75–77]. In cirrhosis, the evidence is even less robust [78–84]. Some metanalyses with significant heterogeneity [85,86] and Markov modeling approaches have evaluated its cost-effectiveness [87–92]. Despite these unrobust data, it is strongly recommended to perform HCC surveillance in the population at risk (Table 1).

This panel considers that the aim of screening tool is to detect a single HCC of <3cm because it is associated with higher likelihood for curative therapies and better survival [78,93–99]. Screening tools for HCC that have been explored are abdominal ultrasound (US), CT or MRI scans and different biomarkers.

3.2.2Comparison of screening tools for HCC

US is the most cost-effective screening tool [100,101]; though it has low sensitivity (range 30–70%), it is operator dependent and a negative test does not completely rule out HCC. On the contrary, it has elevated specificity (>90%). US accuracy should be analyzed within the framework of serial repetition over time, ideally a 6-month interval [96,99]. In this context, a 34% rate of US screening failure is expected, higher when performed by inexperienced operators [98] or inconsistent repetition [97,98,102]. In Argentina the rate of “surveillance failure” has been reported to be 42% [103]; whereas “screening failure” was observed between 26–32% of the cases [103].

The use of alpha-fetoprotein (AFP) alone or in combination with abdominal US has been less effective for detection of single <3cm HCC [75,79,96,99]. There is lack of consensus on AFP thresholds with different sensitivities (Se) and specificities (Sp): AFP >20ng/ml: Se 60% Sp 90%, AFP >100ng/ml Se 31% Sp 98.8%, AFP >200 Se 22% Sp 99.4%, AFP >400 Se 17% Sp 99% [79]. The panel suggests to include AFP values at different thresholds to avoid US screening failure by inexperienced operators [98] (Fig. 1). Other biomarkers including lens-culinaris fraction of AFP (AFP-L3) and desgamma carboxiprothrombin (DCP) have not been better than AFP as screening tools either alone or in combination with US [104–106].

Fig. 1.

Surveillance and diagnostic algorithm for hepatocellular carcinoma.

Note: See additional information in Appendix 2.

(0,34MB).
3.3Diagnosis of HCC based on imaging and pathological features3.3.1Imaging HCC diagnosis

Imaging HCC diagnosis can be performed with high specificity and tumor biopsy is not mandatory in the population at risk [107–112]. Triphasic scans including arterial, portal and delayed phases, either with computed axial tomography (CT) or magnetic resonance imaging (MRI) accurately perform HCC diagnosis. Specificities range from 85% to 100% for liver nodules ≥10mm showing imaging hallmarks including homogenous (non-rim) arterial phase hyperenhancement (APHE) and washout (non-peripheral) in portal phase [107–112]. Typical hallmarks may not be present in all HCCs. The presence of enhacing pseudocapsule during the late phase or increasing diameter of pre-existing nodule with APHE are additional criteria [113]. Reported sensitivities for nodules >20mm of diameter are similar with CT 92% and MRI 95%; however, MRI has superior accuracy for nodules <20mm with sensitivity of 70% and specificity of 83% (67% and 76% for CT, respectively) [114,115].

Liver nodules <10mm are not specific for HCC even showing hallmarks in MRI or CT scans. APHE is more frequently observed in these tiny lesions, whereas washout may not be present in nodules between 10 and 20mm of diameter [115]. In this regard, MRI with hepatobiliary contrast agents (Gadoxetic acid) may be useful to rule out malignancy, but it may decrease HCC specificity [107,108,112,116] (Fig. 1).

Radiological reports should be described in detail and include the LI-RADS system (liver imaging reporting and data system) [108,117]. LI-RADS is not applicable to patients without cirrhosis, without chronic HBV or HCV, or with vascular liver disorders (e.g. Budd Chiari syndrome, portal vein thrombosis, cardiac congestion and diffuse nodular regenerative hyperplasia) [108,118]. LI-RADS categorizes liver nodules in definitely benign (LI-RADS 1), probably benign (LI-RADS 2), with intermediate probability of HCC (LI-RADS 3), probable HCC (LI-RADS 4), definitely HCC (LI-RADS 5), probably or definitely malignant but unspecific for HCC (LI-RADS M) and definite HCC with tumor in vein (LI-RADS-TIV) [130–133]. Although LI-RADS has been validated in retrospective cohort studies [119], it has not been validated in RCT (Table 1).

3.3.2Pathological HCC diagnosis

Pathological diagnosis of HCC should be reserved for unconfirmed imaging diagnosis through fine needle biopsy, core needle biopsy or explant pathological analysis (Fig. 1). Cytological criteria for probable HCC include the presence of moderate to high cellularity, polygonal cells with dense cytoplasm, prominent nucleolus and surrounding sinusoidal cells, absence of biliary ducts cells, among other features. Guided core needle biopsy should be cautiously done for subcapsular liver lesions due to the risk of bleeding or tumor seeding. Histological features of HCC include cells grouped in trabeculae of 3 or more cells, cytological atypia, vascular invasion and loss of reticulin trama. Nuclear grading and histological classification should be assessed according to Edmondson and Steiner and WHO 2010 criteria, respectively. There is no patognomonic immunohistochemistry but the presence of at least 2 out of 3 signs, Glypican 3 or Heat Shock Protein 70 or Glutamine Sintetase, has 60% sensitivity and 100% specificity for HCC [120]. Molecular evaluation of HCC may be used for research purposes.

3.3.3Biomarkers for HCC diagnosis and prognostic purposes

Serum AFP has been extensively studied as HCC biomarker. Although a threshold of 200ng/ml was shown to be the best cut-off [121], most of the patients show AFP values <20ng/ml at HCC diagnosis [80,104,122]. Also, AFP is not completely specific for HCC. For this reason, AFP is not considered a requisite for HCC diagnosis. Nevertheless, AFP has been associated with recurrence risk in patients receiving liver resection (LR) [123,124] or radiofrequency ablation (RFA) [125–127], with tumor progression following transarterial chemoembolization (TACE) [128–131] and as a prognostic marker with systemic therapies [132–134]. Extensive evidence has been published regarding AFP values as a selection tool for LT [135–138]. Other biomarkers have not been included in the daily practice except in Asian countries [104–106] (Table 1).

3.4Staging

Different staging algorithms have been proposed including the Okuda system [139], the GRETCH score [140], the CLIP (Cancer of the Liver Italian Program) [141], CUPI [142], JIS (Japan Integrated System) [143], HKLC (Hong-Kong Liver Cancer) [144] and the BCLC staging (Barcelona-Clinic Liver Cancer) [145].

The BCLC was originally proposed according to the best available evidence, was externally validated in cohort studies [146–148] and adopted in international guidelines (Fig. 2). It includes five different stages according to baseline prognostic variables and the best evidenced-based therapeutic approach. Very early HCC or BCLC stage 0 includes patients with Eastern Cooperative Oncology Group (ECOG) 0, preserved liver function and single HCC <20mm and the best approach is RFA or LR [149–152]. Early HCC or BCLC-A corresponds to patients with preserved liver function, ECOG 0-1 and tumors including (a) single lesion without limit on diameter and (b) up-to 3 nodules less or equal than 30mm. Either CSPH or total bilirubin level >1mg/dl are prognostic variables that preclude LR, whereas these patients are candidates for LT if remaining within transplant criteria [154,155]. Intermediate BCLC-B stage are those patients with ECOG 0-1, preserved liver function with or without evidence of portal hypertension and multinodular tumors or those beyond BCLC-A stage. TACE is the treatment of choice [156–158], whereas in particular cases TARE might be indicated. Advanced HCC or BCLC-C stage includes patients with preserved liver function, with or wihout evidence of portal hypertension, ECOG 0–2 and tumor progressing or not responding after 2 TACEs or those with macrovascular invasion or extrahepatic spread. First and second-line sequencing therapy has achieved a median survival above 26 months in this group of patients [158]. Finally, BCLC-D stage includes patients with unpreserved liver function with any tumor burden or patients with ECOG 2–3; in these patients best supportive care is the treatment of choice. In this latter group, LT is the treatment of choice only if tumor burden is within transplant criteria or other co-morbidities do not exclude them for LT. Preserved liver function should be addressed including biochemical (total bilirubin, prothrombin time, serum creatinine and albumin) and clinical assessment (presence of CSPH including ascites and its complications, hepatic encephalopathy among other complications associated with portal hypertension). Scoring models like Child–Pugh or MELD score may be additional tools [159].

Fig. 2.

Staging for hepatocellular carcinoma according to the Barcelona Clinic Liver Cancer (BCLC).

Note: This figure is adopted from its original and latest version from the BCLC. Some changes were done, particularly for liver transplant candidates regarding BCLC-D or BCLC-B (if these patients are selected through composite models or specific UCSF downstaging protocol+AFP <100ng/ml). Preserved liver is equivalent to Child Pugh A or absence of any of the following decompensation events: jaundice, ascites, hepatic encephalopathy, hepatorenal syndrome, variceal bleeding. ECOG: Eastern Cooperative Oncology Group (performance status).

(0,26MB).

It is important to underline that the BCLC is not a rigid algorithm and some patients may be treated with subsequent recommended therapy (“treatment stage-migration”) [160]. Moreover, dynamic changes over time on liver function before or after each treatment should be considered. Finally, it includes the feasibility of each treatment. Other sub-staging algorithms for BCLC-B have been proposed (score ITALICA, HKLC) [144,161,162] or those avoiding ascites or hepatic encephalopathy grades (ALBI grade score) [163]. Adherence to the BCLC algorithm has been reported between 40% and 60% around the world [164–169] and 53% in Argentina [103].

3.5Locoregional treatments for HCC3.5.1Locoregional-ablative therapies

The most frequently used locoregional ablative therapies include RFA, PEI and microwave ablation (MWA), either percutaneously or laparoscopicaly [127,170,171]. PEI consists of the injection of ethanol solution generating tumor coagulative necrosis [151,172,173]. RFA and MWA generate thermal tissue damage through radiofrequency or microwave mechanisms; the effect of MWA does not decrease when applied near blood vessels (as it happens with RFA). PEI's main limitation is tumor diameter and fibrosis septa and may be an alternative when RFA is unfeasible (e.g. subcapsular or peri-biliary or vascular lesions) [174]. RFA is more effective than PEI for local tumor control, HCC recurrence [173] and overall survival [151] with similar hospital stay and fewer adverse events [172]. In a metanalysis with moderate quality, RFA and MWA presented similar overall survival, recurrence rates and local tumor control but MWA presented fewer adverse events [175]. Other ablative techniques such as cryoablation [176], irreversible electroporation (IRE) [177] and stereotaxic body radiation therapy (SBRT) have been developed but evidence-based supporting the superiority over RFA is lacking [178–180]. The only RCT comparing the superiority of cryoablation over RFA was negative [178]. Table 2 shows level of recommendations and quality of the evidence.

Table 2.

Quality of evidence and level of recommendations for HCC locoregional therapies, liver resection and liver transplantation.

Topic  Statement  Grade of Recommendation  Quality of the evidence 
RFA, PEI and othersEither liver resection or RFA are recommended for patients with single tumor <2cm.  Strong  High 
If LR or RFA are not feasible, PEI or MWA (if costs are comparable) are suggested to be second options.  Conditional  Moderate 
Cryoablation is not recommended as first line option.  Strong  Moderate 
SBRT or IRE are not recommended as first line options but are uncertain as second options.  Conditional  Low to very low 
Liver resectionLR is recommended for single tumors, irrespective of diameter, absence of CSPH, preserved liver function  Strong  High 
RFA is recommended as an alternative option to LR, specifically for patients with up-to 3 tumors less than 3cm each in patients with comorbidities.  Strong  High 
Liver transplantationLT is recommended for BCLC-A patients or with decompensated cirrhosis (BCLC-D).  Strong  High 
Composite criteria (either AFP model or Metroticket 2.0) are suggested to optimize LT candidates selection.  Conditional  Moderate 
AFP >1000ng/ml is recommended as an exclusion independent criteria.  Strong  Moderate 
Bridging therapies in the waitlist period are recommended considering tumor burden, AFP >400ng/ml and expected waitlist times.  Conditional  Low to moderate 
It is suggested to individualize the benefits and risks of downstaging; UCSF plus AFP <100ng/ml is suggested to identify ideal candidates.  Conditional  Low to moderate 
“All-comers” are not recommended for downstaging.  Strong  Low to moderate 
There is uncertain quality of evidence to recommend any particular locoregional modality for bridging or downstaging procedures.  Conditional  Low 
Adjuvant systemic therapiesSorafenib is not recommended for reducing the risk of HCC recurrence after RFA or LR.  Strong  High 
There is no sufficient evidence to recommend adjuvant systemic therapies prior or after LT to avoid HCC recurrence.  Conditional  Very Low 
OtherIt is suggested to use MELD score <10 to select non-ideal LR candidates (with CSPH).  Conditional  Low to very low 
It is uncertain to suggest or recommend multimodal combination therapies for BCLC-A or B patients.  Conditional  Low to very low 
It is suggested to follow BCLC algorithm for HCC recurrence therapies after RFA or PEI or LR or other locoregional therapies.  Conditional  Low to very low 
There is insufficient evidence to recommend any specific therapy for HCC recurrence after LT.  Conditional  Very low 

Abbreviations: BCLC: Barcelona Clinic Liver Cancer; CSPH: clinically significant portal hypertension; IRE: irreversible electroporation; LR: liver resection; LT: liver transplantation; MELD: model for end-stage liver disease; PEI: percutaneous ethanol injection; RFA: radiofrequency ablation; SBRT: stereotaxic body radiation therapy.

3.5.2Liver resection

The selection of appropriate candidates for LR is mandatory. Presence of extrahepatic tumor, main portal trunk invasion or unpreserved liver function are absolute contraindications [181–183]. Patients with CSPH have a higher risk of liver decompensation [153,154,184–186], higher postoperative morbi-mortality and lower 5-year overall survival [HR 1.48 (CI 1.11;1.98)] [154,187,188], even if “minor resections” are done [187]. Not clinically evident CSPH (gastroesophageal varices, ascites, portosystemic shunts) should be assessed through manometry [153,184,185,189] or surrogate markers (splenomegaly >120mm with low platelet count <100,000mm–3) [124,186]. LSM may have a role as a surrogate of CSPH, a threshold of >20kPa has been proposed [190]. Finally, a minimum remnant liver volume of 40% is important to avoid post-operative liver decompensation and can be estimated through CT volumetric assessment or other metabolic tests [191]. The evidence does not suggest any direction in favor or against any specific modality (open vs. laparoscopic LR) [192,193] but anatomical resections have been associated with lower HCC recurrence rates [194].

In BCLC-0 or A stages, LR was associated with lower 5-year recurrence rates (63.8%) compared to RFA 71.7% or PEI 76.9% [150,197,198] but with longer hospitalization and a higher rate of serious adverse events [199]. Cost-effective analysis and Markov modeling have shown that for single HCC <2cm, RFA is as comparable as LR in overall survival. For single lesion BCLC-A, LR is more cost-effective and for BCLC-A with 2–3 nodules, both techniques had similar overall survival, RFA was more cost-efective but LR had lower recurrence rate [200]. Finally, multipolar RFA presented similar survival and recurrence rates for single HCC <40mm compared to LR [201].

Additionally, LR may be performed by expert surgeons in “non-ideal” candidates [182,185,186,189]. However, the quality of the evidence is low. A MELD score less or equal than 9 points may select candidates for minor resections (less than 3 liver segments) [182,185,186,189]. Other authors have proposed LR among BCLC-B patients but with a significant risk of bias [181–183,202]. Recently, a low to moderate quality metanalysis compared LR vs. TACE in BCLC-B/C patients [204]. In the only RCT included, median survival was significantly higher for LR (41 months vs. TACE 14 months) [204]. However, unbalanced co-interventions and an indefinite number of TACEs might have led to ischeamic injury and liver decompensation [204]. Additionally, LR was not superior to sorafenib in patients with macrovascular invasion [205] (Table 2).

3.5.3Management of HCC recurrence following ablative therapies and liver resection

HCC recurrence after locoregional therapies or LR ranges from 40% to 70% at 5 years, either intra or extrahepatic [149,206–208]. Pathology variables associated with a higher risk of recurrence are macro or microvascular invasion, satellites or undifferentiated tumors [152,208,209]. The quality of the evidence supporting either one or the other therapeutic modality for HCC recurrence is low. RFA or re-resection [206,210], “salvage” LT [211,212] or preventive LT or “abb initio”[152,208] have been proposed. Salvage LT has shown similar survival but lower recurrence rates vs. re-resection [212–214]. In spite of being novel approach, preventive or “abb initio” LT needs to be externally validated [152,208]. Several strategies and therapeutic modalities of adjuvance and neo-adjuvance have failed to show any clinical benefit [215], including sorafenib (STORM trial) [216]. Other on-going RCT including pembrolizumab (MK-3475, NCT03867084), nivolumab (CheckMate-9DX, NCT03383458), dendritic cells based therapy [217] and induced natural killer cells [218] have been reported.

3.5.4Locoregional-endovascular therapies

  • (a)

    Transarterial chemoembolization (TACE) consists in the instillation through arterial hepatic branches of a chemotherapeutic agent with lipiodol (e.g. doxorubicin or epirrubicin, among others) followed by arterial embolization either conventionally (e.g. alcohol or spongostan) or through microspheres or drug-eluting beads (DEB) [220,229]. TACE is the standard treatment for BCLC-B patients [155–157]. Neither chemoinfusion alone, “lipiodolization” nor bland embolization (without chemotherapeutic agent) have shown any survival benefit [155,157,221,222]. Some authors have questioned these results in low quality studies with significant risk of bias [223,224]. Eligible candidates for TACE are those with preserved liver function, with a tumor diameter <10cm, less than 50% of total liver involvement, without vascular or biliary tumoral or non-tumoral obstruction, without portosystemic shunts, impaired kidney function or other major comorbidities [220]. Main non-tumoral portal trunk obstruction precludes conventional TACE but it may be done with hyperselective TACE or with DEB.

    In one RCT, TACE with DEB of 300–500μm was not superior to conventional TACE (PRECISION V trial) in overall survival and tumor control [220]. Systemic adverse events were lower in Child–Pugh B >7 patients with DEB [220]. Other uncontrolled studies have shown median survival of 47 months with DEB [225]. Nevertheless, there is lack of evidence comparing conventional TACE vs. smaller DEB (75–100μm).

    The most common adverse event is the post-chemoembolization syndrome, ocurring in 20% of the patients (pain, fever, increasing liver enzymes). Less frequently observed are those associated with the chemotherapeutic agent such as alopecia, mucositis, bone marrow suppression or cardiac dysfunction (more frequently observed with conventional TACE) [220].

  • (b)

    Transarterial Radioembolization (TARE) consists of the endovascular administration of Ytrium-90 (Y-90) radiospheres selectively within the tumor [226]. In some patients with unresectable large tumors (>7cm) or with portal vein thrombosis, TACE maybe not feasible or efficient enough. In these cases, TARE may be more radical with less therapeutic sessions. Ideal candidates are unresectable BCLC-A or B, with preserved liver function, total bilirubin <2mg/dl, absence of extrahepatic metastasis and with up-to 5 HCC liver lesions, a total diameter <20cm or single lesion <10cm, with or without portal vein obstruction [226–232]. Before TARE, other procedures such as arterial liver angiography, a Tecnecium 99 (Tc-99) scan to evaluate intrahepatic-pulmonary shunts (if >15%, TARE is not recommended) or the equivalent to lung exposure of no more than 30Gy for 1 session or 50Gy for >1 sessions should be evaluated [226–232].

    TARE is not associated with a post-embolization syndrome; the most serious adverse event is the Radioembolization Induced Liver Disease or “REILD”; occurring in less than 10% of the patients, within 4 to 8 weeks after TARE including jaundice, ascites and liver decompensation. Radiotoxicity may affect other abdominal organs [226,232].

  • (c)

    Radioembolization vs. TACE. The evidence comparing these techniques is of low to very low quality [227–232]. Two metanalyses with high statistical heterogeneity and significant risk of bias have shown that TARE may be associated with better 1-year survival rate [HR 0.74 (CI 0.61–0.90)], similar time to progression (TTP), shorter length of hospitalization and fewer adverse events [234,235]. However, a small pilot randomized, open-label, controlled trial (SIRTACE) showed no significant differences in 1-year overall survival (TARE 46.2% vs. TACE 66.7%), TTP and rate of adverse events [236]. Other small randomized trials showed longer TTP in favor of TARE but without a significant survival benefit [237,238]. There is an on-going phase III RCT (TRACE trial; NCT01381211) (Table 3).

    Table 3.

    Quality of the evidence and level of recommendations for HCC endovascular and systemic therapies.

    Topic  Statement  Grade of Recommendation  Quality of the evidence 
    TACEIt is recommended as first line option in BCLC-B patients.  Strong  High 
    DEB TACE is not superior to conventional TACE1  Strong  High 
    DEB TACE is suggested for Child Pugh B <9  Conditional  High 
    TAREInsufficient evidence to recommend or suggest TARE over TACE as first option for BCLC-B patients  Conditional  Low to very low 
    In some patients with unresectable large tumors, with portal vein obstruction TARE may have a therapeutic role.  Conditional  Low 
    It is uncertain to recommend or suggest TARE after TACE failure in BCLC-B.  Conditional  High 
    TARE is not recommended for BCLC-B patients with tumor progression or BCLC-C patients (with vascular invasion) over sorafenib.  Strong  High 
    Systemic therapies with TACE/TARE/otherSorafenib or other TKI agents in combination with TACE for BCLC-B patients are not recommended to avoid tumor progression.  Strong  High 
    There is no recommendation to support the combination of TARE with sorafenib for BCLC-B patients to avoid tumor progression.  Strong  High 
    Intra-hepatic arterial chemotherapy in combination with sorafenib is not recommended for BCLC-B or C patients  Strong  High 
    First line systemic therapyThe combination of atezolizumab plus bevacizumab is recommended as first-line systemic therapy2,3  Strong  High 
    If atezo+beva is not available or approved: sorafenib or lenvatinb are first-line systemic options4  Strong  High 
    Nivolumab is not recommended for first-line systemic therapy.  Strong  High 
    Second line systemic therapyRegorafenib, cabozantinib and ramucirumab are recommended second-line systemic therapies for post-sorafenib progression.  Strong  High 
    Cabozantinib or ramucirumab are recommended for intolerant-sorafenib patients.  Strong  High 
    Ramucirumab is only recommended for patients with AFP values ≥400ng/ml.  Strong  High 
    Pembrolizumab may be use in patients with AFP values <400ng/ml, without main portal vein invasion (tolerant or intolerant to sorafenib).  Conditional  High 
    Nivolumab may be suggested as second line (tolerant or intolerant to sorafenib).  Conditional  Moderate 
    There is lack of robust evidence supporting second line options after lenvatinib but these options may be use as for sorafenib.  Conditional  Very Low 
    Third line systemic therapyCabozantinib is recommended as third line (only with prior sorafenib plus other systemic therapy).  Strong  High 
    There is lack of robust evidence supporting any other third line options but some first or second-line agents may be used.  Conditional  Very Low 

    Notes: 1 – Only one RCT compared conventional TACE vs. DEB using large microspheres; uncertain efficacy regarding tiny microspheres. 2- Waiting for approval. 3-Without main portal trunk invasion or any prior autoimmune disease or uncontrolled arterial hypertension for atezolizumab+bevacizumab. 4- Without main portal trunk invasion or no more than 50% liver involvement or uncontrolled arterial hypertension for lenvatinib.

    Abbreviations: BCLC: Barcelona Clinic Liver Cancer; DEB: drug eluting beads; TACE: transarterial chemoembolization; TARE: transarterial radioembolization.

  • (d)

    Radioembolization vs. systemic therapy. Two phase III trials have compared the superiority of TARE over sorafenib in BCLC-B upon tumor progression or failure following sequential TACE and BCLC-C without extrahepatic disease (SARAH and SIRveNIB) [239,240]. Both showed negative results in terms of survival with median survival of 8.0 months for TARE and 9.9 months for sorafenib [HR of 1.17 (CI 0.94;1.41)] (SARAH). Median TTP was similar and although there was a lower rate of intrahepatic tumor progression with TARE, a lower and similar disease control rate (DCR) vs. sorafenib was observed in SARAH and SIRveNIB, respectively [239,240]. A similar rate of all adverse events were observed in SARAH; although there was a higher incidence of grade ≥3 related adverse events with sorafenib (63% vs. TARE 41%) [239]. There was a higer rate of all adverse events as well as higher rate of serious adverse events with sorafenib in SIRveNIB [240]. Opposite results were observed with quality of life, better in SARAH and similar in SIRveNIB [239,240]. Some differences between studies should be underlined. A larger proportion of BCLC-C patients were included in SARAH and total bilirubin was up-to 3mg/dl vs. <2mg/dl in SIRveNIB. In SARAH there were significant cross-overs, un-standardized radiation dose and inexperienced participating centers [239]; whereas there were logistical issues reported in SIRveNIB [240]. In both trials, the median treatment duration with sorafenib was shorter than expected [241,242] (Table 3).

3.5.5Locoregional-endovascular therapies in combination with systemic treatment

Different combinations of TACE with antiangiogenic drugs have been explored aiming at better TTP or disease-free survival (DFS) and in some of them, overall survival. The rationale for this combination is that higher expression of vascular endothelial growth factor (VEGF) following TACE was associated with worse survival [243].

Studies evaluating sorafenib+TACE with TTP as primary outcome include two non-randomized phase II trials (SOCRATES and START trials) [244,245] and three phase III RCT, the double-blinded SPACE trial [246], HELviCA study [247] and the open-label TACTICS trial [248]. The SOCRATES study enrolled patients with unpreserved liver function reporting higher adverse events [244]; whereas the combination was tolerable in patients with preserved liver function (START trial) [245]. Both studies used conventional TACE; DEB was used in another uncontrolled phase II trial [249]. In SPACE and HELviCA trials, the combination of TACE with DEB plus sorafenib did not show better TTP than TACE alone+placebo [247,247]. On the contrary, a longer DFS was observed in the TACTICS trial [median TTP 25.2 months vs. 13.5 months; HR 0.56 (CI 0.38;0.83)] [248]. These opposite results may be due to different elegibility criteria and different definition of tumor progression between trials. However, in both trials the combination showed a longer time to extrahepatic progression. The STAH trial evaluated overall survival as primary outcome with negative results but it included BCLC-C patients [250].

Other antiangiogenic agents in combination with TACE were negative including brivaniv (BRISK-TA phase III RCT) [251], bevacizumab [252] and axitinib [253]. More recently, intra-arterial chemotherapeutic agents in combination with sorafenib have also failed to show any survival benefit vs. sorafenib alone [254]. In another RCT including BCLC-C patients with main portal trunk invasion, the combination of intra-hepatic arterial infusion of FOLFOX plus sorafenib vs. sorafenib alone showed promising survival but with higher myelotoxicity [255]. Finally, the combination of sorafenib plus TARE did not show any survival benefit (SORAMIC trial) [242,256]. There are on-going trials exploring the TACE plus immunotherapy with nivolumab (IMMUTACE NCT03572582 and NCT03143270) (Table 3).

3.6Liver transplantation3.6.1Transplant eligibility criteria and outcomes following LT

Taking into account that it has the potential to cure not only HCC but also the underlying disease, LT is the HCC treatment with the highest survival probability. As it deals with donor scarcity and social justice, it should only be offered to patients with a minimum 60% 5-year survival rate. Moreover, HCC recurrence, occuring in approximately 10–30% of the cases, is the most frequent cause of death after LT in these patients [257,258]. Thus, to select the best candidates with the lowest risk of HCC recurrence is another main transplant end-point.

Although Milan criteria (single nodule up-to 5cm or up-to 3 nodules none of them more than 3cm in diameter) have been the gold standard for transplant eligibilty, some issues should be addressed [259,260]. On the one hand, tumor burden based only on number and diameter has a 25% discordance compared to explant pathology analysis [261–263]. On the other hand, other variables are associated with post-LT outcomes [136,264–269]. Different authors have proposed extended criteria beyond Milan including pre-LT AFP values as a biomarker for candidate selection [136,138,266–268] or histological pre-LT evaluation assessing tumor differentiation (Toronto criteria) [269]. Nevertheless, pre-LT histological evaluation is not always feasible [270].

The fact is that when extending criteria from beyond Milan without including AFP as a biomarker, higher HCC recurrence and lower post-LT survival have been reported [136,138,266–268,271]. AFP values are independently associated with post-LT survival and HCC recurrence and correlate with biologically aggressive tumor features at explant pathology [136,138,266–268,271]. Particularly, AFP levels >1000ng/ml have been proposed as a contraindication for LT [135]. Consequently, composite criteria have been recently proposed. Duvoux et al. included AFP values to number and tumor diameter (French AFP model: range 0 to 9 points with a threshold of up-to 2 points) [136]. This model was externally validated in Latin America and afterwards, in other regions around the world [271–273]. Other composite models included the sum of the largest diameter plus the total number of HCC nodules with the logarithmic transformation of AFP values (the Metroticket 2.0) [138]. Finally, dynamic AFP changes, as well as tumor progression in the waitlist period, should also be considered [274–277] (Table 2).

3.6.2Locoregional therapies as adyuvant treatment and “downstaging” before LT

Bridging” or adjuvant locoregional therapies before LT, either TACE or RFA/PEI among others, aim to avoid tumor progression and remain within LT criteria [277–279]. Tumor progression beyond Milan criteria is 7–11% at 6 months and 38% at 12 months of listing [72,265,275,276]. Variables associated with a higher risk of waitlist tumor progression are tumors beyond Milan criteria, AFP >400ng/ml and unfeasible or unresponsive to “bridging” therapies [275,276]. On the contrary, patients with complete response (CR) following locoregional treatments have lower rates of tumor progression and waitlist drop-out when compared to stable disease (SD) or progressive disease (PD) (1% vs. 10% at 12 months and 1% vs. 51% at 24 months) [275,280].

Downstaging” consists of reduction of tumor burden, either in tumor diameter or number, to values within Milan criteria [276,281–285]. However, there is no consensus on the “upper limits” beyond Milan for downstaging. Most popular LT downstaging protocol are those proposed by the University of California San Francisco (UCSF) (1 nodule >5cm but ≤8cm or 2–3 nodules all ≤5cm and sum of diameters ≤8cm or 4–5 lesions all ≤3cm and sum of diameters ≤8cm, plus AFP values below 1000ng/ml) [285,286]. There was a significant statistical heterogeneity in a metanalysis reporting the effect of “downstaging” upon survival [287]. On the one hand, effective “downstaging” was 48% (CI 39;58%), even lower in those beyond UCSF (“all-comers”) [288]. This latter group was associated with higher drop-out rate, lower probability of tumor reduction and worse outcomes after LT [288]. It seems that there should be a limit in tumor burden for “downstaging”. In this regard, AFP values below 100ng/ml were recently proposed to select better candidates within the UCSF protocol [289]. Also, a minimum observation period of 3 months to assess stable disease is required [290]. Finally, there is no robust data supporting the superiority of any therapeutic modality [178,287,291] (Table 2).

3.6.3Systemic treatment as adjuvant therapy before or after LT

There is no robust data in favor or against the use of sorafenib or other multikinase inhibitors, or other chemotherapeutic agents before or after transplantation as adjuvant therapy to reduce the risk of HCC recurrence [247,292–294]. Although immunosuppression with mTOR inhibitors was promising [295–297], there was neither survival nor recurrence benefit in a recent RCT [298].

3.6.4Surveillance and management of HCC recurrence after LT

The risk of HCC recurrence should be reassessed at explant pathology analysis. Presence of extensive tumor burden (beyond Up-to 7 criteria) [270], microvascular invasion, undifferentiated tumors or absence of complete necrosis are associated with higher recurrence risk [137,299,300]. Surveillance for HCC recurrence is controversial since it is considered an advanced stage with a high mortality rate independently from its location and there is no robust data to support any particular therapy. Moreover, most recurrences occur during the first 18 months after LT [137,257] and its earlier detection has been associated with worse survival [257,258]. Nevertheless, international consensus recommends surveillance for HCC recurrence with CT or MRI scans plus AFP values with a minimum interval of 6 months up-to the 3rd to 5th years following LT [299]. Locoregional therapies have been reported in retrospective studies for single intra or extrahepatic lesions [258,301], re-transplantation is strongly not recommended and in patients in whom locoregional therapies are not feasible, sequential systemic therapy has prolonged overall survival in retrospective studies [302–306]. No robust data supports any change in the type of immunosuppression [302–306] (Table 2).

3.7Systemic therapies for advanced HCC3.7.1First-line systemic treatment agents

During the last decade, an enormous improvement in the treatment of these patients has been achieved, with unthinkable survival rates years ago. Systemic conventional chemotherapeutic agents such as FOLFOX or GEMOX [307–309] or other agents as tamoxifen [157] have failed to show any survival benefit.

Sorafenib, a tyrosine multikinase inhibitor (TKI) was the first drug to show a survival benefit over placebo in two double-blind, phase III RCT (SHARP and Asia-Pacific) [241,310]. The exact mechanism of action is unknown with anti-angiogenic, hypoxic and antiproliferative effects. Eligibility criteria included preserved liver function, ECOG 0-1 and BCLC-C or BCLC-B under tumor progression [241,310]. Sorafenib (400mg bid) had a 70% relative risk reduction of death [HR of 0.69 (CI 0.55;0.87)] [241,310] and better TTP. However, radiological CR or PR were infrequently observed with a DCR of 43–53% [241,310]. Other real-world uncontrolled studies have been published so far [311], suggesting that sorafenib may be extended up-to Child–Pugh B7; however, lower survival and higher rate of adverse events with unpreserved liver function were reported [312–315].

Since then, other non-inferiority [e.g. sunitinib [316] and brivanib [317] or superiority trials [erlotinib [318], linifanib [319], dovitinib [320]] have failed to show any survival benefit or have shown excessive toxicity vs. sorafenib [bevacizumab+sorafenib [321]. More recently, lenvatinib showed non-inferiority (REFLECT trial) [322] and atezolizumab plus bevacizumab superiority over sorafenib (IMbrave trial, NCT03434379).

The REFLECT phase III, open-label RCT, showed non-inferior survival of lenvatinib (8mg day if <60kg or 12mg day if >60kg) vs. sorafenib [322]. This TKI blocks VEGF as well as FGF and PDGF pathways. In this trial, eligibility criteria excluded patients with main portal trunk tumor invasion and those with >50% of total liver volume involvement [322,323]. Median survival was 13.6 months with lenvatinib vs. 12.3 months with sorafenib [HR 0.92 (CI 0.79;1.06)] [322]. TTP, as well as higher rates of partial response and objective response rates (ORR) were observed with lenvatinib. Higher rates of severe adverse events were observed in lenvatinib arm (57% vs. 49%), mainly hypertension, hypothyroidism and proteinuria (Table 3). In Argentina, lenvatinib was approved in late 2018.

3.7.2Immunotherapy alone or in combination for first-line systemic therapy

Immunotherapy for cancer treatment has evolved into a complete novel paradigm. Cancer cells avoid lymphocyte T cell activation and proliferation, highly expressing “programmed cell death ligands” PD-1 ligand 1 and 2 (PD-L1, PD-L2) or “cytotoxic T lymphocyte protein 4” ligands (CTLA-4 and its ligands). Blockade of these unions through monoclonal antibodies (anti-PD-(L)1 or anti-CTLA-4) unblocks the anti-cancer immune host response (“immune check-point inhibitors”). The first anti-PD1 IgG4 monoclonal antibody was nivolumab. Later on, pembrolizumab (IgG4 anti-PD1), tremelimumab and ipilimumab (IgG1 anti CTLA-4) and atezolizumab, durvalumab and avelumab (IgG1 anti PD-L1).

Recently, the CheckMate-459 phase III, open-label RCT (NCT02576509) for first-line therapy in patients with advanced HCC naïve of systemic therapy, Child A, ECOG 0-1, BCLC-B or C in the absense of main portal trunk invasion did not show superior survival of nivolumab (240mg iv every 3 weeks) over sorafenib [median survival nivolumab 16.4 months vs. sorafenib 14.7 months; HR 0.85 (CI 0.72–1.02); P=0.07]. There was not a significant difference of DFS, even showing a higher rate of ORR in favor of nivolumab (15% vs. 7% with sorafenib). Nivolumab was associated with a lower rate of grade 3/4 adverse events (22% vs. 49%).

More recently, another phase III, open-label RCT, IMbrave-150 (NCT03434379) showed superiority of atezolizumab 1200mg iv (anti-PD-L1) plus bevacizumab 15mg/kg iv every 3 weeks vs. sorafenib [median survival nor reach vs. 13.2 months for sorafenib; HR 0.58 (CI 0.42–0.79); P=0.0006] [324]. Eligibility criteria included preserved liver function, systemic-naïve advanced HCC, ECOG 0-1 in the absense of main portal trunk invasion. Longer progression free survival (PFS) with a significant higher ORR rate in the combination arm 27% vs. 12% and DCR of 74% vs. 55% (P<.0001) were observed. Similar incidence of all grade adverse events and a lower incidence of grades 3/4 related adverse events were observed with the combination arm (36% vs. 46%). The most frequent adverse events were systemic hypertension, diarrhea, proteinuria, hyporexia, elevated liver enzymes and infusional reaction. However, treatment discontinuation was higher in atezolizumab+bevacizumab arm (16% and 10%, respectively) (Table 3).

Other combinations are being explored in phase III trials: pembrolizumab+lenvatinib vs. lenvatinib (LEAP-002, NCT03713593), sorafenib vs. durvalumab (anti PDL-1) vs. durvalumab+tremelimumab (CTLA-4) (HIMALAYA, NCT03298451), cabozantinib with/without atezolizumab vs. sorafenib (COSMIC-312; NCT03755791) and nivolumab+ipilimumab vs. sorafenib or lenvatinib (CheckMate-9DW; NCT04039607).

3.7.3Second-line systemic treatment agents

Tumor progression under sorafenib is the natural course of the disease in most of the patients and 10–20% of the patients may not tolerate this treatment. Thus, several second-line phase III trials have been explored after sorafenib progression or intolerance. Some agents failed to show any survival benefit vs. placebo including brivanib (BRISK-PS, phase III RCT) [325], axitinib (phase II trial) [326], everolimus (EVOLVE-1, phase III RCT) [327] and tivantinib (METIV, phase III RCT) [328].

The RESORCE trial included patients with advanced HCC, preserved liver function, with tumor progression and tolerant to sorafenib (a minimum dose of 400mg daily during the last month). Dose was scheduled in 160mg day in a 4 week-cycle (3 weeks ON-1 week OFF). Median survival was 10.6 months for regorafenib and 7.8 months for placebo [HR of 0.62 (CI 0.50;0.79)]; this survival benefit was observed in all stratified analyses even in patients with worse baseline prognosis (AFP >400ng/ml or macrovascular invasion) [329]. A benefit on TTP was also observed, with a significant difference in DCR (65% vs. 36%). Adverse events were common for TKIs (fatigue, hyporexia, hypertension, diarrhea and hand-foot skin reaction-HFSR); 46% were grade III and 4% grade IV, with a discontinuation rate of 10% [329]. In Argentina, it was approved during 2018.

The CELESTIAL trial compared cabozantinib 60mg/day vs. placebo [134] including unresectable BCLC-B or C patients, Child–Pugh A, ECOG 0-1, with previous sorafenib treatment (either tolerant or intolerant), with tumor progression after up-to 2 prior systemic therapies. Median survival was 10.2 months for cabozantinib and 8 months for placebo [HR of 0.76 (CI 0.63;0.92)]; this survival benefit was also observed in all post hoc analyses, even in patients with sorafenib as the only prior systemic therapy [HR 0.74 (IC 0.59–0.92)]. However, the efficacy of cabozantinib in tolerant vs. intolerant to sorafenib is uncertain. TTP was also in favor of cabozantinib with a DCR of 64% vs. 33%. Dose reductions and discontinuations were more common in cabozantinib arm, likewise for adverse events such as HFSR, asthenia and diarrhea [134].

The REACH I failed to show any survival benefit of ramucirumab (8mg/kg iv every 2 weeks), a monoclonal antibody that blocks VEGF receptors, vs. placebo. However, in a post hoc analysis there was a specific survival benefit among patients with baseline AFP values ≥400ng/ml, a group with an expected lower survival rate [133]. This motivated to performed the REACH II RCT including patients with histologically or cytologically confirmed HCC with AFP ≥400ng/ml, with prior sorafenib therapy – tolerant or intolerant – and tumor progression [330]. Median survival was 8.5 months for ramucirumab and 7.3 months for placebo [HR of 0.71 (CI 0.53;0.95)]. Stratified analyses confirmed the efficacy of ramucirumab in all groups but uncertain in prior-intolerant to sorafenib. TTP was longer and there were higher ORR and DCR rates over placebo (60% vs. 39%). Serious adverse events were similar between arms; ramucirumab presented more frequently hypertension (13%) and hyponatremia (6%) with a discontinuation rate due to adverse events of 11% [330]. Neither cabozantinib nor ramucirumab has been approved for HCC in Argentina (Table 3).

3.7.4Immunotherapy for second-line systemic therapy

Tremelimumab has been explored in an uncontrolled phase II trial in patients with HCV+; it was well tolerated with no HCV biochemical flares, even with decreasing HCV viral loads [331]. In another phase I/II, uncontrolled trial of nivolumab (CheckMate-040) in Child–Pugh A-B <9 patients under progression with sorafenib (tolerant or intolerant) [332]. There were escalating and expansion cohorts (3mg/kg iv every 3 weeks). The survival rate at 9 months was 74% (CI 67%;79%) with ORR of 20% and a DCR of 64%. PD-L 1 expression on tumor biopsies (≥1% vs. <1%) was not associated with a better survival but higher ORR was observed. Nivolumab showed a very good safety profile; common adverse events were rash, pruritus, diarrhea or colitis and liver or pancreatic enzymes elevation. This granted FDA temporary approval and motivated the CheckMate-459 at frontline (NCT02576509).

More robust data comes from a phase III, double-blind, RCT of pembrolizumab 200mg iv every 3 weeks vs. placebo (KEYNOTE-240) in patients under progression with sorafenib or intolerant to sorafenib, Child A, ECOG 0-1, BCLC B or C without main portal trunk invasion and AFP levels <400ng/ml [333]. The trial was negative in terms of survival benefit according to its null hypothesis alfa level; however, median survival was significantly longer for pembrolizumab vs. placebo [13.9 vs. 10.6 months, HR of 0.78 (CI 0.71–0.99); P=0.024]. DFS did not reach its primary efficacy endpoint too, although there was a higher ORR in favor of pembrolizumab (18.3% vs. 4.4%; P=0.00007), with a longer median duration of response. The incidence of immune-related adverse events (irAEs) was 18.3% with pembrolizumab, 7.2% grade 3/4 and in 3.6% motivated treatment discontinuation. The most frequent adverse events were liver enzymes elevation, fatigue, pruritus, hyporexia and diarrhea. In a stratified analysis, patients with AFP <200ng/ml or with post-progression (vs. intolerant to sorafenib) or with HBV had the highest benefit of pembrolizumab [333]. In Argentina, nivolumab and pembrolizumab were approved in late 2019 (Table 3).

3.8Clinical and radiological assessment after treatments for HCC

The main outcome of the treatment of HCC patients is overall survival. Secondary outcomes should be cancer erradication for curative therapies or delaying tumor progression with palliative therapies [334]. Liver decompensation events are competing events (jaundice, ascites, hepatic encephalopathy) leading to higher mortality and lower survival. These events are considered to be “untreatable progression”[335,336]. On the other hand, TTP or DFS are not surrogate markers of overall survival in patients with HCC; both are lower robust end-points with different risk of bias [291,317].

Target lesions” are those potentially treatable with at least APHE [337,338]; which should not be confounded with inflammatory signals. In cases of CR for curative therapies, imaging controls should be done every 3 months thereafter. Tumor response evaluation should be assessed with triphasic CT or MRI scans 4–6 weeks following TACE; particularly, with MRI after conventional TACE avoiding confounding effect with lipiodol. TARE response should be assessed not earlier than 8 weeks; with the best response usually occurring even after 6 months. Tumor re-assessment for systemic therapies is done every 8–12 weeks if second or third lines are available. Different imaging criteria to address tumor response have been proposed through the Response Evaluation Criteria for Solid Tumors (RECIST), including different versions (RECIST 1.0, RECIST 1.1), modified RECIST (mRECIST) [337,338]. Other criteria include the EASL or WHO [337,338]. mRECIST and EASL consider residual hyperenhacement diameters [337,338]. For locoregional therapies, mRECIST or EASL are preferable and for systemic therapies, RECIST 1.1 or mRECIST have been validated in RCT [339]. LIRADS tumor response assessment has not been validated in RCT [108,118]. Finally, evaluation after immunotherapy should consider that inflammatory infiltrates may increase tumor diameters at the very beginning (iRECIST).

3.8.1Clinical management after locoregional endovascular therapies

Although it has been shown that TACE number of sessions is directly proportional to tumor diameter [340] and CR has been associated with better survival [340], it is important to underline the risk of liver decompensation with unnecessary sequential TACEs when no response or tumor progression is observed, due to ischemic injury on the non-tumoral liver. Thus, it may be prudential to perform no more than 2 or 3 TACE sessions to achieve local tumor control and if not achieved, start systemic therapy [337,341]. Although the risk of liver ischemic injury with TARE is much lower, potential REILD should be evaluated.

Some scoring models have been published to identify candidates for sequential re-TACE, including the ART, STATE-ART and HAP scores [342–344]. However, significant risk of selection and information bias precluded these scores to be robust enough. Patients with “untreatable progression” are neither candidates for sequential TACE nor systemic therapy [335,336]. The rate of progression from Child–Pugh A to B or C following TACE was 13.8% and 15.9%, respectively (RELPEC cohort study) [345]. In Argentina, patients with “untreatable progression” following TACE had lower survival compared to those with sequential TACE-systemic therapy [311]. Data from first and second line RCT considered two intrahepatic tumor progression following TACE in BCLC-B as eligible for systemic therapy [241,317,322]. TARE as a rescue therapy following TACE failure was reported in the SARAH and SIRveNIB trials [240,241].

3.8.2Clinical management and radiological assessment after systemic therapies

Clinical evaluation and management of patients receiving systemic therapies should include “untreatable progression”, PD and tolerability. Any type of PD may not defined prognosis [134,329,330,332]. Whereas vascular invasion, new or growing extrahepatic lesions are prognostic markers, at least two consecutive new intrahepatic or growing intrahepatic lesions may be needed to start a second line [335].

Adverse events following TKIs should be addressed at every clinical visit; the need for dose reduction or treatment discontinuation. Intolerance to sorafenib was differently defined in the RESORCE and KEYNOTE-224 trials [329,333]. Treatment discontinuation rates were 9% with lenvatinib [322], 10% with regorafenib [329], 11% with sorafenib [241,310] and 16% with cabozantinib [134]. This comparison should consider different elegibility criteria and assessment of adverse events [346]. Most common adverse events for TKIs are fatigue, hyporexia, diarrhea, dermatologic events including rash and HFSR, hypertension, hypothyroidism, elevated liver enzymes, elevated total bilirubin and proteinuria [134,241,310,322,329]. Ramucirumab presented hypertension and hyponatremia [330]. Dermatological adverse events during the first 45 days of sorafenib therapy have been associated with better overall survival [314,347]. In general, grades I-II adverse event should prompt specific treatment and may continue with TKI dose until resolution of symptoms; grades III demand TKI reduction or transient interruption until resolution and grades IV definite treatment discontinuation [346]. Schemes of dose reduction according to RCT are shown in Table 4.

Table 4.

Radiological tumor response assessment, management of adverse events and dose scheme for HCC systemic therapies.

Topic  Statement  Grade of Recommendation  Quality of the evidence 
Radiological response assessmentmRECIST is recommended for post-locoregional therapies tumor reassessment.  Strong  Moderate to high 
RECIST 1.1 or mRECIST are recommended to assess tumor response under systemic treatments.  Strong  High 
There is no robust evidence to support tumor reassessment by LIRADS.  Strong  Low to very low 
Stopping rulesGrade 4 adverse events for any therapy.  Strong  High 
Unresolved grade 3 adverse events for any therapy.  Strong  High 
Grades 2 or higher for irAEs1  Strong  High 
“Untreatableprogression2 for locoregional and systemic therapies.  Strong  High 
Dosing schedules for systemic therapiesDose is fixed for atezolizumab 1200mg iv+bevacizumab 15mg/kg iv; cycles every 21 days may be postpone or interrupted for adverse events.  Strong  High 
Dose scheme reduction for sorafenib is 400mg bid to 400mg/day and 400mg every other day.  Strong  High 
Dose scheme reduction for lenvatinib depends or initial weight but it is 12mg/day to 8mg/day, 4mg/day to 4mg every other day.  Strong  High 
Dose scheme reduction for regorafenib is 160mg/day to 120mg/day and 80mg/day (ON-OFF cycles).  Strong  High 
Dose scheme reduction for cabozantinib is 60mg/day to 40mg/day and 20mg/day.  Strong  High 
Dose is fixed for ramucirumab 8mg/kg iv; cycles every 14 days may be postpone or interrupted for adverse events.  Strong  High 
Dose is fixed for pembrolizumab 200mg iv; cycles every 21 days may be postpone or interrupted for adverse events.  Strong  High 
Dose is fixed for nivolumab 3mg/kg or up-to 240mg; cycles every 14 days may be postpone or interrupted for adverse events.  Strong  High 

Notes: 1-Specific management for each irAE should be addressed with other practice guidelines. 2- Defined as jaundice, ascites, hepatic encephalopathy and other related complications (hepatorenal syndrome, among others).

Abbreviations: iv: intravenous infusion. irAE: immune-related adverse event.

Management of irAEs should be addressed following other international guidelines [348]. The irAEs have been reported in 83% of treated patients with less than 10% being grades 3 or 4 [332,333,349,350]. Although well-tolerated, irAEs could be life-threatening and demand a low threshold of suspicion [348]. Every organ can be involved, with a median occurrence between 3 to 6 months of initiation, earlier for anti-CTLA-4 compared to anti PD-(L)1 [348]. Most frequently involved organs are the skin (rash, pruritus), gastrointestinal (diarrhea or colitis) and the liver (hepatitis) [333]. Most of irAEs respond to steroid treatment and resolve between 6 to 8 weeks; steroid treatment has not been associated with lower efficacy in other tumors. However, steroids should be cautiously used in patients with cirrhosis due to a higher risk of infections or liver decompensation [351]. Recurrence of irAEs has been reported 55% of the cases, associated with time of occurrence with the first event [348]. Viral load for HBV before immunotherapy should be controlled with antivirals below 100 or 500UI/ml because of the risk of HBV flares [332].

3.9Special populations

Hepatocelluar carcinoma in non-cirrhotic or non-advanced fibrotic patients, mixed hepato-cholangio carcinoma and fibrolamellar HCC are special populations with low quality evidence to support for or go against any specific recommendations and these patients have been excluded from RCT. Although the BCLC could be applied, these patients should be discussed in multidisciplinary boards and LR should be the first-line option.

3.10Final considerations

With the coming new diagnostic and novel therapies for HCC, these patients will demand multidisciplinary team-working specifically trained on liver-related complications and HCC including hepatologist, gastroenterologists, liver surgeons, oncologists, radiologists, palliativies and pathologists [7]. Hepatologists have had a key role in research in this field [73,91,136,138,155,157,228,241,285,322,328]. Consequently, international associations and guidelines should not only underline the main role of hepatologists but also the need for multidisciplinary team-working [352–355]. Similarities and differences between this and other guidelines are described on Table 5. Differences between these practice guidelines with the former may be related to feasibility and availability of diagnostic and therapeutic modalities but also to cost-effective, and healthcare system barriers. Cost-effective analysis in countries with limited health resources should be done, as well as local patient's preferences. Approval of some interventions in these countries should be carefully addressed. For this reason, it is important to have local or regional data, analyzing access to health care [358,359]. In this clinical practice guideline, in 7.5% of the total included references, Argentinian colleagues participated as first authors, or as participating co-authors from international observational and RCT. However, the first authorship rate was 4.4%, this is a common problem in Latin America, a region where there is a low rate in the clinical setting [356–358].

Table 5.

Comparison between A.A.E.E.H and other international guidelines.

Topic/Guideline  AAEEH  AASLD [1]  EASL [2]  ALEH [3]  APASL [4] 
General organization  Consensus meeting 2015Key clinical questions.GRADE approach  Ten key clinical questions.GRADE approachPublished in 2018  GRADE approachPublished in 2018  GRADE approachPublished in 2014  Consensus meetingGRADE approachPublished in 2017 
EpidemiologyDefinition of the population at high risk for HCCDefinition of the population at high risk for HCC.Definition of the population at high risk for HCCDefinition of the population at high risk for HCCDefinition of the population at high risk for HCC.Special focus on HBV HCC
Primary, secondary and tertiary prevention detailed.  Primary, secondary and tertiary prevention detailed.  Primary and secondary prevention detailed.  Primary, secondary and tertiary prevention detailed. 
Strongly recommends treatment of liver disease to avoid progression of liver disease.HCV therapy after curative therapies for HCC: benefit vs. recurrence risk to be individualized.Uncertainty for recommending coffee consumption.  Strongly recommends treatment of liver disease to avoid progression of liver disease.HCV therapy after curative therapies for HCC: benefit vs. recurrence risk to be individualized.Strongly recommends coffee consumption.    Universal HBV vaccination in infants, specially HBV-endemic areas.Long-term antiviral HBV therapy as secondary prophylaxis.Recommends achievement of viral eradication for HCV but in high risk patients warrants surveillance.Further studies to assess risk and benefits of DAA after curative therapies for HCC
SurveillanceRecommends surveillance with US.Suggests surveillance with US with AFP in cases of unknown or inexpert US operators based on 3 AFP cut-off values.  Suggests surveillance with US with or without AFPSuggest not performing surveillance in ChildPugh C if not listed for LT.  Recommends surveillance with US.Does not recommend biomarkersRecommended in patients with preserved liver function or listed for LT.  Recommends US.  Recommends US with AFP (suggested cut-off 200ng/ml) or even a lower cut-off after HBV or HCV viral eradication.Suggests simultaneous use of biomarkers: AFP, AFP-L3 and DCP
Every 6 months.  Time-frame for surveillance every 4–8 months.  Every 6 months.  Every 6 to 12 months.  Every 6 months. 
Imaging Diagnosis  Imaging diagnosis strongly recommended either with triphasicCT or MRI.Suggests using LI-RADS in a specific algoritm.  Imaging diagnosis strongly recommended either with triphasicCT or MRIImaging diagnosis strongly recommended either with triphasic CT or MRI or contrast-US.PET-CT not recommended.MRI with hepatobiliary contrast agents included in algorithm but recalling lower specificity.  Imaging diagnosis strongly recommended either with triphasic CT or MRI  Imaging diagnosis strongly recommended either with triphasic CT or MRI or gadoxetic acid MRI for non-hypervascular nodules and contrast US. 
Tumor biopsy  Tumor biopsy for non-cirrhotic and indeterminate nodules.Maybe assessed after gadoxetic acid contrast MRI.Tumor biopsy only in non-cirrhotic patients.  Tumor biopsy conditional.Against routine biopsy of every indeterminate nodule.  Tumor biopsy only in non-cirrhotic patients.  Tumor biopsy in non-cirrhotic patients.  Tumor biopsy for non-cirrhotic and indeterminate nodules after gadoxetic acid contrast MRI
StagingBCLC staging.  BCLC not included.  BCLC staging.  BCLC staging.  Asia-Pacific treatment algorithm. 
Includes specific key question for LT candidates in BCLC-B and D stages.  Staging based on TNM.  “Not optimal surgical candidate” with portal hypertension may be done including extension of hepatectomy and MELD <10.     
Endovascular therapies  Strongly recommends TACE for BCLC-B patients.TACE with DEB not recommended over conventional TACE.Suggests evaluating TARE in patients with unresectable large tumors, with/without portal vein thrombosis (non-tumoral invasion).  Strongly recommends with TACE, TARE or external radiation over BSC for T2 or T3 HCC not candidates for surgery or LT.Does not recommend one form of therapy over another.  Strongly recommends TACE, either conventional or with DEB, over BSC.Does not recommend TARE or other therapy for intermediate HCCStrongly recommends TACE over BSC.TACE with DEB although “showing benefits over conventional TACE, not recommended due to higher costs”.  TACE, either conventional or with DEB, over BSC in multinodular, un-resectable and without extrahepatic or vascular disease HCCTARE or SBRT conditional options.Concept or “TACE failure/refractoriness” (2 or more consecutive ineffective responses or 2 or more intrahepatic progressions). 
Locoregional therapies  Either RFA or LR for very early HCC. PEI if RFA not feasible.Does not recommend cryoablation.Uncertain recommendation for SBRT or IRE.CSPH and preserved liver function recommended to select candidates for LR.  Suggests resection over RFA: for T1 or T2.  Either RFA or LR for very early HCCSuggests “resection” in non-ideal candidates: MELD <10.  Either RFA or PEI for very early HCC not suitable for surgery.  RFA first line for single <2cm, Child A or B.RFA as alternative for LR. PEI for cases in which RFA is not feasible in patients with up-to 3 nodules <3cm each. 
Liver Transplantation  Suggests using composite criteria for candidate selection.Suggests bridging therapies while on the waitlist period depending on tumor burden, AFP >400ng/ml and expected time on the waitlist.Suggests downstaging for patients beyond Milan criteria but with AFP <100ng/ml.  Suggest for T1 HCC listed for LT to undergo observation period.Suggests bridging therapies for T2 while on the waitlist period; without any specific treatment recommendation.Suggests downstaging for patients beyond Milan criteria within T3 stage.  Recommends Milan criteria for appropriate candidate selection but recalls for composite models including biomarkers.Suggests bridging therapies while on the waitlist period; without any specific treatmentWeak recommendation for downstagingRecommends Milan criteria for appropriate candidate selection.Conditional recommendation for downstaging.Recall for AFP cut-offs for LT selection.  Resectability vs. LT based on Child Pugh and “surgeons evaluation”. 
Adjuvance  Against adjuvant therapy after LR or ablation.  Against adjuvant therapy after LR or ablation.  Against adjuvant therapy after LR or ablation.  Not up-dated.   
Systemic treatment  Specific recommendations for available options in first-line (sorafenib, lenvatinb) and second line (regorafenib, pembrolizumab and nivolumab).Not yet approved: atezolizumab+bevacizumab in first-line and cabozantinib in second-line.Against TARE over sorafenib.  Strongly recommends systemic therapy over BSC for Child A or B in advanced HCC patients.No specific recommendation for first or second lines. Not up-dated.  Specific recommendations for first and second lines (sorafenib, regorafenib, cabozantinib).Against TARE over sorafenibNot up-dated.  Only sorafenib.Not up-dated.  Sorafenib for first line therapy.Regorafenib as second line.Not up-dated. 
Other remarks  Specific recommendations for adverse events, stopping rules and radiological response assessment.Recalls resection as main therapy in non-cirrhotic patients.Multidisciplinary team working.    Multidisciplinary team workingRecalls resection as main therapy in non-cirrhotic patients.  Recalls resection as main therapy in non-cirrhotic patients.LT may be offer in these patients.HCC in pediatric population.  Multidisciplinary team working. 

Notes:

[1] Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts LR, et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology 2018;67:358–80. doi:10.1002/hep.29086/suppinfo.

[2] Galle PR, Forner A, Llovet JM, Mazzaferro V, Piscaglia F, Raoul J-L, et al. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. Journal of Hepatology 2018;69:182–236. doi:10.1016/j.jhep.2018.03.019.

[3] Méndez-Sánchez N, Ridruejo E. Latin American Association for the Study of the Liver (LAASL) Clinical Practice Guidelines: Management of Hepatocellular Carcinoma. Annals of … 2014.

[4] Omata M, Cheng A-L, Kokudo N, Kudo M, Lee JM, Jia J, et al. Asia–Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update 2017:1–54. doi:10.1007/s12072-017-9799-9.

On the other hand, genetic regional variations should also be considered [357]. During the coming years, advancement of medical technology, based on novel genetic [357,359], epigenetic researches [360], as well as radiomics and artificial intelligence [361], may help to enhance surveillance, diagnosis and treatment response assessment in the clinical setting of HCC patients. In this regard, clinical practice guidelines should be appropriately updated [357].

Conflict-of-interest

F Piñero has received Advisory Board and speaker honoraria and he is consultant of Bayer Healthcare; has received speaker honoraria from Raffo and research grants from the Argentinean National Institute of Cancer (INC ID-190), Argentinean National Ministry of Science and Technology Development (PICT 2017, FONCYT) and from the Latin American Liver Research Educational and Awareness Network (LALREAN). G Aballay Soteras has received speaker honoraria and Advisory Board fees and research grants from Bayer Healthcare, MSD; he is consultant for Bayer and has received Advisory Board fees from Roche. S Marciano has received speaker honoraria from Gador, Gilead and Abbvie; has received educational grants from Gilead, Bristol-Myers Squibb, Bayer, Biotoscana and Gador and research grants from Gilead and Bristol-Myers Squibb. M Silva has received has received speaker honoraria and is a consultant for Abbvie, Gador, Bristol-Myers Squibb, Merck, Bayer and research grants from the Argentinean National Institute of Cancer (INC ID-190), Argentinean National Ministry of Science and Technology Development (PICT 2017, FONCYT). A Ruf has received Advisory Board and speaker honoraria and he is consultant of Bayer Healthcare, Novartis, Gador, Astellas and has received research grants from the Executive Secretariat of the Group of 77 at the United Nations. M Dirchwolf has received Advisory Board and speaker honoraria and he is consultant of Bayer Healthcare and has received a research grant from Novartis. M Tanno, M Tisi Baña, E Fassio, S Mengarelli, S Borzi, N Fernandez, E Ridruejo, V Descalzi, M Anders, G Mazzolini, V Reggiardo, JC Spina, L McCormack, M Maraschio, C Lagues, F Villamil, F Cairo and B Ameigeiras do not have any conflict of interest to disclose.

Acknowledgments

On behalf of those who have participated in the development of this clinical practice guideline: (1) General Coordination: F Piñero, M Tanno, G Aballay Soteras. (2) GRADE supportive board: F Piñero, M Tisi Baña. (3) Epidemiology: E Fassio, V Reggiardo, A Gadano, S Marciano. (4) Surveillance: S Mengarelli, M Dirchwolf, S Borzi. (5) Diagnosis: M Volpachio, J P Perotti, F Diaz Telli, J C Spina. (6) Staging: N Fernández, A Ruf. (7) Pathology: C Lagues, M Amante, MT García de Dávila. (8) Treatment: L McCormack, M Maraschio, E de Santibañes, G Podestá, E Ridruejo, V Descalzi, M Anders, F Villamil, G Eiselle, R García Mónaco, G Mazzolini, F Perazzo (9) Special populations: S Marciano, F Cairo, M Silva.

To the A.A.E.E.H 2018–2020 boards: Beatriz Ameigeiras, Diana Krasniansky, Fernando Cairo, Paola Casciato, Omar Galdame and Manuel Mendizabal.

Appendix A
Supplementary data

The following are Supplementary data to this article:

References
[1]
G.H. Guyatt, A.D. Oxman, G.E. Vist, R. Kunz, Y. Falck-Ytter, P. Alonso-Coello, et al.
GRADE: an emerging consensus on rating quality of evidence and strength of recommendations.
[2]
G.H. Guyatt, A.D. Oxman, R. Kunz, Y. Falck-Ytter, G.E. Vist, A. Liberati, et al.
Going from evidence to recommendations.
[3]
Schulz Altman Moher.
The CONSORT statement: revised recommendations for improving the quality of reports of parallel-group randomised trials.
[4]
D. Moher, A. Liberati, J. Tetzlaff, D.G. Altman, PRISMA Group.
Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement.
J Clin Epidemiol, 62 (2009), pp. 1006-1012
[5]
E. Elm von, D.G. Altman, M. Egger, S.J. Pocock, P.C. Gøtzsche, J.P. Vandenbroucke, et al.
The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies.
Lancet, 370 (2007), pp. 1453-1457
[6]
A.P. Venook, C. Papandreou, J. Furuse, L. Ladron de Guevara.
The incidence and epidemiology of hepatocellular carcinoma: a global and regional perspective.
[7]
Global Burden of Disease Liver Cancer Collaboration, T. Akinyemiju, S. Abera, M. Ahmed, N. Alam, M.A. Alemayohu, et al.
The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level.
[8]
M.-H. Chang, S.L. You, C.-J. Chen, C.-J. Liu, C.-M. Lee, S.-M. Lin, et al.
Decreased incidence of hepatocellular carcinoma in hepatitis B vaccines: a 20-year follow-up study.
JNCI J Natl Cancer Inst, 101 (2009), pp. 1348-1355
[9]
B. Njei, Y. Rotman, I. Ditah, J.K. Lim.
Emerging trends in hepatocellular carcinoma incidence and mortality.
Hepatology, 61 (2015), pp. 191-199
[10]
R.J. Wong, R. Cheung, A. Ahmed.
Nonalcoholic steatohepatitis is the most rapidly growing indication for liver transplantation in patients with hepatocellular carcinoma in the U.S..
Hepatology, 59 (2014), pp. 2188-2195
[11]
Z.M. Younossi, D. Blissett, R. Blissett, L. Henry, M. Stepanova, Y. Younossi, et al.
The economic and clinical burden of nonalcoholic fatty liver disease in the United States and Europe.
Hepatology, 64 (2016), pp. 1577-1586
[12]
V. Santi, D. Buccione, A. Di Micoli, G. Fatti, M. Frigerio, F. Farinati, et al.
The changing scenario of hepatocellular carcinoma over the last two decades in Italy.
J Hepatol, 56 (2012), pp. 397-405
[13]
K. Tokushige, E. Hashimoto, Y. Horie, M. Taniai, S. Higuchi.
Hepatocellular carcinoma in Japanese patients with nonalcoholic fatty liver disease, alcoholic liver disease, and chronic liver disease of unknown etiology: report of the nationwide survey.
J Gastroenterol, 46 (2011), pp. 1230-1237
[14]
K.A. McGlynn, J.L. Petrick, W.T. London.
Global epidemiology of hepatocellular carcinoma.
Clin Liver Dis, 19 (2015), pp. 223-238
[15]
P. Bertuccio, F. Turati, G. Carioli, T. Rodriguez, C. La Vecchia, M. Malvezzi, et al.
Global trends and predictors in hepatocellular carcinoma.
J Hepatol, 67 (2017), pp. 302-309
[16]
F.X. Bosch, J. Ribes, M. Díaz, R. Cléries.
Primary liver cancer: worldwide incidence and trends.
Gastroenterology, 127 (2004), pp. S5-S16
[17]
K. Tokushige, E. Hashimoto, Y. Horie, M. Taniai, S. Higuchi.
Hepatocellular carcinoma in Japanese patients with nonalcoholic fatty liver disease, alcoholic liver disease, and chronic liver disease of unknown etiology: report of the nationwide survey.
J Gastroenterol, 46 (2011), pp. 1230-1237
[18]
D. Kim, A.A. Li, B.J. Perumpail, C. Gadiparthi, W. Kim, G. Cholankeril, et al.
Changing trends in etiology- and ethnicity-based annual mortality rates of cirrhosis and hepatocellular carcinoma in the United States.
[19]
E. Fassio, S. Díaz, C. Santa, M.E. Reig, Y. Martínez Artola, A. Alves de Mattos, et al.
Etiology of hepatocellular carcinoma in Latin America: a prospective, multicenter, international study.
Ann Hepatol, 9 (2010), pp. 63-69
[20]
J.D. Debes, A.J. Chan, D. Balderramo, L. Kikuchi, E. Gonzalez Ballerga, J.E. Prieto, et al.
Hepatocellular carcinoma in South America: evaluation of risk factors, demographics and therapy.
Liver Int, 38 (2018), pp. 136-143
[21]
F.J. Carrilho, L. Kikuchi, F. Branco, C.S. Goncalves, A.A.de Mattos.
Clinical and epidemiological aspects of hepatocellular carcinoma in Brazil.
[22]
E. Fassio, C. Miguez, S. Soria, F. Palazzo, A. Gadano, R. Adrover, et al.
Etiology of hepatocellular carcinoma in Argentina: results of a multicenter retrospective study.
Acta Gastroenterol Latinoam, 39 (2009), pp. 47-52
[23]
F. Piñero, J. Pages, S. Marciano, N. Fernández, J. Silva, M. Anders, et al.
Fatty liver disease, an emerging etiology of hepatocellular carcinoma in Argentina.
World J Hepatol, 10 (2018), pp. 41-50
[24]
V. Virlogeux, P. Pradat, K. Hartig-Lavie, F. Bailly, M. Maynard, G. Ouziel, et al.
Eradication of hepatitis C virus infection and the development of hepatocellular carcinoma: a meta-analysis of observational studies.
Ann Intern Med, 158 (2017), pp. 1122-1127
[25]
H.B. El-Serag, F. Kanwal, P. Richardson, J. Kramer.
Risk of hepatocellular carcinoma after sustained virological response in Veterans with hepatitis C virus infection.
Hepatology, 64 (2016), pp. 130-137
[26]
N. Ganne-Carrié, R. Layese, V. Bourcier, C. Cagnot, P. Marcellin, D. Guyader, et al.
Association between sustained virological response and all-cause mortality among patients with chronic hepatitis C and advanced hepatic fibrosis.
JAMA, 308 (2012), pp. 2584-2593
[27]
Y. Hoshida.
Molecular epidemiology of hepatocellular carcinoma.
Clin Liver Dis, 1 (2013), pp. 177-179
[28]
A.S. Lok, L.B. Seeff, T.R. Morgan, A.M. Di Bisceglie, R.K. Sterling, T.M. Curto, et al.
Incidence of hepatocellular carcinoma and associated risk factors in hepatitis C-related advanced liver disease.
Gastroenterology, 136 (2009), pp. 138-148
[29]
R. Masuzaki, R. Tateishi, H. Yoshida, E. Goto, T. Sato, T. Ohki, et al.
Prospective risk assessment for hepatocellular carcinoma development in patients with chronic hepatitis C by transient elastography.
Hepatology, 49 (2009), pp. 1954-1961
[30]
C. Ripoll, R.J. Groszmann, G. Garcia-Tsao, J. Bosch, N. Grace, A. Burroughs, et al.
Hepatic venous pressure gradient predicts development of hepatocellular carcinoma independently of severity of cirrhosis.
J Hepatol, 50 (2009), pp. 923-928
[31]
G.N. Ioannou, P.K. Green, K. Berry.
HCV eradication induced by direct-acting antiviral agents reduces the risk of hepatocellular carcinoma.
J Hepatol, S0168-8278 (2017), pp. 32273
[32]
P. Nahon, R. Layese, V. Bourcier, C. Cagnot, P. Marcellin, D. Guyader, et al.
Incidence of hepatocellular carcinoma after direct antiviral therapy for HCV in patients with cirrhosis included in surveillance programs.
Gastroenterology, 155 (2018), pp. 1436-1450
[33]
L.S. Belli, G. Perricone, R. Adam, P.A. Cortesi, M. Strazzabosco, R. Facchetti, et al.
Impact of DAAs on liver transplantation: major effects on the evolution of in- dications and results. An ELITA study based on the ELTR registry.
J Hepatol, 69 (2018), pp. 810-817
[34]
A. Romano, P. Angeli, S. Piovesan, F. Noventa, G. Anastassopoulos, L. Chemello, et al.
Newly diagnosed hepatocellular carcinoma in patients with advanced hepatitis C treated with DAAs: a prospective population study.
J Hepatol, 69 (2018), pp. 345-352
[35]
F. Piñero, M. Mendizabal, E. Ridruejo, F. Herz Wolff, B. Ameigeiras, M. Anders, et al.
Treatment with direct-acting antivirals for HCV decreases but does not eliminate the risk of hepatocellular carcinoma.
Liver Int, 39 (2019), pp. 1033-1043
[36]
A.S. Lok, J.E. Everhart, E.C. Wright, A.M. Di Bisceglie, H.Y. Kim, R.K. Sterling, et al.
Maintenance peginterferon therapy and other factors associated with hepatocellular carcinoma in patients with advanced hepatitis C.
Gastroenterology, 140 (2011), pp. 840-841
[37]
C.-J. Chen, H.I. Yang, J. Su, C.-L. Jen, S.L. You, S.N. Lu, et al.
Risk of hepatocellular carcinoma across a biological gradient of serum hepatitis B virus DNA level.
JAMA, 295 (2006), pp. 65-73
[38]
J.D. Chen, H.I. Yang, U.H. Iloeje, S.L. You, S.N. Lu, L.Y. Wang, et al.
Carriers of inactive hepatitis B virus are still at risk for hepatocellular carcinoma and liver-related death.
Gastroenterology, 138 (2010), pp. 1747-1754
[39]
J.M. Lee, S.H. Ahn, H.S. Kim, H. Park, H.Y. Chang, D.Y. Kim, et al.
Quantitative hepatitis B surface antigen and hepatitis B e antigen titers in prediction of treatment response to entecavir.
Hepatology, 53 (2011), pp. 1486-1493
[40]
T.C. Tseng, C.J. Liu, H.C. Yang, T.H. Su, C.C. Wang, C.L. Chen, et al.
High levels of hepatitis B surface antigen increase risk of hepatocellular carcinoma in patients with low HBV load.
Gastroenterology, 142 (2012), pp. 1140-1149
[41]
H.I. Yang, S.H. Yeh, P.J. Chen, U.H. Iloeje, C.L. Jen, J. Su, et al.
Associations between hepatitis B virus genotype and mutants and the risk of hepatocellular carcinoma.
JNCI J Natl Cancer Inst, 100 (2008), pp. 1134-1143
[42]
K.S. Jung, S.U. Kim, S.H. Ahn, Y.N. Park, D.Y. Kim, J.Y. Park, et al.
Risk assessment of hepatitis B virus-related hepatocellular carcinoma development using liver stiffness measurement (FibroScan).
Hepatology, 53 (2011), pp. 885-894
[43]
H.I. Yang, M.-F. Yuen, H.L.-Y. Chan, K.-H. Han, P.-J. Chen, D.Y. Kim, et al.
Risk estimation for hepatocellular carcinoma in chronichepatitis B (REACH-B): development and validation of apredictive score.
Lancet Oncol, 12 (2011), pp. 568-574
[44]
G. Papatheodoridis, G. Dalekos, V. Sypsa, C. Yurdaydin, M. Buti, J. Goulis, et al.
PAGE-B predicts the risk of developing hepatocellular carcinoma in Caucasians with chronic hepatitis B on 5-year antiviral therapy.
J Hepatol, 64 (2016), pp. 800-806
[45]
G.V. Papatheodoridis, R. Idilman, G.N. Dalekos, M. Buti, H. Chi, F. van Boemmel, et al.
The risk of hepatocellular carcinoma decreases after the first 5 years of entecavir or tenofovir in Caucasians with chronic hepatitis B.
Hepatology, 66 (2017), pp. 1444-1453
[46]
P. Jepsen, P. Ott, P.K. Andersen, H.T. Sørensen, H. Vilstrup.
Risk for hepatocellular carcinoma in patients with alcoholic cirrhosis: a Danish nationwide cohort study.
[47]
F. Turati, C. Galeone, M. Rota, C. Pelucchi, E. Negri, V. Bagnardi, et al.
Alcohol and liver cancer: a systematic review and meta-analysis of prospective studies.
Ann Oncol, 25 (2014), pp. 1526-1535
[48]
H. Vandenbulcke, C. Moreno, I. Colle, J.-F. Knebel, S. Francque, T. Sersté, et al.
Alcohol intake increases the risk of hepatocellular carcinoma in patients with hepatitis C virus-related compensated cirrhosis: a prospective study.
[49]
R. Loomba, H.I. Yang, J. Su, D. Brenner, U. Iloeje, C.J. Chen.
Obesity and alcohol synergize to increase the risk of incident hepatocellular carcinoma in men.
Clin Gastroenterol Hepatol, 8 (2010), pp. 891-892
[50]
J.A. Marrero, R.J. Fontana, S. Fu, H.S. Conjeevaram, G.L. Su, A.S. Lok.
Alcohol, tobacco and obesity are synergistic risk factors for hepatocellular carcinoma.
J Hepatol, 42 (2005), pp. 218-224
[51]
F. Piscaglia, G. Svegliati-Baroni, A. Barchetti, A. Pecorelli, S. Marinelli, C. Tiribelli, et al.
Clinical patterns of hepatocellular carcinoma in nonalcoholic fatty liver disease: a multicenter prospective study.
Hepatology, 63 (2016), pp. 827-838
[52]
G.V. Papatheodoridis, Pietro Lampertico, S. Manolakopoulos, A. Lok.
Incidence of hepatocellular carcinoma in chronic hepatitis B patients receiving nucleos(t)ide therapy: a systematic review.
J Hepatol, 53 (2010), pp. 348-356
[53]
A.K. Singal, H. Salameh, Y.F. Kuo, R.J. Fontana.
Meta-analysis: the impact of oral anti-viral agents on the incidence of hepatocellular carcinoma in chronic hepatitis B.
Aliment Pharmacol Ther, 38 (2013), pp. 98-106
[54]
J.Y. Cho, Y.H. Paik, W. Sohn, H.U.C. Cho, G.-Y. Gwak, M.S. Choi, et al.
Patients with chronic hepatitis B treated with oral antiviral therapy retain a higher risk for HCC compared with patients with inactive stage disease.
[55]
G.-A. Kim, H.C. Lee, M.-J. Kim, Y. Ha, E.J. Park, J. An, et al.
Incidence of hepatocellular carcinoma after HBsAg seroclearance in chronic hepatitis B patients: a need for surveillance.
J Hepatol, 62 (2015), pp. 1092-1099
[56]
A. Mancebo, M.L.G. Diéguez, V. Cadahía, M. Varela, R. Pérez, C.A. Navascués, et al.
Annual incidence of hepatocellular carcinoma among patients with alcoholic cirrhosis and identification of risk groups.
Clin Gastroenterol Hepatol, 11 (2013), pp. 95-101
[57]
R. Loomba, H.I. Yang, J. Su, D. Brenner, U. Iloeje, C.J. Chen.
Obesity and alcohol synergize to increase the risk of incident hepatocellular carcinoma in men.
Clin Gastroenterol Hepatol, 8 (2010), pp. 891-892
[58]
C. Wang, X. Wang, G. Gong, Q. Ben, W. Qiu, Y. Chen, et al.
Increased risk of hepatocellular carcinoma in patients with diabetes mellitus: a systematic review and meta-analysis of cohort studies.
Int J Cancer, 130 (2011), pp. 1639-1648
[59]
D. Saunders, D. Seidel, M. Allison, G. Lyratzopoulos.
Systematic review: the association between obesity and hepatocellular carcinoma – epidemiologic evidence.
Aliment Pharmacol Ther, 31 (2010), pp. 1051-1063
[60]
M. Mendizabal, F. Piñero, E. Ridruejo, F.H. Wolff, M. Anders, V. Reggiardo, et al.
Disease progression in patients with hepatitis C virus infection treated with direct-acting antiviral agents.
Clin Gastroenterol Hepatol, (2020),
[61]
U. Gelatti, L. Covolo, M. Franceschini, F. Pirali, A. Tagger, M. Ribero, et al.
Coffee consumption reduces the risk of hepatocellular carcinoma independently of its aetiology: a case–control study.
J Hepatol, 42 (2005), pp. 528-534
[62]
F. Bravi, A. Tavani, C. Bosetti, P. Boffetta, C. La Vecchia.
Coffee and the risk of hepatocellular carcinoma and chronic liver disease.
Eur J Cancer Prev, 26 (2017), pp. 368-377
[63]
J.-C. Wu, Y.-H. Huang, G.-Y. Chau, C.-W. Su, C.-R. Lai, P.-C. Lee, et al.
Risk factors for early and late recurrence in hepatitis B-related hepatocellular carcinoma.
J Hepatol, 51 (2009), pp. 890-897
[64]
I.F.-N. Hung, D.K.-H. Wong, R.T.-P. Poon, D.Y.-T. Fong, A.H.-W. Chui, W.-K. Seto, et al.
Risk factors and post-resection independent predictive score for the recurrence of hepatitis B-related hepatocellular carcinoma.
[65]
L.-T. Chen, M.-F. Chen, L.-A. Li, P.-H. Lee, L.-B. Jeng, D.-Y. Lin, et al.
Long-term results of a randomized, observation-controlled, phase iii trial of adjuvant interferon alfa-2b in hepatocellular carcinoma after curative resection.
[66]
C.-Y. Wu, Y.-J. Chen, H.J. Ho, Y.-C. Hsu, K.N. Kuo, M.-S. Wu, et al.
Association between nucleoside analogues and risk of hepatitis B virus–related hepatocellular carcinoma recurrence following liver resection.
[67]
S. Kubo, S. Nishiguchi, K. Hirohashi, H. Tanaka, T. Shuto, O. Yamazaki, et al.
Effects of long-term postoperative interferon-alpha therapy on intrahepatic recurrence after resection of hepatitis C virus-related hepatocellular carcinoma. A randomized, controlled trial.
[68]
V. Mazzaferro, R. Romito, M. Schiavo, L. Mariani, T. Camerini, S. Bhoori, et al.
Sustained virologic response by direct antiviral agents reduces the incidence of hepatocellular carcinoma in patients with HCV infection.
[69]
M.A. Reig, Z. Mariño, C. Perelló, M. Iñarrairaegui, A. Ribeiro, S. Lens, et al.
Unexpected early tumor recurrence in patients with hepatitis C virus-related hepatocellular carcinoma undergoing interferon-free therapy: a note of caution.
J Hepatol, 65 (2016), pp. 719-726
[70]
F. Conti, F. Buonfiglioli, A. Scuteri, C. Crespi, L. Bolondi, P. Caraceni, et al.
Early occurrence and recurrence of hepatocellular carcinoma in HCV-related cirrhosis treated with direct-acting antivirals.
[71]
A.C. Huang, N. Mehta, J.L. Dodge, F.Y. Yao, N.A. Terrault.
Direct-acting antivirals do not increase the risk of hepatocellular carcinoma recurrence after local-regional therapy or liver transplant waitlist dropout.
Hepatology, 68 (2018), pp. 449-461
[72]
F. Piñero, I. Boin, A. Chagas, E. Quinonez, S. Marciano, M. Vilatoba, et al.
Direct-acting antivirals and hepatocellular carcinoma: no evidence of higher waitlist progression or post-transplant recurrence.
Liver Transplant, 26 (2020), pp. 640-650
[73]
M. Sherman, K.M. Peltekian, C. Lee.
Screening for hepatocellular carcinoma in chronic carriers of hepatitis B virus: incidence and prevalence of hepatocellular carcinoma in a North American urban population.
Hepatology, 22 (1995), pp. 432-438
[74]
C.E. Costentin, R. Layese, V. Bourcier, C. Cagnot, P. Marcellin, D. Guyader, et al.
Compliance with hepatocellular carcinoma surveillance guidelines associated with increased lead-time adjusted survival of patients with compensated viral cirrhosis: a multi-center cohort study.
[75]
B. Zhang, B. Yang.
Combined alpha fetoprotein testing and ultrasonography as a screening test for primary liver cancer.
J Med Screen, 6 (1999), pp. 108-110
[76]
J.-G. Chen, D.M. Parkin, Q.-G. Chen, J.-H. Lu, Q.-J. Shen, B.-C. Zhang, et al.
Screening for liver cancer: results of a randomised controlled trial in Qidong, China.
J Med Screen, 10 (2003), pp. 204-209
[77]
B.-H. Zhang, B.-H. Yang, Z.-Y. Tang.
Randomized controlled trial of screening for hepatocellular carcinoma.
J Cancer Res Clin Oncol, (2004), pp. 130
[78]
M. Zoli, D. Magalotti, G. Bianchi, C. Gueli, G. Marchesini, E. Pisi.
Efficacy of a surveillance program for early detection of hepatocellular carcinoma.
[79]
Franco Trevisani, P.E. DIntino, A.M. Morselli-Labate, G. Mazzella, E. Accogli, P. Caraceni, et al.
Serum a-fetoprotein for diagnosis of hepatocellular carcinoma in patients with chronic liver disease: influence of HBsAg and anti-HCV status.
J Hepatol, 2001 (2001), pp. 570-575
[80]
S. van Meer, R.A. de Man, M.J. Coenraad, D. Sprengers, K.M.J. van Nieuwkerk, H.-J. Klümpen, et al.
Surveillance for hepatocellular carcinoma is associated with increased survival: results from a large cohort in the Netherlands.
J Hepatol, 63 (2015), pp. 1156-1163
[81]
A.G. Singal, J. Tiro, X. Li, B. Adams-Huet, J. Chubak.
Hepatocellular carcinoma surveillance among patients with cirrhosis in a population-based integrated health care delivery system.
J Clin Gastroenterol, 51 (2017), pp. 650-655
[82]
P. Johnson, S. Berhane, C. Kagebayashi, S. Satomura, M. Teng, R. Fox, et al.
Impact of disease stage and aetiology on survival in hepatocellular carcinoma: implications for surveillance.
Br J Cancer, 116 (2017), pp. 441-447
[83]
H.Y. Kim, J.Y. Nam, J.H. Lee, Y.J. Chang, H. Lee, H. Cho, et al.
Intensity of surveillance for hepatocellular carcinoma determines survival in patients at risk in a hepatitis B-endemic area.
Aliment Pharmacol Ther, 47 (2018), pp. 1490-1501
[84]
H. Toyoda, T. Kumada, T. Tada, K. Mizuno, A. Hiraoka, K. Tsuji, et al.
Impact of hepatocellular carcinoma etiology and liver function on the benefit of surveillance: a novel approach for the adjustment of lead-time bias.
Liver Int, 2018 (2018), pp. 1-35
[85]
A.G. Singal, M.L. Volk, A. Waljee, R. Salgia, M. Higgins, A.B. Rogers, et al.
Meta-analysis: surveillance with ultrasound for early-stage hepatocellular carcinoma in patients with cirrhosis.
Aliment Pharmacol Ther, 30 (2009), pp. 37-47
[86]
A.G. Singal, A. Pillai, J. Tiro.
Early detection, curative treatment, and survival rates for hepatocellular carcinoma surveillance in patients with cirrhosis: a meta-analysis.
[87]
J. Thompson Coon, G. Rogers, P. Hewson, D. Wright, R. Anderson, S. Jackson, et al.
Surveillance of cirrhosis for hepatocellular carcinoma: a cost–utility analysis.
Br J Cancer, 98 (2008), pp. 1166-1175
[88]
K.L. Andersson, J.A. Salomon, S.J. Goldie, R.T. Chung.
Cost effectiveness of alternative surveillance strategies for hepatocellular carcinoma in patients with cirrhosis.
Clin Gastroenterol Hepatol, 6 (2008), pp. 1418-1424
[89]
A. Cucchetti, F. Trevisani, M. Cescon, G. Ercolani, F. Farinati, P. Del Poggio, et al.
Cost-effectiveness of semi-annual surveillance for hepatocellular carcinoma in cirrhotic patients of the Italian Liver Cancer population.
J Hepatol, 56 (2012), pp. 1089-1096
[90]
A. Mourad, S. Deuffic-Burban, N. Ganne-Carrié, T. Renaut-Vantroys, I. Rosa, A.-M. Bouvier, et al.
Hepatocellular carcinoma screening in patients with compensated hepatitis C virus (HCV)-related cirrhosis aware of their HCV status improves survival: a modeling approach.
Hepatology, 59 (2014), pp. 1471-1481
[91]
A. Cucchetti, F. Trevisani, A. Pecorelli, V. Erroi, F. Farinati, F. Ciccarese, et al.
Estimation of lead-time bias and its impact on the outcome of surveillance for the early diagnosis of hepatocellular carcinoma.
J Hepatol, 61 (2014), pp. 333-341
[92]
A. Cucchetti, F. Trevisani, L. Bucci, M. Ravaioli, F. Farinati, E.G. Giannini, et al.
Years of life that could be saved from prevention of hepatocellular carcinoma.
Aliment Pharmacol Ther, 43 (2016), pp. 814-824
[93]
L.L. Wong, W.M. Limm, R. Severino, L.M. Wong.
Improved survival with screening for hepatocellular carcinoma.
Liver Transplant, 6 (2000), pp. 320-325
[94]
L. Bolondi, S. Sofia, S. Siringo, S. Gaiani, A. Casali, G. Zironi, et al.
Surveillance programme of cirrhotic patients for early diagnosis and treatment of hepatocellular carcinoma: a cost effectiveness analysis.
Gut, 48 (2001), pp. 251-259
[95]
F. Trevisani, S. De Notariis, G. Rapaccini, F. Farinati, L. Benvegnù, M. Zoli, et al.
Semiannual and annual surveillance of cirrhotic patients for hepatocellular carcinoma: effects on cancer stage and patient survival (Italian experience).
Am J Gastroenterol, 97 (2002), pp. 734-744
[96]
V. Santi, F. Trevisani, A. Gramenzi, A. Grignaschi, F. Mirici-Cappa, P. Del Poggio, et al.
Semiannual surveillance is superior to annual surveillance for the detection of early hepatocellular carcinoma and patient survival.
J Hepatol, 53 (2010), pp. 291-297
[97]
A.G. Singal, M. Nehra, B. Adams-Huet, A.C. Yopp, J.A. Tiro, J.A. Marrero, et al.
Detection of hepatocellular carcinoma at advanced stages among patients in the HALT-C trial: where did surveillance fail?.
Am J Gastroenterol, 108 (2013), pp. 425-432
[98]
P. Del Poggio, S. Olmi, F. Ciccarese, M. Di Marco, G.L. Rapaccini, L. Benvegnù, et al.
Factors that affect efficacy of ultrasound surveillance for early.
[99]
J.-C. Trinchet, C. Chaffaut, V. Bourcier, F. Degos, J. Henrion, H. Fontaine, et al.
Ultrasonographic surveillance of hepatocellular carcinoma in cirrhosis: a randomized trial comparing 3- and 6-month periodicities.
Hepatology, 54 (2011), pp. 1987-1997
[100]
S.Y. Kim, J. An, Y.-S. Lim, S. Han, J.-Y. Lee, J.H. Byun, et al.
MRI with liver-specific contrast for surveillance of patients with cirrhosis at high risk of hepatocellular carcinoma.
[101]
C. Pocha, E. Dieperink, K.A. McMaken, A. Knott, P. Thuras, S.B. Ho.
Surveillance for hepatocellular cancer with ultrasonography vs. computed tomography – a randomised study.
Aliment Pharmacol Ther, 38 (2013), pp. 303-312
[102]
J.A. Davila, L. Henderson, J.R. Kramer, F. Kanwal, P.A. Richardson, Z. Duan, et al.
Utilization of surveillance for hepatocellular carcinoma among hepatitis C virus-infected veterans in the United States.
[103]
F. Piñero, S. Marciano, N. Fernández, J. Silva, Y. Zambelo, M. Cobos, et al.
Adherence to Barcelona Clinic Liver Cancer therapeutic algorithm for hepatocellular carcinoma in the daily practice.
Eur J Gastroenterol Hepatol, 30 (2018), pp. 376-383
[104]
A.S. Lok, R.K. Sterling, J.E. Everhart, E.C. Wright, J.C. Hoefs, A.M. Di Bisceglie, et al.
Des-γ-carboxy prothrombin and α-fetoprotein as biomarkers for the early detection of hepatocellular carcinoma.
Ygast, 138 (2010), pp. 493-502
[105]
J. Choi, G.-A. Kim, S. Han, W. Lee, S. Chun, Y.-S. Lim.
Longitudinal assessment of three serum biomarkers to detect very early-stage hepatocellular carcinoma.
Hepatology, 69 (2019), pp. 1983-1994
[106]
J. Marrero, Z. Feng, W. YY, M.H. Nguyen, A.S. Befeler, L.R. Roberts, et al.
Alpha-fetoprotein, des-gamma carboxyprothrombin, and lectin-bound alpha-fetoprotein in early hepatocellular carcinoma.
Gastroenterology, 137 (2009), pp. 110-118
[107]
C. Ayuso, J. Rimola, R. Vilana, M. Burrel, A. Darnell, Á. García-Criado, et al.
Diagnosis and staging of hepatocellular carcinoma (HCC): current guidelines.
[108]
A. Tang, M.R. Bashir, M.T. Corwin, I. Cruite, C.F. Dietrich, R.K.G. Do, et al.
Evidence supporting LI-RADS major features for CT- and MR imaging-based diagnosis of hepatocellular carcinoma: a systematic review.
Radiology, 286 (2018), pp. 29-48
[109]
C. Wald, M.W. Russo, J.K. Heimbach, H.K. Hussain, E.A. Pomfret, J. Bruix.
New OPTN/UNOS policy for liver transplant allocation: standardization of liver imaging, diagnosis, classification, and reporting of hepatocellular carcinoma.
Radiology, 266 (2013), pp. 376-382
[110]
Y.J. Lee, J.M. Lee, J.S. Lee, H.Y. Lee, B.H. Park, Y.H. Kim, et al.
Hepatocellular carcinoma: diagnostic performance of multidetector CT and MR imaging—a systematic review and meta-analysis.
Radiology, 275 (2015), pp. 97-109
[111]
R. Chou, C. Cuevas, R. Fu, B. Devine, N. Wasson, A. Ginsburg, et al.
Imaging techniques for the diagnosis of hepatocellular carcinoma.
Ann Intern Med, 162 (2015), pp. 697
[112]
R.F. Hanna, V.Z. Miloushev, A. Tang, L.A. Finklestone, S.Z. Brejt, R.S. Sandhu, et al.
Comparative 13-year meta-analysis of the sensitivity and positive predictive value of ultrasound, CT, and MRI for detecting hepatocellular carcinoma.
Abdom Radiol, (2015), pp. 1-20
[113]
K. Ishigami, K. Yoshimitsu, Y. Nishihara, H. Irie, Y. Asayama, T. Tajima, et al.
Hepatocellular carcinoma with a pseudocapsule on gadolinium-enhanced MR images: correlation with histopathologic findings.
Radiology, 250 (2009), pp. 435-443
[114]
M. Burrel, J.M. Llovet, C. Ayuso, C. Iglesias, M. Sala, R. Miquel, et al.
MRI angiography is superior to helical CT for detection of HCC prior to liver transplantation: an explant correlation.
Hepatology, 38 (2003), pp. 1034-1042
[115]
J. Rimola, A. Forner, S. Tremosini, M. Reig, R. Vilana, L. Bianchi, et al.
Non-invasive diagnosis of hepatocellular carcinoma 62cm in cirrhosis. Diagnostic accuracy assessing fat, capsule and signal intensity at dynamic MRI.
J Hepatol, 56 (2012), pp. 1317-1323
[116]
M. Renzulli, R. Golfieri.
Bologna Liver Oncology Group (BLOG). Diagnostic performance of gadoxetic acid–enhanced liver MR imaging in the detection of hccs and allocation of transplant recipients on the basis of the Milan criteria and UnOs guidelines: correlation with histopathologic findings.
J Gastroenterol Hepatol, 31 (2015), pp. 69-80
[117]
H.-J. Jang, T.K. Kim, S.R. Wilson.
Small nodules (1–2cm) in liver cirrhosis: characterization with contrast-enhanced ultrasound.
Eur J Radiol, 72 (2009), pp. 418-424
[118]
H.-D. Kim, Y.-S. Lim, S. Han, J. An, G.-A. Kim, S.Y. Kim, et al.
Evaluation of early-stage hepatocellular carcinoma by magnetic resonance imaging with gadoxetic acid detects additional lesions and increases overall survival.
Gastroenterology, 148 (2015), pp. 1371-1382
[119]
E. Terzi, M. Iavarone, M. Pompili, L. Veronese, G. Cabibbo, M. Fraquelli, et al.
Contrast enhanced ultrasound identifies hepatocellular carcinoma in cirrhosis: a large multicenter retrospective study.
[120]
S. Tremosini, A. Forner, L. Boix, R. Vilana, L. Bianchi, M. Reig, et al.
Prospective validation of an immunohistochemical panel (glypican 3, heat shock protein 70 and glutamine synthetase) in liver biopsies for diagnosis of very early hepatocellular carcinoma.
[121]
R. Tateishi, H. Yoshida, Y. Matsuyama, N. Mine, Y. Kondo, M. Omata.
Diagnostic accuracy of tumor markers for hepatocellular carcinoma: a systematic review.
[122]
F. Farinati, D. Marino, M. De Giorgio, A. Baldan, M. Cantarini, C. Cursaro, et al.
Diagnostic and prognostic role of alpha-fetoprotein in hepatocellular carcinoma: both or neither?.
Am J Gastroenterol, 101 (2006), pp. 524-532
[123]
W.-J. Ma, H.-Y. Wang, L.-S. Teng.
Correlation analysis of preoperative serum alpha-fetoprotein (AFP) level and prognosis of hepatocellular carcinoma (HCC) after hepatectomy.
World J Surg Oncol, 11 (2013), pp. 212
[124]
R. Santambrogio, E. Opocher, M. Costa, M. Barabino, M. Zuin, E. Bertolini, et al.
Hepatic resection for “BCLC Stage A” hepatocellular carcinoma. The prognostic role of alpha-fetoprotein.
Ann Surg Oncol, 19 (2011), pp. 426-434
[125]
D. Choi, H.K. Lim, H. Rhim, Y.-S. Kim, W.J. Lee, S.W. Paik, et al.
Percutaneous radiofrequency ablation for early-stage hepatocellular carcinoma as a first-line treatment: long-term results and prognostic factors in a large single-institution series.
Eur Radiol, 17 (2006), pp. 684-692
[126]
W.Y. Kao, Y.Y. Chiou, H.H. Hung, C.W. Su, Y.H. Chou, J.C. Wu, et al.
Serum alpha-fetoprotein response can predict prognosis in hepatocellular carcinoma patients undergoing radiofrequency ablation therapy.
Clin Radiol, 67 (2012), pp. 429-436
[127]
D.H. Lee, J.M. Lee, J.Y. Lee, S.H. Kim, J.-H. Yoon, Y.J. Kim, et al.
Radiofrequency ablation of hepatocellular carcinoma as first-line treatment: long-term results and prognostic factors in 162 patients with cirrhosis.
Radiology, 270 (2014), pp. 900-909
[128]
Y. Wang, Y. Chen, N. Ge, L. Zhang, X. Xie, J. Zhang, et al.
Prognostic significance of alpha-fetoprotein status in the outcome of hepatocellular carcinoma after treatment of transarterial chemoembolization.
Ann Surg Oncol, 19 (2012), pp. 3540-3546
[129]
Q. Lai, A.W. Avolio, I. Graziadei, G. Otto, M. Rossi, G. Tisone, et al.
Alpha-fetoprotein and modified response evaluation criteria in solid tumors progression after locoregional therapy as predictors of hepatocellular cancer recurrence and death after transplantation.
Liver Transplant, 19 (2013), pp. 1108-1118
[130]
T. Kuzuya, Y. Asahina, K. Tsuchiya, K. Tanaka, Y. Suzuki, T. Hoshioka, et al.
Early decrease in α-fetoprotein, but not des-γ-carboxy prothrombin, predicts sorafenib efficacy in patients with advanced hepatocellular carcinoma.
Oncology, 81 (2011), pp. 251-258
[131]
N. Personeni, S. Bozzarelli, T. Pressiani, L. Rimassa, M.C. Tronconi, F. Sclafani, et al.
Usefulness of alpha-fetoprotein response in patients treated with sorafenib for advanced hepatocellular carcinoma.
J Hepatol, 57 (2012), pp. 101-107
[132]
J. Bruix, A.-L. Cheng, G. Meinhardt, K. Nakajima, Y. De Sanctis, J. Llovet.
Prognostic factors and predictors of sorafenib benefit in patients with hepatocellular carcinoma: analysis of two phase III studies.
J Hepatol, 67 (2017), pp. 999-1008
[133]
A.X. Zhu, J.O. Park, B.-Y. Ryoo, C.-J. Yen, R. Poon, D. Pastorelli, et al.
Ramucirumab versus placebo as second-line treatment in patients with advanced hepatocellular carcinoma following first-line therapy with sorafenib (REACH): a randomised, double-blind, multicentre, phase 3 trial.
Lancet Oncol, 16 (2015), pp. 859-870
[134]
G.K. Abou-Alfa, T. Meyer, A.-L. Cheng, A.B. El-Khoueiry, L. Rimassa, B.-Y. Ryoo, et al.
Cabozantinib in patients with advanced and progressing hepatocellular carcinoma.
N Engl J Med, 379 (2018), pp. 54-63
[135]
B. Hameed, N. Mehta, G. Sapisochin, J.P. Roberts, F.Y. Yao.
Alpha-fetoprotein level >1000ng/mL as an exclusion criterion for liver transplantation in patients with hepatocellular carcinoma meeting the Milan criteria.
Liver Transplant, 20 (2014), pp. 945-951
[136]
C. Duvoux, F.R. Thoraval, T. Decaens, F. Pessione, H. Badran, T. Piardi, et al.
Liver transplantation for hepatocellular carcinoma: a model including – fetoprotein improves the performance of Milan criteria.
[137]
N. Mehta, J. Heimbach, D.M. Harnois, G. Sapisochin, J.L. Dodge, D. Lee, et al.
Validation of a Risk Estimation of Tumor Recurrence After Transplant (RETREAT) score for hepatocellular carcinoma recurrence after liver transplant.
JAMA Oncol, 3 (2017), pp. 493-500
[138]
V. Mazzaferro, C. Sposito, J. Zhou, A.D. Pinna, L. De Carlis, J. Fan, et al.
Metroticket 2.0 model for analysis of competing risks of death after liver transplantation for hepatocellular carcinoma.
Gastroenterology, 154 (2018), pp. 128-139
[139]
K. Okuda, T. Ohtsuki, H. Obata, M. Tomimatsu, N. Okazaki, H. Hasegawa, et al.
Natural history of hepatocellular carcinoma and prognosis in relation to treatment. Study of 850 patients.
[140]
S. Chevret, J.C. Trinchet, D. Mathieu, A.A. Rached, M. Beaugrand, C. Chastang.
A new prognostic classification for predicting survival in patients with hepatocellular carcinoma. Groupe d’Etude et de Traitement du Carcinome Hépatocellulaire.
J Hepatol, 31 (1999), pp. 133-141
[141]
CLIP TCOTLIP.
A new prognostic system for hepatocellular carcinoma: a retrospective study of 435 patients: the Cancer of the Liver Italian Program (CLIP) investigators.
Hepatology, 28 (1998), pp. 751-755
[142]
T.W.T. Leung, A.M.Y. Tang, B. Zee, W.Y. Lau, P.B.S. Lai, K.L. Leung, et al.
Construction of the Chinese University Prognostic Index for hepatocellular carcinoma and comparison with the TNM staging system, the Okuda staging system, and the Cancer of the Liver Italian Program staging system.
Cancer, 94 (2002), pp. 1760-1769
[143]
M. Kudo, H. Chung, Y. Osaki.
Prognostic staging system for hepatocellular carcinoma (CLIP score): its value and limitations, and a proposal for a new staging system, the Japan Integrated Staging Score (JIS score).
J Gastroenterol, 38 (2003), pp. 207-215
[144]
T. Yau, V.Y.F. Tang, T.-J. Yao, S.-T. Fan, C.M. Lo, R.T.P. Poon.
Development of Hong Kong liver cancer staging system with treatment stratification for patients with hepatocellular carcinoma.
Gastroenterology, 146 (2014), pp. 1691-1693
[145]
J.M. Llovet, C. Brú, J. Bruix.
Prognosis of hepatocellular carcinoma: the BCLC staging classification.
Semin Liver Dis, 19 (1999), pp. 329-338
[146]
J.A. Marrero, R.J. Fontana, A. Barrat, F. Askari, H.S. Conjeevaram, G.L. Su, et al.
Prognosis of hepatocellular carcinoma: comparison of 7 staging systems in an American cohort.
Hepatology, 41 (2005), pp. 707-715
[147]
U. Cillo, A. Vitale, F. Grigoletto, F. Farinati, A. Brolese, G. Zanus, et al.
Prospective validation of the Barcelona Clinic Liver Cancer staging system.
J Hepatol, 44 (2006), pp. 723-731
[148]
A. Guglielmi, A. Ruzzenente, S. Pachera, A. Valdegamberi, M. Sandri, M. D’Onofrio, et al.
Comparison of seven staging systems in cirrhotic patients with hepatocellular carcinoma in a cohort of patients who underwent radiofrequency ablation with complete response.
Am J Gastroenterol, 103 (2008), pp. 597-604
[149]
S. Roayaie, K. Obeidat, C. Sposito, L. Mariani, S. Bhoori, A. Pellegrinelli, et al.
Resection of hepatocellular cancer ≤2cm: results from two Western centers.
Hepatology, 57 (2013), pp. 1426-1435
[150]
K. Hasegawa, N. Kokudo, M. Makuuchi, N. Izumi, T. Ichida, M. Kudo, et al.
Comparison of resection and ablation for hepatocellular carcinoma: a cohort study based on a Japanese nationwide survey.
J Hepatol, 58 (2013), pp. 724-729
[151]
Y.K. Cho, J.K. Kim, M.Y. Kim, H. Rhim, J.K. Han.
Systematic review of randomized trials for hepatocellular carcinoma treated with percutaneous ablation therapies.
Hepatology, 49 (2008), pp. 453-459
[152]
M. Sala, J. Fuster, J.M. Llovet, M. Navasa, M. Sol, M.A. Varela, et al.
High pathological risk of recurrence after surgical resection for hepatocellular carcinoma: an indication for salvage liver transplantation.
Liver Transplant, 10 (2009), pp. 1294-1300
[153]
J. Bruix, A. Castells, J. Bosch, F. Feu, J. Fuster, J.C. Garcia-Pagan, et al.
Surgical resection of hepatocellular carcinoma in cirrhotic patients: prognostic value of preoperative portal pressure.
Gastroenterology, 111 (1996), pp. 1018-1022
[154]
A. Berzigotti, M. Reig, J.G. Abraldes, J. Bosch.
Portal hypertension and the outcome of surgery for hepatocellular carcinoma in compensated cirrhosis: a systematic review and meta-analysis.
[155]
J.M. Llovet, M.I. Real, X. Montañá, R. Planas, S. Coll, J. Aponte, et al.
Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial.
Lancet, 359 (2002), pp. 1734-1739
[156]
C. Lo, C.-M. Lo, H. Ngan, W.-K. Tso, C.-L. Liu, C.-M. Lam, et al.
Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma.
Hepatology, 35 (2002), pp. 1164-1171
[157]
J. Llovet.
Systematic review of randomized trials for unresectable hepatocellular carcinoma: chemoembolization improves survival.
Hepatology, 37 (2003), pp. 429-442
[158]
R.S. Finn, P. Merle, A. Granito, Y.-H. Huang, G. Bodoky, M. Pracht, et al.
Outcomes of sequential treatment with sorafenib followed by regorafenib for HCC: additional analyses from phase III RESORCE trial.
[159]
F. Durand, D. Valla.
Assessment of the prognosis of cirrhosis: Child–Pugh versus MELD.
J Hepatol, 42 (2005), pp. S100-S107
[160]
M. Reig, A. Darnell, A. Forner, J. Rimola, C. Ayuso, J. Bruix.
Systemic therapy for hepatocellular carcinoma: the issue of treatment stage migration and registration of progression using the BCLC-refined RECIST.
Semin Liver Dis, 34 (2014), pp. 444-455
[161]
L. Bolondi, A. Burroughs, J.-F. Dufour, P. Galle, V. Mazzaferro, F. Piscaglia, et al.
Heterogeneity of patients with intermediate (BCLC B) hepatocellular carcinoma: proposal for a subclassification to facilitate treatment decisions.
Semin Liver Dis, 32 (2013), pp. 348-359
[162]
X. Adhoute, G. Penaranda, J.-P. Bronowicki, J.-L. Raoul.
Usefulness of the HKLC vs. the BCLC staging system in a European HCC cohort.
J Hepatol, 62 (2015), pp. 492-493
[163]
A. Hiraoka, T. Kumada, M. Kudo, M. Hirooka, K. Tsuji, E. Itobayashi, et al.
Albumin-bilirubin (ALBI) grade as part of the evidence-based clinical practice guideline for HCC of the Japan Society of Hepatology: a comparison with the liver damage and child-pugh classifications.
Liver Cancer, 6 (2017), pp. 204-215
[164]
S. Leoni, F. Piscaglia, I. Serio, E. Terzi, I. Pettinari, L. Croci, et al.
Adherence to AASLD guidelines for the treatment of hepatocellular carcinoma in clinical practice: experience of the Bologna Liver Oncology Group.
Digest Liver Dis, 46 (2014), pp. 549-555
[165]
L. Gashin, E. Tapper, A. Babalola, K.-C. Lai, R. Miksad, R. Malik, et al.
Determinants and outcomes of adherence to recommendations from a multidisciplinary tumour conference for hepatocellular carcinoma.
HPB (Oxford), 16 (2014), pp. 1009-1015
[166]
K.M. Kim, D.H. Sinn, S.-H. Jung, G.-Y. Gwak, Y.H. Paik, M.S. Choi, et al.
The recommended treatment algorithms of the BCLC and HKLC staging systems: does following these always improve survival rates for HCC patients?.
Liver Int, 36 (2016), pp. 1490-1497
[167]
M.C. Wallace, Y. Huang, D.B. Preen, G. Garas, L.A. Adams, G. MacQuillan, et al.
HKLC triages more hepatocellular carcinoma patients to curative therapies compared to BCLC and is associated with better survival.
Dig Dis Sci, (2017), pp. 1-11
[168]
M. Guarino, R. Tortora, G. de Stefano, C. Coppola, F. Morisco, A. Salomone Megna, et al.
Adherence to Barcelona Clinic Liver Cancer guidelines in field practice: results of Progetto Epatocarcinoma Campania.
J Gastroenterol Hepatol, 33 (2018), pp. 1123-1130
[169]
L. Kikuchi, A.L. Chagas, R.S. Alencar, C. Tani, M.A. Diniz, L.A. D’Albuquerque, et al.
Adherence to BCLC recommendations for the treatment of hepatocellular carcinoma: impact on survival according to stage.
Clinics, 72 (2017), pp. 454-460
[170]
R. Lencioni, D. Cioni, L. Crocetti, C. Franchini, C.D. Pina, J. Lera, et al.
Early-stage hepatocellular carcinoma in patients with cirrhosis: long-term results of percutaneous image-guided radiofrequency ablation.
Radiology, 234 (2005), pp. 961-967
[171]
D. Choi, H.K. Lim, H. Rhim, Y.-S. Kim, W.J. Lee, S.W. Paik, et al.
Percutaneous radiofrequency ablation for early-stage hepatocellular carcinoma as a first-line treatment: long-term results and prognostic factors in a large single-institution series.
Eur Radiol, 17 (2006), pp. 684-692
[172]
A. Orlando, G. Leandro, M. Olivo, A. Andriulli, M. Cottone.
Radiofrequency thermal ablation vs. percutaneous ethanol injection for small hepatocellular carcinoma in cirrhosis: meta-analysis of randomized controlled trials.
Am J Gastroenterol, 104 (2009), pp. 514-524
[173]
G. Germani, M. Pleguezuelo, K. Gurusamy, T. Meyer, G. Isgrò, A.K. Burroughs.
Clinical outcomes of radiofrequency ablation, percutaneous alcohol and acetic acid injection for hepatocelullar carcinoma: a meta-analysis.
J Hepatol, 52 (2010), pp. 380-388
[174]
T.W. Kang, H.K. Lim, M.W. Lee, Y.-S. Kim, H. Rhim, W.J. Lee, et al.
Long-term therapeutic outcomes of radiofrequency ablation for subcapsular versus nonsubcapsular hepatocellular carcinoma: a propensity score matched study.
Radiology, 280 (2016), pp. 300-312
[175]
A. Facciorusso.
Microwave ablation versus radiofrequency ablation for the treatment of hepatocellular carcinoma: a systematic review and meta-analysis.
Int J Hyperth, 32 (2016), pp. 339-344
[176]
C. Wang, H. Wang, W. Yang, K. Hu, H. Xie, K.-Q. Hu, et al.
Multicenter randomized controlled trial of percutaneous cryoablation versus radiofrequency ablation in hepatocellular carcinoma.
Hepatology, 61 (2015), pp. 1579-1590
[177]
O. Sutter, J. Calvo, G. N’Kontchou, J.-C. Nault, R. Ourabia, P. Nahon, et al.
Safety and efficacy of irreversible electroporation for the treatment of hepatocellular carcinoma not amenable to thermal ablation techniques: a retrospective single-center case series.
Radiology, 284 (2017), pp. 877-886
[178]
G. Sapisochin, A. Barry, M. Doherty, S. Fischer, N. Goldaracena, R. Rosales, et al.
J Hepatol, 67 (2017), pp. 92-99
[179]
N.G. Berger, M.N. Tanious, A.Y. Hammad, J.T. Miura, H. Mogal, C.N. Clarke, et al.
External radiation or ablation for solitary hepatocellular carcinoma: a survival analysis of the SEER database.
J Surg Oncol, 116 (2017), pp. 307-312
[180]
M. Feng, K. Suresh, M.J. Schipper, L. Bazzi, E. Ben-Josef, M.M. Matuszak, et al.
Individualized adaptive stereotactic body radiotherapy for liver tumors in patients at high risk for liver damage.
[181]
S. Roayaie, G. Jibara, P. Tabrizian, J.-W. Park, J. Yang, L. Yan, et al.
The role of hepatic resection in the treatment of hepatocellular cancer.
Hepatology, 62 (2015), pp. 440-451
[182]
A. Vitale, P. Burra, A.C. Frigo, F. Trevisani, F. Farinati, G. Spolverato, et al.
Survival benefit of liver resection for patients with hepatocellular carcinoma across different Barcelona Clinic Liver Cancer stages: a multicentre study.
J Hepatol, 62 (2015), pp. 617-624
[183]
G.K. Glantzounis, A. Paliouras, M.C. Stylianidi, H. Milionis, P. Tzimas, D. Roukos, et al.
The role of liver resection in the management of intermediate and advanced stage hepatocellular carcinoma. A systematic review.
Eur J Surg Oncol, (2017), pp. 1-14
[184]
J.M. Llovet, J. Fuster, J. Bruix.
Intention-to-treat analysis of surgical treatment for early hepatocellular carcinoma: resection versus transplantation.
Hepatology, 30 (1999), pp. 1434-1440
[185]
C. Ripoll, R. Groszmann, G. Garcia-Tsao, N. Grace, A. Burroughs, R. Planas, et al.
Hepatic venous pressure gradient predicts clinical decompensation in patients with compensated cirrhosis.
Gastroenterology, 133 (2007), pp. 481-488
[186]
A. Cucchetti, G. Ercolani, M. Vivarelli, M. Cescon, M. Ravaioli, G. Ramacciato, et al.
Is portal hypertension a contraindication to hepatic resection?.
Ann Surg, 250 (2009), pp. 922-928
[187]
S.B. Choi, H.J. Kim, T.J. Song, H.S. Ahn, S.Y. Choi.
Influence of clinically significant portal hypertension on surgical outcomes and survival following hepatectomy for hepatocellular carcinoma: a systematic review and meta-analysis.
J Hepatobiliary Pancreat Sci, 21 (2014), pp. 639-647
[188]
J. Liu, H. Zhang, Y. Xia, T. Yang, Y. Gao, J. Li, et al.
Impact of clinically significant portal hypertension on outcomes after partial hepatectomy for hepatocellular carcinoma: a systematic review and meta-analysis.
Int Hepato-Pancreato-Biliary Assoc, (2018), pp. 1-13
[189]
A. Cucchetti, M. Cescon, R. Golfieri, F. Piscaglia, M. Renzulli, F. Neri, et al.
Hepatic venous pressure gradient in the preoperative assessment of patients with resectable hepatocellular carcinoma.
J Hepatol, 64 (2016), pp. 79-86
[190]
R. de Franchis, B.V. Faculty.
Expanding consensus in portal hypertension.
J Hepatol, 63 (2015), pp. 743-752
[191]
A. Lisotti, F. Azzaroli, F. Buonfiglioli, M. Montagnani, P. Cecinato, L. Turco, et al.
Indocyanine green retention test as a noninvasive marker of portal hypertension and esophageal varices in compensated liver cirrhosis.
Hepatology, 59 (2013), pp. 643-650
[192]
A. Twaij, P.H. Pucher, M.H. Sodergren, T. Gall, A. Darzi, L.R. Jiao.
Laparoscopic vs open approach to resection of hepatocellular carcinoma in patients with known cirrhosis: systematic review and meta-analysis.
World J Gastroenterol, 20 (2014), pp. 8274-8281
[193]
V. Molina, J. Sampson-Dávila, J. Ferrer, C. Fondevila, R. Díaz Del Globbo, D. Calatayud, et al.
Benefits of laparoscopic liver resection in patients with hepatocellular carcinoma and portal hypertension: a case-matched study.
[194]
S. Eguchi, T. Kanematsu, S. Arii, M. Okazaki, K. Okita, M. Omata, et al.
Comparison of the outcomes between an anatomical subsegmentectomy and a non-anatomical minor hepatectomy for single hepatocellular carcinomas based on a Japanese nationwide survey.
Surgery, 143 (2008), pp. 469-475
[197]
M.-S. Chen, J.-Q. Li, Y. Zheng, R.-P. Guo, H.-H. Liang, Y.-Q. Zhang, et al.
A prospective randomized trial comparing percutaneous local ablative therapy and partial hepatectomy for small hepatocellular carcinoma.
[198]
P.-H. Liu, C.-Y. Hsu, C.-Y. Hsia, Y.-H. Lee, Y.-H. Huang, Y.-Y. Chiou, et al.
Surgical resection versus radiofrequency ablation for single hepatocellular carcinoma ≤2cm in a propensity score model.
Ann Surg, 263 (2016), pp. 538-545
[199]
A. Majumdar, D. Roccarina, D. Thorburn, B.R. Davidson, E. Tsochatzis, K.S. Gurusamy.
Management of people with early- or very early-stage hepatocellular carcinoma.
Cochrane Database Syst Rev, 44 (2017), pp. 494a
[200]
A. Cucchetti, F. Piscaglia, M. Cescon, A. Colecchia, G. Ercolani, L. Bolondi, et al.
Cost-effectiveness of hepatic resection versus percutaneous radiofrequency ablation for early hepatocellularcarcinoma.
J Hepatol, 59 (2013), pp. 300-307
[201]
K. Feng, J. Yan, X. Li, F. Xia, K. Ma, S. Wang, et al.
A randomized controlled trial of radiofrequency ablation and surgical resection in the treatment of small hepatocellular carcinoma.
J Hepatol, 57 (2012), pp. 794-802
[202]
P.-H. Liu, Y.-H. Lee, C.-Y. Hsu, C.-Y. Hsia, Y.-H. Huang, Y.-Y. Chiou, et al.
Surgical resection is better than transarterial chemoembolization for hepatocellular carcinoma beyond Milan criteria independent of performance status.
J Gastrointest Surg, 18 (2014), pp. 1623-1631
[204]
L. Yin, H. Li, A.-J. Li, W.-Y. Lau, Z.-Y. Pan, E.C.H. Lai, et al.
Partial hepatectomy vs. transcatheter arterial chemoembolization for resectable multiple hepatocellular carcinoma beyond Milan criteria: a RCT.
[205]
C.E. Costentin, G. Amaddeo, T. Decaens, K. Boudjema, P. Bachellier, F. Muscari, et al.
Comparison between different D-Dimer cutoff values to assess the individual risk of recurrent venous thromboembolism: analysis of results obtained in the DULCIS study.
Int J Lab Hem, 38 (2015), pp. 1515-1525
[206]
Y. Zhou, C. Sui, B. Li, Z. Yin, Y. Tan, J. Yang, et al.
Repeat hepatectomy for recurrent hepatocellular carcinoma: a local experience and a systematic review.
World J Surg Oncol, 8 (2010), pp. 55
[207]
S. Lee, C. Hyuck David Kwon, J. Man Kim, J.-W. Joh, S. Woon Paik, B.-W. Kim, et al.
Time of hepatocellular carcinoma recurrence after liver resection and alpha-fetoprotein are important prognostic factors for salvage liver transplantation.
Liver Transplant, 20 (2014), pp. 1057-1063
[208]
J.F. Fàbrega, A. Forner, A. Liccioni, R. Miquel.
Prospective validation of ab initio liver transplantation in hepatocellular carcinoma upon detection of risk factors for recurrence after resection.
Hepatology, 63 (2016), pp. 839-849
[209]
A. Villanueva, Y. Hoshida, C. Battiston, V. Tovar, D. Sia, C. Alsinet, et al.
Combining clinical, pathology, and gene expression data to predict recurrence of hepatocellular carcinoma.
Gastroenterology, 140 (2011), pp. 1501-1502
[210]
P. Gavriilidis, A. Askari, D. Azoulay.
Survival following redo hepatectomy vs radiofrequency ablation for recurrent hepatocellular carcinoma: a systematic review and meta-analysis.
Int Hepato-Pancreato-Biliary Assoc, (2016), pp. 1-7
[211]
A.C.Y. Chan, S.C. Chan, K.S.H. Chok, T.T. Cheung, D.W. Chiu, R.T.P. Poon, et al.
Treatment strategy for recurrent hepatocellular carcinoma: salvage transplantation, repeated resection, or radiofrequency ablation?.
Liver Transplant, 19 (2013), pp. 411-419
[212]
C. Lim, H. Shinkawa, K. Hasegawa, P. Bhangui, C. Salloum, C. Gomez Gavara, et al.
Salvage liver transplantation or repeat hepatectomy for recurrent hepatocellular carcinoma: an intent-to-treat analysis.
Liver Transplant, 23 (2017), pp. 1553-1563
[213]
R.J. de Haas, C. Lim, P. Bhangui, C. Salloum, P. Compagnon, C. Feray, et al.
Curative salvage liver transplantation in patients with cirrhosis and hepatocellular carcinoma: an intention-to-treat analysis.
Hepatology, 67 (2018), pp. 204-215
[214]
H. Muaddi, D.P. Al-Adra, R. Beecroft, A. Ghanekar, C.-A. Moulton, A. Doyle, et al.
Liver transplantation is equally effective as a salvage therapy for patients with hepatocellular carcinoma recurrence following radiofrequency ablation or liver resection with curative intent.
Ann Surg Oncol, (2018), pp. 1-9
[215]
M. Samuel, P.K.H. Chow, E. Chan Shih-Yen, D. Machin, K.C. Soo.
Neoadjuvant and adjuvant therapy for surgical resection of hepatocellular carcinoma.
Cochrane Database Syst Rev, 20 (2009), pp. 295
[216]
J. Bruix, T. Takayama, V. Mazzaferro, G.-Y. Chau, J. Yang, M. Kudo, et al.
Articles Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): a phase 3, randomised, double-blind, placebo-controlled trial.
Lancet Oncol, 16 (2015), pp. 1344-1354
[217]
C. Cong, M. Yin-Hua, Z. Ya-Ting, Z. Fan, Z. Ning, W. Xiang, et al.
Effect of dendritic cell-based immunotherapy on hepatocellular carcinoma: a systematic review and meta-analysis.
Cytotherapy, 20 (2018), pp. 975-989
[218]
H. Wang, A. Liu, W. Bo, X. Feng, Y. Hu, L. Tian, et al.
Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma patients after curative resection, a systemic review and meta-analysis.
Digest Liver Dis, 48 (2016), pp. 1275-1282
[220]
On Behalf of the PRECISION V Investigators, J. Lammer, K. Malagari, T. Vogl, F. Pilleul, A. Denys, et al.
Prospective randomized study of doxorubicin-eluting-bead embolization in the treatment of hepatocellular carcinoma: results of the PRECISION V study.
Cardiovasc Intervent Radiol, 33 (2009), pp. 41-52
[221]
J. Bruix, J.M. Llovet, A. Castells, X. Montañá, C. Brú, M.C. Ayuso, et al.
Transarterial embolization versus symptomatic treatment in patients with advanced hepatocellular carcinoma: results of a randomized, controlled trial in a single institution.
Hepatology, 27 (1998), pp. 1578-1583
[222]
R.S. Oliveri, J. Wetterslev, C. Gluud.
Transarterial (chemo)embolisation for unresectable hepatocellular carcinoma.
Cochrane Database Syst Rev, 24 (2011), pp. 625
[223]
T. Meyer, A. Kirkwood, M. Roughton, S. Beare, E. Tsochatzis, D. Yu, et al.
A randomised phase II/III trial of 3-weekly cisplatin-based sequential transarterial chemoembolisation vs embolisation alone for hepatocellular carcinoma.
Br J Cancer, 108 (2013), pp. 1252-1259
[224]
K.T. Brown, R.K. Do, M. Gonen, A.M. Covey, G.I. Getrajdman, C.T. Sofocleous, et al.
Randomized trial of hepatic artery embolization for hepatocellular carcinoma using doxorubicin-eluting microspheres compared with embolization with microspheres alone.
J Clin Oncol, 34 (2016), pp. 2046-2053
[225]
M. Burrel, Maria Reig, Alejandro Forner, M. Barrufet, C.R. de Lope, S. Tremosini, et al.
Survival of patients with hepatocellular carcinoma treated by transarterial chemoembolisation (TACE) using Drug Eluting Beads. Implications for clinical practice and trial design.
J Hepatol, 56 (2016), pp. 1330-1335
[226]
B. Sangro, M. Iñarrairaegui, J. Bilbao.
Radioembolization for hepatocellular carcinoma.
J Hepatol, 56 (2012), pp. 464-473
[227]
R.J. Lewandowski, L.M. Kulik, A. Riaz, S. Senthilnathan, M.F. Mulcahy, R.K. Ryu, et al.
A comparative analysis of transarterial downstaging for hepatocellular carcinoma: chemoembolization versus radioembolization.
Am J Transplant, 9 (2009), pp. 1920-1928
[228]
B. Sangro, L. Carpanese, R. Cianni, R. Golfieri, D. Gasparini, S. Ezziddin, et al.
Survival after yttrium-90 resin microsphere radioembolization of hepatocellular carcinoma across Barcelona clinic liver cancer stages: a European evaluation.
Hepatology, 54 (2011), pp. 868-878
[229]
V. Mazzaferro, C. Sposito, S. Bhoori, R. Romito, C. Chiesa, C. Morosi, et al.
Yttrium-90 radioembolization for intermediate-advanced hepatocellular carcinoma: a phase 2 study.
Hepatology, 57 (2013), pp. 1826-1837
[230]
K. Memon, L. Kulik, R.J. Lewandowski, M.F. Mulcahy, A.B. Benson, D. Ganger, et al.
Radioembolization for hepatocellular carcinoma with portal vein thrombosis: impact of liver function on systemic treatment options at disease progression.
J Hepatol, 58 (2013), pp. 73-80
[231]
A. Fouly El, J. Ertle, A. Dorry El, M.K. Shaker, A. Dechêne, H. Abdella, et al.
In intermediate stage hepatocellular carcinoma: radioembolization with yttrium 90 or chemoembolization?.
Liver Int, 35 (2014), pp. 627-635
[232]
P.C.-H. Kwok, K.C. Leung, M.T. Cheung, T.W. Lam, L.T. Szeto, S.Q.-H. Chou, et al.
Survival benefit of radioembolization for inoperable hepatocellular carcinoma using yttrium-90 microspheres.
J Gastroenterol Hepatol, 29 (2014), pp. 1897-1904
[234]
Y. Zhang, Y. Li, H. Ji, X. Zhao, H. Lu.
Transarterial Y90 radioembolization versus chemoembolization for patients with hepatocellular carcinoma: a meta-analysis.
Bst, 9 (2015), pp. 289-298
[235]
J.-Y. Teo, J.C. Allen Jr., D.C. Ng, S.-P. Choo, D.W.M. Tai, J.P.E. Chang, et al.
A systematic review of contralateral liver lobe hypertrophy after unilobar selective internal radiation therapy with Y90.
Int Hepato-Pancreato-Biliary Assoc, 18 (2016), pp. 7-12
[236]
F.T. Kolligs, J.I. Bilbao, T. Jakobs, M. Iñarrairaegui, J.M. Nagel, M. Rodriguez, et al.
Pilot randomized trial of selective internal radiation therapy vs. chemoembolization in unresectable hepatocellular carcinoma.
Liver Int, 35 (2015), pp. 1715-1721
[237]
R. Salem, A.C. Gordon, S. Mouli, R. Hickey, J. Kallini, A. Gabr, et al.
Y90 radioembolization significantly prolongs time to progression compared with chemoembolization in patients with hepatocellular carcinoma.
[238]
M.B. Pitton, R. Kloeckner, C. Ruckes, G.M. Wirth, W. Eichhorn, M.A. Wörns, et al.
Randomized comparison of selective internal radiotherapy (SIRT) versus drug-eluting bead transarterial chemoembolization (DEB-TACE) for the treatment of hepatocellular carcinoma.
Cardiovasc Intervent Radiol, 38 (2014), pp. 352-360
[239]
V. Vilgrain, H. Pereira, E. Assenat, B. Guiu, A.D. Ilonca, G.-P. Pageaux, et al.
Efficacy and safety of selective internal radiotherapy with yttrium-90 resin microspheres compared with sorafenib in locally advanced and inoperable hepatocellular carcinoma (SARAH): an open-label randomised controlled phase 3 trial.
Lancet Oncol, 18 (2017), pp. 1624-1636
[240]
P.K.H. Chow, M. Gandhi, S.-B. Tan, M.W. Khin, A. Khasbazar, J. Ong, et al.
SIRveNIB: selective internal radiation therapy versus sorafenib in Asia-Paci.
J Clin Oncol, (2018), pp. 1-12
[241]
J.M. Llovet, S. Ricci, V. Mazzaferro, P. Hilgard, E. Gane, J.-F. Blanc, et al.
Sorafenib in advanced hepatocellular carcinoma.
N Engl J Med, 359 (2008), pp. 378-390
[242]
J. Ricke, K. Bulla, F. Kolligs, M. Peck-Radosavljevic, P. Reimer, B. Sangro, et al.
Safety and toxicity of radioembolization plus Sorafenib in advanced hepatocellular carcinoma: analysis of the European multicentre trial SORAMIC.
Liver Int, 35 (2014), pp. 620-626
[243]
A. Sergio, C. Cristofori, R. Cardin, G. Pivetta, R. Ragazzi, A. Baldan, et al.
Transcatheter Arterial Chemoembolization (TACE) in Hepatocellular Carcinoma (HCC): the role of angiogenesis and invasiveness.
Am J Gastroenterol, 103 (2008), pp. 914-921
[244]
A. Erhardt, F. Kolligs, M. Dollinger, E. Schott, H. Wege, M. Bitzer, et al.
TACE plus sorafenib for the treatment of hepatocellular carcinoma: results of the multicenter, phase II SOCRATES trial.
Cancer Chemother Pharmacol, 74 (2014), pp. 947-954
[245]
Y. Chao, Y.-H. Chung, G. Han, J.-H. Yoon, J. Yang, J. Wang, et al.
The combination of transcatheter arterial chemoembolization and sorafenib is well tolerated and effective in Asian patients with hepatocellular carcinoma: final results of the START trial.
Int J Cancer, 136 (2014), pp. 1458-1467
[246]
R. Lencioni, J.M. Llovet, G. Han, W.Y. Tak, J. Yang, A. Guglielmi, et al.
Sorafenib or placebo plus TACE with doxorubicin-eluting beads for intermediate stage HCC: The SPACE trial.
J Hepatol, 64 (2016), pp. 1090-1098
[247]
K. Hoffmann.
Impact of neo-adjuvant Sorafenib treatment on liver transplantation in HCC patients – a prospective, randomized, double-blind, phase III trial.
[248]
M. Kudo, K. Ueshima, M. IKEDA, T. Torimura, N. Tanabe, H. Aikata, et al.
Randomised, multicentre prospective trial of transarterial chemoembolisation (TACE) plus sorafenib as compared with TACE alone in patients with hepatocellular carcinoma: TACTICS trial.
[249]
T.M. Pawlik, D.K. Reyes, D. Cosgrove, I.R. Kamel, N. Bhagat, J.F.H. Geschwind.
Phase II trial of sorafenib combined with concurrent transarterial chemoembolization with drug-eluting beads for hepatocellular carcinoma.
J Clin Oncol, 29 (2011), pp. 3960-3967
[250]
J.-W. Park, Y.J. Kim, Do Young Kim, S.-H. Bae, S.W. Paik, Y.-J. Lee, et al.
Sorafenib with or without concurrent transarterial chemoembolization in patients with advanced hepatocellular carcinoma: the phase III STAH trial.
J Hepatol, 70 (2019), pp. 684-691
[251]
M. Kudo, G. Han, R.S. Finn, R.T.P. Poon, J.-F. Blanc, L. Yan, et al.
Brivanib as adjuvant therapy to transarterial chemoembolization in patients with hepatocellular carcinoma: a randomized phase III trial.
Hepatology, 60 (2014), pp. 1697-1707
[252]
M. Pinter, G. Ulbrich, W. Sieghart, C. Kölblinger, T. Reiberger, S. Li, et al.
Hepatocellular carcinoma: a phase II randomized controlled double-blind trial of transarterial chemoembolization in combination with biweekly intravenous administration of bevacizumab or a placebo.
Radiology, 277 (2015), pp. 903-912
[253]
S.L. Chan, W. Yeo, F. Mo, A.W.H. Chan, J. Koh, L. Li, et al.
A phase 2 study of the efficacy and biomarker on the combination of transarterial chemoembolization and axitinib in the treatment of inoperable hepatocellular carcinoma.
Cancer, 123 (2017), pp. 3977-3985
[254]
M. Kudo, K. Ueshima, O. Yokosuka, S. Ogasawara, S. Obi, N. Izumi, et al.
Sorafenib plus low-dose cisplatin and fluorouracil hepatic arterial infusion chemotherapy versus sorafenib alone in patients with advanced hepatocellular carcinoma (SILIUS): a randomised, open label, phase 3 trial.
Lancet Gastroenterol Hepatol, 3 (2018), pp. 424-432
[255]
M. He, Q. Li, R. Zou, J. Shen, W. Fang, G. Tan, et al.
Sorafenib plus hepatic arterial infusion of oxaliplatin, fluorouracil, and leucovorin vs sorafenib alone for hepatocellular carcinoma with portal vein invasion.
[256]
L. Kulik, M. Vouche, S. Koppe, R.J. Lewandowski, M.F. Mulcahy, D. Ganger, et al.
Prospective randomized pilot study of Y90+/− sorafenib as bridge to transplantation in hepatocellular carcinoma.
J Hepatol, 61 (2014), pp. 309-317
[257]
C. Toso, S. Cader, A. Mentha-Dugerdil, G. Meeberg, P. Majno, I. Morard, et al.
Factors predicting survival after post-transplant hepatocellular carcinoma recurrence.
J Hepatobiliary Pancreat Sci, 20 (2012), pp. 342-347
[258]
S. Roayaie, J.D. Schwartz, M.W. Sung, S.H. Emre, C.M. Miller, G.E. Gondolesi, et al.
Recurrence of hepatocellular carcinoma after liver transplant: patterns and prognosis.
Liver Transplant, 10 (2004), pp. 534-540
[259]
V.V. Mazzaferro, E.E. Regalia, R.R. Doci, S.S. Andreola, A.A. Pulvirenti, F.F. Bozzetti, et al.
Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis.
N Engl J Med, 334 (1996), pp. 693-699
[260]
V. Mazzaferro, S. Bhoori, C. Sposito, M. Bongini, M. Langer, R. Miceli, et al.
Milan criteria in liver transplantation for hepatocellular carcinoma: an evidence-based analysis of 15 years of experience.
Liver Transplant, 17 (2011), pp. S44-S57
[261]
L. McCormack, A. Gadano, J. Lendoire, E. Quiñonez, O. Imventarza, O. Andriani, et al.
Model for end-stage liver disease exceptions committee activity in Argentina: does it provide justice and equity among adult patients waiting for a liver transplant?.
[262]
N.G. Cejas, F.G. Villamil, J.C. Lendoire, V. Tagliafichi, A. Lopez, D.H. Krogh, et al.
Improved waiting-list outcomes in Argentina after the adoption of a model for end-stage liver disease-based liver allocation policy.
Liver Transplant, 19 (2013), pp. 711-720
[263]
F. Piñero, S. Marciano, M. Anders, F. Orozco Ganem, A. Zerega, J. Cagliani, et al.
Identifying patients at higher risk of hepatocellular carcinoma recurrence after liver transplantation in a multicenter cohort study from Argentina.
Eur J Gastroenterol Hepatol, 28 (2016), pp. 421-427
[264]
F.Y. Yao, L. Ferrell, N.M. Bass, J.J. Watson, P. Bacchetti, A. Venook, et al.
Liver transplantation for hepatocellular carcinoma: expansion of the tumor size limits does not adversely impact survival.
Hepatology, 33 (2001), pp. 1394-1403
[265]
F.Y. Yao, N.M. Bass, B. Nikolai, T.J. Davern, R. Kerlan, V. Wu, et al.
Liver transplantation for hepatocellular carcinoma: analysis of survival according to the intention-to-treat principle and dropout from the waiting list.
Liver Transplant, 8 (2002), pp. 873-883
[266]
C. Toso, S. Asthana, D.L. Bigam, A.M.J. Shapiro, N.M. Kneteman.
Reassessing selection criteria prior to liver transplantation for hepatocellular carcinoma utilizing the scientific registry of transplant recipients database.
Hepatology, 49 (2008), pp. 832-838
[267]
K. Berry, G.N. Ioannou.
Serum alpha-fetoprotein level independently predicts posttransplant survival in patients with hepatocellular carcinoma.
Liver Transplant, 19 (2013), pp. 634-645
[268]
X. Xu, D. Lu, Q. Ling, X. Wei, J. Wu, L. Zhou, et al.
Liver transplantation for hepatocellular carcinoma beyond the Milan criteria.
[269]
G. Sapisochin, N. Goldaracena, J.M. Laurence, M. Dib, A. Barbas, A. Ghanekar, et al.
The extended Toronto criteria for liver transplantation in patients with hepatocellular carcinoma: a prospective validation study.
Hepatology, 64 (2016), pp. 2077-2088
[270]
V. Mazzaferro, J.M. Llovet, R. Miceli, S. Bhoori, M. Schiavo, L. Mariani, et al.
Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis.
Lancet Oncol, 10 (2009), pp. 35-43
[271]
F. Piñero, M. Tisi Baña, E.C. de Ataide, S. Hoyos Duque, S. Marciano, A. Varón, et al.
Liver transplantation for hepatocellular carcinoma: evaluation of the AFP model in a multicenter cohort from Latin America.
Liver Int, 36 (2016), pp. 1657-1667
[272]
A. Notarpaolo, R. Layese, P. Magistri, M. Gambato, M. Colledan, G. Magini, et al.
Validation of the AFP model as a predictor of HCC recurrence in patients with viral hepatitis-related cirrhosis who had received a liver transplant for HCC.
J Hepatol, 66 (2017), pp. 552-559
[273]
J. Rhu, J.M. Kim, G.S. Choi, Kwon C-HD, J.-W. Joh.
Validation of the α-fetoprotein model for hepatocellular carcinoma recurrence after transplantation in an Asian Population.
Transplantation, 102 (2018), pp. 1316-1322
[274]
T. Decaens, F. Roudot-Thoraval, S. Bresson-Hadni, C. Meyer, J. Gugenheim, F. Durand, et al.
Impact of pretransplantation transarterial chemoembolization on survival and recurrence after liver transplantation for hepatocellular carcinoma.
Liver Transplant, 11 (2005), pp. 767-775
[275]
A. Vitale, F. D’Amico, A.C. Frigo, F. Grigoletto, A. Brolese, G. Zanus, et al.
Response to therapy as a criterion for awarding priority to patients with hepatocellular carcinoma awaiting liver transplantation.
Ann Surg Oncol, 17 (2010), pp. 2290-2302
[276]
A. Cucchetti, M. Cescon, E. Bigonzi, F. Piscaglia, R. Golfieri, G. Ercolani, et al.
Priority of candidates with hepatocellular carcinoma awaiting liver transplantation can be reduced after successful bridge therapy.
Liver Transplant, 17 (2011), pp. 1344-1354
[277]
M.L. Samoylova, J.L. Dodge, F.Y. Yao, J.P. Roberts.
Time to transplantation as a predictor of hepatocellular carcinoma recurrence after liver transplantation.
Liver Transplant, 20 (2014), pp. 937-944
[278]
P. Majno, R. Lencioni, F. Mornex, N. Girard, R.T. Poon, D. Cherqui.
Is the treatment of hepatocellular carcinoma on the waiting list necessary?.
Liver Transplant, 17 (2011), pp. S98-S108
[279]
T. Bittermann, M.A. Hoteit, P.L. Abt, K.A. Forde, D. Goldberg.
Waiting time and explant pathology in transplant recipients with hepatocellular carcinoma: a novel study using national data.
Am J Transplant, 14 (2014), pp. 1657-1663
[280]
N. Mehta, J.L. Dodge, A. Goel, J.P. Roberts, R. Hirose, F.Y. Yao.
Identification of liver transplant candidates with hepatocellular carcinoma and a very low dropout risk: implications for the current organ allocation policy.
Liver Transplant, 19 (2013), pp. 1343-1353
[281]
F.Y. Yao, R. Hirose, J.M. LaBerge, T.J. Davern, N.M. Bass, R.K. Kerlan, et al.
A prospective study on downstaging of hepatocellular carcinoma prior to liver transplantation.
Liver Transplant, 11 (2005), pp. 1505-1514
[282]
F.Y. Yao, R.K. Kerlan Jr., R. Hirose, T.J. Davern III, N.M. Bass, S. Feng, et al.
Excellent outcome following down-staging of hepatocellular carcinoma prior to liver transplantation: an intention-to-treat analysis.
Hepatology, 48 (2008), pp. 819-827
[283]
M. Ravaioli, G.L. Grazi, F. Piscaglia, F. Trevisani, M. Cescon, G. Ercolani, et al.
Liver transplantation for hepatocellular carcinoma: results of down-staging in patients initially outside the milan selection criteria.
Am J Transplant, 8 (2008), pp. 2547-2557
[284]
O. Barakat, R.P. Wood, C.F. Ozaki, V. Ankoma-Sey, J. Galati, M. Skolkin, et al.
Morphologic features of advanced hepatocellular carcinoma as a predictor of downstaging and Liver Transplantation: an intention-to-treat analysis.
Liver Transplant, (2010),
[285]
F.Y. Yao, N. Mehta, J. Flemming, J. Dodge, B. Hameed, O. Fix, et al.
Downstaging of hepatocellular cancer before liver transplant: long-term outcome compared to tumors within Milan criteria.
Hepatology, 61 (2015), pp. 1968-1977
[286]
F. Yao.
A follow-up analysis of the pattern and predictors of dropout from the waiting list for liver transplantation in patients with hepatocellular carcinoma: implications for the current organ allocation policy.
Liver Transplant, 9 (2003), pp. 684-692
[287]
N.D. Parikh, A.K. Waljee, A.G. Singal.
Downstaging hepatocellular carcinoma: a systematic review and pooled analysis.
Liver Transplant, 21 (2015), pp. 1142-1152
[288]
J. Sinha, N. Mehta, J.L. Dodge, E. Poltavskiy, J. Roberts, F. Yao.
Are there upper limits in tumor burden for down-staging of hepatocellular carcinoma to liver transplant? Analysis of the all-comers protocol.
Hepatology, 70 (2019), pp. 1185-1196
[289]
N. Mehta, J.L. Dodge, J.D. Grab, F.Y. Yao.
National experience on down-staging of hepatocellular carcinoma before liver transplant: influence of tumor burden, AFP, and wait time.
[290]
F.Y. Yao, S. Breitenstein, C.E. Broelsch, J.-F. Dufour, M. Sherman.
Does a patient qualify for liver transplantation after the down-staging of hepatocellular carcinoma?.
Liver Transplant, 17 (2011), pp. S109-S116
[291]
Radioembolization Results in Longer Time-to-Progression and Reduced Toxicity Compared With Chemoembolization in Patients With Hepatocellular Carcinoma 2011;140:497–507.e2. doi:10.1053/j.gastro.2010.10.049.
[292]
J. Xu, Z.-Y. Shen, X.-G. Chen, Q. Zhang, H.-J. Bian, P. Zhu, et al.
A randomized controlled trial of licartin for preventing hepatoma recurrence after liver transplantation.
Hepatology, 45 (2007), pp. 269-276
[293]
A.B. Siegel, A.B. El-Khoueiry, R.S. Finn, K.A. Guthrie, A. Goyal, A.P. Venook, et al.
Phase I trial of sorafenib following liver transplantation in patients with high-risk hepatocellular carcinoma.
Liver Cancer, 4 (2015), pp. 115-125
[294]
A.E. Alsina, A. Makris, V. Nenos, E. Sucre, J. Arrobas, E. Franco, et al.
Can sorafenib increase survival for recurrent hepatocellular carcinoma after liver transplantation? A pilot study.
Am Surg, 80 (2014), pp. 680-684
[295]
C. Toso, G.A. Meeberg, D.L. Bigam, J. Oberholzer, A.M.J. Shapiro, K. Gutfreund, et al.
De novo sirolimus-based immunosuppression after liver transplantation for hepatocellular carcinoma: long-term outcomes and side effects.
Transplantation, 83 (2007), pp. 1162-1168
[296]
C. Toso, S. Merani, D.L. Bigam, A.M.J. Shapiro, N.M. Kneteman.
Sirolimus-based immunosuppression is associated with increased survival after liver transplantation for hepatocellular carcinoma.
Hepatology, 51 (2009), pp. 1237-1243
[297]
C. Toso, G. Mentha, Pietro Majno.
Integrating sorafenib into an algorithm for the management of post-transplant hepatocellular carcinoma recurrence.
[298]
E.K. Geissler, A.A. Schnitzbauer, C. Zülke, P.E. Lamby, A. Proneth, C. Duvoux, et al.
Sirolimus use in liver transplant recipients with hepatocellular carcinoma: a randomized, multicenter, open-label phase 3 trial.
Transplantation, 100 (2017), pp. 116-125
[299]
N. Kneteman, T. Livraghi, D. Madoff, E. de Santibañez, M. Kew.
Tools for monitoring patients with hepatocellular carcinoma on the waiting list and after liver transplantation.
Liver Transplant, 17 (2011), pp. S117-S127
[300]
T. Decaens, F. Roudot-Thoraval, H. Badran, P. Wolf, F. Durand, R. Adam, et al.
Impact of tumour differentiation to select patients before liver transplantation for hepatocellular carcinoma.
[301]
E. Davis, R. Wiesner, J. Valdecasas, Y. Kita, M. Rossi, M. Schwartz.
Treatment of recurrent hepatocellular carcinoma after liver transplantation.
Liver Transplant, 17 (2011), pp. S162-S166
[302]
C. Sposito, L. Mariani, A. Germini, M.F. Reyes, M. Bongini, G. Grossi, et al.
Comparative efficacy of sorafenib versus best supportive care in recurrent hepatocellular carcinoma after liver transplantation: a case–control study.
J Hepatol, 59 (2013), pp. 59-66
[303]
A. Waghray, B. Balci, G. El-Gazzaz, R. Kim, R. Pelley, K.V. Narayanan Menon, et al.
Safety and efficacy of sorafenib for the treatment of recurrent hepatocellular carcinoma after liver transplantation.
Clin Transplant, 27 (2013), pp. 555-561
[304]
A. Vitale, P. Boccagni, X. Kertusha, G. Zanus, F. D’Amico, E. Lodo, et al.
Sorafenib for the treatment of recurrent hepatocellular carcinoma after liver transplantation?.
Transplant Proc, 44 (2012), pp. 1989-1991
[305]
K. Staufer, L. Fischer, B. Seegers, E. Vettorazzi, B. Nashan, M. Sterneck.
High toxicity of sorafenib for recurrent hepatocellular carcinoma after liver transplantation.
Transplant Int, 25 (2012), pp. 1158-1164
[306]
M. Iavarone, F. Invernizzi, C. Czauderna, M. Sanduzzi Zamparelli, S. Bhoori, G. Amaddeo, et al.
Preliminary experience on safety of regorafenib after sorafenib failure in recurrent hepatocellular carcinoma after liver transplantation.
Am J Transplant, 19 (2019), pp. 3176-3184
[307]
Y. Liu, H. Yue, S. Xu, F. Wang, N. Ma, K. Li, et al.
First-line gemcitabine and oxaliplatin (GEMOX) plus sorafenib, followed by sorafenib as maintenance therapy, for patients with advanced hepatocellular carcinoma: a preliminary study.
Int J Clin Oncol, 20 (2015), pp. 952-959
[308]
E. Assenat, G.-P. Pageaux, S. Thézenas, J.-M. Péron, Y. Bécouarn, J.-F. Seitz, et al.
Sorafenib alone vs. sorafenib plus GEMOX as 1.
Br J Cancer, (2019), pp. 1-7
[309]
A. Liccioni, M. Reig, J. Bruix.
FOLFOX-4 vs. doxorubicin for hepatocellular carcinoma: Could a negative result be accepted as positive?.
J Hepatol, 61 (2014), pp. 164-165
[310]
A.-L. Cheng, Y.-K. Kang, Z. Chen, C.-J. Tsao, S. Qin, J.S. Kim, et al.
Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial.
Lancet Oncol, 10 (2009), pp. 25-34
[311]
F. Piñero, S. Marciano, N. Fernández, J. Silva, M. Anders, A. Zerega, et al.
Intermediate-advanced hepatocellular carcinoma in Argentina: treatment and survival analysis.
[312]
M. Iavarone, G. Cabibbo, F. Piscaglia, C. Zavaglia, A. Grieco, E. Villa, et al.
Field-practice study of sorafenib therapy for hepatocellular carcinoma: a prospective multicenter study in Italy.
Hepatology, 54 (2011), pp. 2055-2063
[313]
R. Lencioni, M. Kudo, S.L. Ye, J.P. Bronowicki, X.P. Chen, L. Dagher, et al.
GIDEON (Global Investigation of therapeutic DEcisions in hepatocellular carcinoma and Of its treatment with sorafeNib): second interim analysis.
Int J Clin Pract, 68 (2013), pp. 609-617
[314]
M. Reig, T. Ferran, C. Rodríguez-Lope, A. Forner, N. LLarch, J. Rimola, et al.
Early dermatologic adverse events predict better outcome in HCC patients treated with sorafenib.
J Hepatol, 61 (2014), pp. 318-324
[315]
J.A. Marrero, M. Kudo, A.P. Venook, S.-L. Ye, J.-P. Bronowicki, X.-P. Chen, et al.
Observational registry of sorafenib use in clinical practice across Child-Pugh subgroups: the GIDEON study.
[316]
A.-L. Cheng, Y.-K. Kang, D.-Y. Lin, J.-W. Park, M. Kudo, S. Qin, et al.
Sunitinib versus sorafenib in advanced hepatocellular cancer: results of a randomized phase III trial.
J Clin Oncol, 31 (2013), pp. 4067-4075
[317]
P.J. Johnson, S. Qin, J.-W. Park, R.T.P. Poon, J.-L. Raoul, P.A. Philip, et al.
Brivanib versus sorafenib as first-line therapy in patients with unresectable, advanced hepatocellular carcinoma: results from the randomized phase III BRISK-FL study.
J Clin Oncol, 31 (2013), pp. 3517-3524
[318]
A.X. Zhu, O. Rosmorduc, T.R.J. Evans, P.J. Ross, A. Santoro, F.J. Carrilho, et al.
SEARCH: a phase III, randomized, double-blind, placebo-controlled trial of sorafenib plus erlotinib in patients with advanced hepatocellular carcinoma.
J Clin Oncol, 33 (2015), pp. 559-566
[319]
C. Cainap, S. Qin, W.-T. Huang, I.J. Chung, H. Pan, Y. Cheng, et al.
Linifanib versus sorafenib in patients with advanced hepatocellular carcinoma: results of a randomized phase III trial.
J Clin Oncol, 33 (2015), pp. 172-179
[320]
A.L. Cheng, S. Thongprasert, H.Y. Lim, W. Sukeepaisarnjaroen, T.S. Yang, C.C. Wu, et al.
Randomized, open-label phase 2 study comparing frontline dovitinib versus sorafenib in patients with advanced hepatocellular carcinoma.
Hepatology, 64 (2016), pp. 774-784
[321]
J.M. Hubbard, M.R. Mahoney, W.S. Loui, L.R. Roberts, T.C. Smyrk, Z. Gatalica, et al.
Phase I/II randomized trial of sorafenib and bevacizumab as first-line therapy in patients with locally advancedor metastatic hepatocellular carcinoma: north central cancer treatment group trial N0745 (Alliance).
Target Oncol, 12 (2016), pp. 201-209
[322]
M. Kudo, R.S. Finn, S. Qin, K.-H. Han, K. Ikeda, F. Piscaglia, et al.
Lenvatinib versus sorafenib in rst-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial.
Lancet, 391 (2018), pp. 1163-1173
[323]
K. Ikeda, M. Kudo, S. Kawazoe, Y. Osaki, M. IKEDA, T. Okusaka, et al.
Phase 2 study of lenvatinib in patients with advanced hepatocellular carcinoma.
J Gastroenterol, 52 (2016), pp. 512-519
[324]
R. Finn, S. Qin, M. Ikeda, P. Galle, M. Ducreux, T. Kim, et al.
Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma.
New Eng J Med, 382 (2020), pp. 1894-1905
[325]
J.M. Llovet, T. Decaens, J.-L. Raoul, E. Boucher, M. Kudo, C. Chang, et al.
Brivanib in patients with advanced hepatocellular carcinoma who were intolerant to sorafenib or for whom sorafenib failed: results from the randomized phase III BRISK-PS study.
J Clin Oncol, 31 (2013), pp. 3509-3516
[326]
Y.K. Kang, T. Yau, J.W. Park, H.Y. Lim, T.Y. Lee, S. Obi, et al.
Randomized phase II study of axitinib versus placebo plus best supportive care in second-line treatment of advanced hepatocellular carcinoma.
Ann Oncol, 18 (2015), pp. 2290-2300
[327]
A.X. Zhu, M. Kudo, E. Assenat, S. Cattan, Y.-K. Kang, H.-Y. Lim, et al.
Effect of everolimus on survival in advanced hepatocellular carcinoma after failure of sorafenib.
[328]
L. Rimassa, E. Assenat, M. Peck-Radosavljevic, M. Pracht, V. Zagonel, P. Mathurin, et al.
Articles Tivantinib for second-line treatment of MET-high, advanced hepatocellular carcinoma (METIV-HCC): a final analysis of a phase 3, randomised, placebo-controlled study.
Lancet Oncol, 19 (2018), pp. 682-693
[329]
J. Bruix, S. Qin, P. Merle, A. Granito, Y.-H. Huang, G. Bodoky, et al.
Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial.
[330]
A.X. Zhu, Y.-K. Kang, C.-J. Yen, R.S. Finn, D.B. Shin, P.R. Galle, et al.
Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased (-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial.
Lancet Oncol, 20 (2019), pp. 282-296
[331]
B. Sangro, C. Gomez-Martin, M. la Mata de, M. Iñarrairaegui, E. Garralda, P. Barrera, et al.
A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C.
J Hepatol, 59 (2013), pp. 81-88
[332]
A.B. El-Khoueiry, B. Sangro, T. Yau, T. Crocenzi, M. Kudo, C. Hsu, et al.
Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial.
Lancet, 389 (2017), pp. 2492-2502
[333]
R.S. Finn, B.-Y. Ryoo, P. Merle, M. Kudo, M. Bouattour, H.-Y. Lim, et al.
Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase III trial.
J Clin Oncol, 38 (2020), pp. 193-202
[334]
J.J. Bruix, M.M. Reig, J.J. Rimola, A.A. Forner, M.M. Burrel, R.R. Vilana, et al.
Clinical decision making and research in hepatocellular carcinoma: pivotal role of imaging techniques.
Hepatology, 54 (2011), pp. 2238-2244
[335]
M. Reig, J. Rimola, T. Ferrran, A. Darnell, C. Rodríguez-Lope, A. Forner, et al.
Postprogression survival of patients with advanced hepatocellular carcinoma: rationale for second-line trial design.
Hepatology, 58 (2013), pp. 2023-2031
[336]
M. Iavarone, G. Cabibbo, M. Biolato, C. Corte Della, M. Maida, M. Barbara, et al.
Predictors of survival in patients with advanced hepatocellular carcinoma who permanently discontinued sorafenib.
Hepatology, 62 (2015), pp. 784-791
[337]
R. Lencioni, J. Llovet.
Modified RECIST (mRECIST) assessment for hepatocellular carcinoma.
Semin Liver Dis, 30 (2010), pp. 052-60
[338]
M. Vouche, L. Kulik, R. Atassi, K. Memon, R. Hickey, D. Ganger, et al.
Radiological-pathological analysis of WHO, RECIST, EASL, mRECIST and DWI: imaging analysis from a prospective randomized trial of Y90±sorafenib.
Hepatology, 58 (2013), pp. 1655-1666
[339]
R. Lencioni, R. Montal, T. Ferrran, J.-W. Park, T. Decaens, J.-L. Raoul, et al.
Objective response by mRECIST as a predictor and potential surrogate end-point of overall survival in advanced HCC.
J Hepatol, 66 (2017), pp. 1166-1172
[340]
J. Choi, J.H. Shim, Y.M. Shin, K.M. Kim, Y.-S. Lim, H.C. Lee.
Clinical significance of the best response during repeated transarterial chemoembolization in the treatment of hepatocellular carcinoma.
J Hepatol, 60 (2014), pp. 1212-1218
[341]
E.S. Jung, J.H. Kim, E.L. Yoon, H.J. Lee, S.J. Lee, S.J. Suh, et al.
Comparison of the methods for tumor response assessment in patients with hepatocellular carcinoma undergoing transarterial chemoembolization.
J Hepatol, 58 (2013), pp. 1181-1187
[342]
W. Sieghart, F. Hucke, M. Pinter, I. Graziadei, W. Vogel, C. Müller, et al.
The ART of decision making: retreatment with transarterial chemoembolization in patients with hepatocellular carcinoma.
Hepatology, 57 (2013), pp. 2261-2273
[343]
F. Hucke, M. Pinter, I. Graziadei, W. Vogel, C. Müller, H. Heinzl, et al.
How to STATE suitability and START transarterial chemoembolization in patients with intermediate stage hepatocellular carcinoma.
[344]
L. Kadalayil, R. Benini, L. Pallan, J. O’Beirne, L. Marelli, D. Yu, et al.
A simple prognostic scoring system for patients receiving transarterial embolisation for hepatocellular cancer.
Ann Oncol, 24 (2013), pp. 2565-2570
[345]
A. Hiraoka, T. Kumada, M. Kudo, M. Hirooka, Y. Koizumi, Y. Hiasa, et al.
Hepatic function during repeated tace procedures and prognosis after introducing sorafenib in patients with unresectable hepatocellular carcinoma: multicenter analysis.
Dig Dis, 35 (2017), pp. 602-610
[346]
National Cancer Institute.
Common terminology criteria for adverse events (CTCAE).
(2010), pp. 1-196
[347]
A.D. González, M. Sanduzzi Zamparelli, V. Sapena, T. Ferrran, N. LLarch, I. Gemma, et al.
Systematic review with meta-analysis: the critical role of dermatological events in patients with hepatocellular carcinoma treated with sorafenib.
Aliment Pharmacol Ther, (2018), pp. 1-10
[348]
J. Brahmer, C. Lacchetti, B.J. Schneider, M.B. Atkins, K.J. Brassil, J.M. Caterino, et al.
Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American society of clinical oncology clinical practice guideline.
J Clin Oncol, 36 (2018), pp. 1714-1768
[349]
T. Yau, C. Hsu, T.-Y. Kim, S.-P. Choo, Y.-K. Kang, M.-M. Hou, et al.
Nivolumab in advanced hepatocellular carcinoma: sorafenib-experienced Asian cohort analysis.
J Hepatol, 71 (2019), pp. 543-552
[350]
A. Rammohan, M.S. Reddy, M. Farouk, J. Vargese, M. Rela.
Pembrolizumab for metastatic hepatocellular carcinoma following live donor liver transplantation: the silver bullet?.
Hepatology, 67 (2018), pp. 1166-1168
[351]
B. Sangro, S.L. Chan, T. Meyer, M. Reig, A. El-Khoueiry, P.R. Galle.
Diagnosis and management of toxicities of immune checkpoint inhibitors in hepatocellular carcinoma.
J Hepatol, 72 (2020), pp. 320-341
[352]
W.E. Naugler, A.E. Alsina, C.T. Frenette, L. Rossaro, M.T. Sellers.
Building the multidisciplinary team for management of patients with hepatocellular carcinoma.
Clin Gastroenterol Hepatol, 13 (2015), pp. 827-835
[353]
J. Guy, R.K. Kelley, J. Roberts, R. Kerlan, F. Yao, N. Terrault.
Multidisciplinary management of hepatocellular carcinoma.
Clin Gastroenterol Hepatol, 10 (2012), pp. 354-362
[354]
T.T. Chang, R. Sawhney, A. Monto, J.B. Davoren, J.G. Kirkland, L. Stewart, et al.
Implementation of a multidisciplinary treatment team for hepatocellular cancer at a Veterans Affairs Medical Center improves survival.
HPB (Oxford), 10 (2008), pp. 405-411
[355]
K.K. Christians, H.A. Pitt, W.S. Rilling, J. Franco, F.A. Quiroz, M.B. Adams, et al.
Hepatocellular carcinoma: multimodality management.
Surgery, 130 (2001), pp. 554-560
[356]
O. Campollo, S. Roman.
Consensus and clinical practice guidelines in Latin America: who, where and how.
Ann Hepatol, 19 (2019), pp. 1-2
[357]
A. Panduro, S. Roman.
Advancements in genomic medicine and the need for updated regional clinical practice guidelines in the field of hepatology.
Ann Hepatol, 19 (2020), pp. 1-2
[358]
S. Roman.
Occult hepatitis B and other unexplored risk factors for hepatocellular carcinoma in Latin America.
Ann Hepatol, 17 (2018), pp. 541-543
[359]
J.-C. Nault, Y. Martin, S. Caruso, T.Z. Hirsch, Q. Bayard, J. Calderaro, et al.
Clinical impact of genomic diversity from early to advanced hepatocellular carcinoma.
Hepatology, 71 (2020), pp. 164-182
[360]
J. Bayo, E.J. Fiore, L.M. Dominguez, A. Real, M. Malvicini, M. Rizzo, et al.
A comprehensive study of epigenetic alterations in hepatocellular carcinoma identifies potential therapeutic targets.
J Hepatol, 71 (2019), pp. 78-90
[361]
Z. Yao, Y. Dong, G. Wu, Q. Zhang, D. Yang, J.-H. Yu, et al.
Preoperative diagnosis and prediction of hepatocellular carcinoma: radiomics analysis based on multi-modal ultrasound images.
BMC Cancer, 18 (2018), pp. 1089

Azcuénaga 1222 (1115), Ciudad de Buenos Aires, Argentina. E-mail: secretaria@aaeeh.org.ar.

Copyright © 2020. Fundación Clínica Médica Sur, A.C.
Opciones de artículo
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

Quizás le interese:
10.1016/j.aohep.2023.101185
No mostrar más