Show PDF
Show PDF
BCLC strategy for prognosis prediction and treatment recommendations: The 2025
update
PII: S0168-8278(25)02571-1
DOI: https://doi.org/10.1016/j.jhep.2025.10.020
Reference: JHEPAT 10323
Please cite this article as: Reig M, Sanduzzi-Zamparelli M, Forner A, Rimola J, Ferrer-Fàbrega J,
Burrel M, Garcia-Criado Á, Díaz A, Llarch N, Iserte G, Mollà M, Kelley RK, Galle PR, Mazzaferro V,
Salem R, Sangro B, Singal A, Vogel A, Yanagihara TK, Ayuso C, Torres F, Bruix J, BCLC strategy for
prognosis prediction and treatment recommendations: The 2025 update, Journal of Hepatology, https://
doi.org/10.1016/j.jhep.2025.10.020.
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.
© 2025 European Association for the Study of the Liver. Published by Elsevier B.V. All rights are
reserved, including those for text and data mining, AI training, and similar technologies.
Title: BCLC strategy for prognosis prediction and treatment recommendations:
The 2025 update
f
Amit Singal15, Arndt Vogel16,17, Ted K. Yanagihara18,19, Carmen Ayuso1,3,5, Ferran
oo
Torres20, Jordi Bruix1,3,4
1BCLC
pr
group. Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS),
-
re
Barcelona, Spain; 2Liver Oncology Unit, Liver Unit, ICMDM, Hospital Clínic Barcelona,
Barcelona, Spain; 3University of Barcelona, Barcelona, Spain; 4Centro de
lP
IDIBAPS, Hospital Clínic, c/ Villarroel, 170, Escala 11, 4a planta, 08036 Barcelona.
Spain. Tel.: +34 932279803, fax: +34 932275792. E-mail address: [email protected]
(M. Reig)
f
oo
Keywords: HCC, survival, BCLC, ablation, surgery, liver transplantation TACE,
TARE, EBRT, systemic treatment, immunotherapy, ALBI score, AFP, CUSE.
- pr
re
Electronic word count: 9418
lP
Eli Lilly, Gilead, Roche, Biotoscana Farma; lecture fees from Bayer, BMS, Gilead,
Jo
AstraZeneca Roche, Guerbet and Lilly and research grants (to the Institution) from
Bayer, ROCHE and Ipsen. Educational Support (to the institution): Bayer,
Astrazeneca, Eisai- Merck MSD, Roche, Ipsen, Lilly, Terumo, BMS, Next, Boston
Scientific, Ciscar Medical, Eventy C3 LLC (Egypt), Servier, Guerbet; MSZ: received
consultancy/advisory fee from Captor Therapeutics, speaker fees from Bayer,
AstraZeneca and Roche and travel grants from Bayer, BTG, Eisai and Roche; AF:
received advisory fee from AstraZeneca, Incyte, Boston Scientific and Taiho and
speaker fees from AstraZeneca, Roche and Boston Scientific; JR: has consulted for
Roche and received speaker fees from Bayer and Roche and travel grants from Bayer
JFF; received Lecture fees from AstraZeneca; MB: received honoraria from: Terumo,
Boston Scientific, Guerbet, Bayer, Astrazeneca and Travel support: Terumo, Boston
Scientific; AGC: received lectures fee from Roche; AD: ; none NLL: received
consultancy fees from Bayer, Universal DX and AstraZeneca, speaker fees from
2
Roche and AstraZeneca, travel funding from Bayer and congress registration from
Eisai; GI: received travel grants from Bayer and received speaker fees from ROCHE
MM; I have received honoraria as a speaker and moderator in educational sessions
from the Clinic Foundation for Biomedical Research (Clinic Barcelona); RK; received
consulting fee from: Compass Therapeutics, CVS Caremark, Elevar, GSK, Jazz, J
Pharma, Moderna, Regeneron, Tyra Biosciences; travel grants from: Astra Zeneca,
Merck; Data Safety Monitoring Board from: Genentech/Roche; PG; received
honoraria from Bayer, Boston Scientific, AstraZeneca, Adaptimmune, BMS, Eisai,
MSD, Sirtex, Lilly, Roche, Guerbet and grants from ; VM; none; RS: received
honoraria from: Boston Scientific, Cook, Bard, Genentech, Astrazeneca, Eisai, Autem,
f
Terumo, Astellas, Siemens, Sirtex, Trisalus, Terumo; BS; reports grants from Bristol
oo
Myers Squibb and Roche; consulting fees from AstraZeneca, Bayer, Boston Scientific,
pr
Bristol Myers Squibb, Eisai, Incyte, Roche/Genentech, Sanofi Pasteur, and Sirtex
Medical; honoraria from AstraZeneca, Eisai, Incyte, and Roche; and travel support
-
re
from AstraZeneca, Bristol Myers Squibb, Eisai, Roche, and Sirtex Medical; AS: has
served as a consultant or on advisory boards for Genentech, AstraZeneca, Eisai,
lP
Bayer, Exelixis, Merck, Elevar, Boston Scientific, Sirtex, FujiFilm Medical Sciences,
Exact Sciences, Helio Genomics, Curve Biosciences, Glycotest, Abbott, DELFI,
na
Mursla Bio, ImCare, and Universal Dx; AV; Roche, AstraZenca, Böhringer-Ingelheim,
ur
Ipsen, Incyte, Cogent, EISAI, Zymeworks, Biologix, BMS, Terumo, Elevar, Servier,
MSD, Tahio, Jazzpharma, Medivir, Abbvie, Tyra, Falk, Janssen, Lilly; Roche,
Jo
f
PRYCO234831); MSZ: received grant support from Instituto de Salud Carlos III
oo
(FI19/00222) and Centro de Investigación Biomédica en Red en Enfermedades
pr
Hepáticas y Digestivas - CIBEREHD (EHD25PI02); AF: Grant from Instituto de Salud
Carlos III (PI15/01229 and PI18/00542) and from Centro de Investigación Biomédica
-
re
en Red - CIBER (Immune4Al, S2300092_3); JR: registry grant from European
Association for the Study of the Liver (EASL); JFF; none; MB; none; AGC: none; AD:
lP
iniciació a la recerca 2022” from Societat Catalana de Digestologia (SCD); MM; none;
ur
RK; Agios, Astra Zeneca, Bayer, BMS, Compass Therapeutics, Eli Lilly, EMD Serono,
Exelixis, Genentech/Roche, Merck, Partner Therapeutics, QED, Relay Therapeutics,
Jo
Servier, Surface Oncology, Taiho, Tyra Biosciences; PG: none; VM: none; RS: none;
BS; received grant support from Centro de Investigación Biomédica en Red - CIBER
(Immune4Al, S2300092_3); AS: none; AV; none; TKY: Radiation Oncology Institute
Research Support; Lineberger Comprehensive Cancer Center Research Support;
Medtronic Inc.; CA: none; FT: JB: received grant support from Instituto de Salud
Carlos III (PI18/00768), the Spanish Health Ministry (National Strategic Plan against
Hepatitis C), AECC (PI044031) and WCR (AICR) 16-0026. CIBERehd is funded by
the Instituto de Salud Carlos III.
4
Authors contributions: a list of the authors' contributions to the study; concept and
design MSZ, AF, JB, MR, experiments and procedures MSZ, AF, JB, MR, JR, JF,
MB, AGC, AD, NLL, GI, MM, CA; all the authors contributed to writing the article
Data availability statement: The 2025 BCLC treatment strategy (cut-off August 5,
2025) builds on prior iterations to guide individualized decision-making. To generate
the evidence synthesis, we conducted a PubMed search restricted to humans, adults
(≥19 years), and English-language records. We performed (1) an individual-
participant-data (IPD) meta-analysis of external beam radiotherapy (EBRT) that
included approximately 5,000 patients (Moon et al.), and (2) an aggregate-data meta-
f
analysis comprising 73 studies of patients with macrovascular invasion (MVI) (Fortuny
oo
et al., submitted). We also reviewed all additional relevant information according to
5
Impact and Implications
The 2025 BCLC update provides the scientific justification for integrating recent
advances into a cohesive, evidence-based framework that remains the global
reference for prognostication and treatment allocation in HCC. By incorporating the
CUSE framework, it provides multidisciplinary teams with a structured approach to
navigate evidence gaps, explicitly address uncertainty, and tailor therapy to patient
goals and context. These updates are particularly important for clinicians, tumor
boards, and guideline developers, as well as for patients and caregivers who face
complex choices in real-world practice. In practical terms, the prognostic capacity of
f
BCLC is embedded within person-centered decision-making, fostering iterative
oo
treatment planning and laying the foundation for future studies assessing the clinical
impact of CUSE, while acknowledging the need for validation and variability in
implementation across healthcare settings.
- pr
re
lP
Summary
na
The 2025 BCLC update incorporates recent advances in the field of treatment
ur
6
patient values and goals; and (iv) select a plan with regular check-ins as new
information or needs arise.
Lay Summary
The 2025 BCLC update maintains its clear, stage-based framework and ties each
stage to the strongest evidence-based first treatment (see Figure 4). It also introduces
the concept of CUSE—Complexity, Uncertainty, Subjectivity, Emotion—to guide care
teams when direct comparisons are not possible, data are lacking, or clinical scenarios
are complex (see Figure 1a-b). Health care teams start the clinical decision by defining
f
each person’s treatment goals (e.g., longer survival, tumor control), identifying patient
oo
priorities (e.g., safety, quality of life, symptom relief), and recognizing real-world
pr
practice constraints outside clinical trials (e.g., appointments burden, travel distance,
and costs). Thereafter, the health care team, in collaboration with the patient,
-
re
assesses strengths and gaps in the data, tailor options to that person’s health and
values, and finally select a plan with regular check-ins as new information or needs
lP
arise.
na
ur
Jo
7
Introduction
Across successive updates [2–9], Barcelona Clínic Liver Cancer (BCLC) prognosis
and staging system has refined hepatocellular carcinoma (HCC) staging and
treatment allocation. We underscore those developments that justify a strategic shift
and explain why other promising—but still nascent—approaches remain premature for
inclusion. All findings are graded by study design—recognizing both the insights and
limitations of observational data—and, as before, we engage external experts to
broaden the BCLC group’s perspective. Here, we have incorporated new datasets
analyzable by BCLC stage, and we have omitted data without clear, stage-specific
f
results. The 2025 update also preserves all previous prognosis refinements—such as
oo
albumin–bilirubin (ALBI) and alpha-fetoprotein (AFP) stratification; a detailed
pr
assessment of decompensating events (such as prior variceal bleeding and ascites
grade or response); performance status (PS); and frailty, which is defined as a
-
re
multidimensional syndrome of reduced reserve and increased vulnerability. It further
provides a clearer definition of multifocal disease in intermediate stage B. Finally, we
lP
Life expectancy in HCC is modelled by three interacting forces: tumor burden, liver
function, and cancer-related symptoms. Hence, a single multivariable model to predict
outcomes across all evolutionary stages of the disease would bear excessive
complexity.[10] In addition, some parameters that would be key for some stages
would be of no value in others, as happens at the time of treatment recommendation.
Therefore, from its inception, the BCLC strategy has followed a two-step approach: (i)
delineate broad evolutionary stages with the major prognostic variables, and (ii)
develop more granular prognostic and therapeutic models within each stage. This
8
architecture was first advanced in 2000[2] and has been validated repeatedly and
adopted by most guidelines worldwide as the starting point for describing individuals
with HCC. It has also served to characterize the study population in most HCC-related
investigations, including phase III clinical trials.
Tumor burden should be assessed with contrast-enhanced multi-phase computed
tomography (CT) or magnetic resonance imaging (MRI) by liver cancer–dedicated
radiologists to avoid under- or over-staging, as even one additional nodule or a more
accurate diameter may shift stage. Chest CT is advised to complete staging when
appropriate[11,12]. Contrast-enhanced ultrasound (CEUS) is accepted as a
complementary tool for diagnosis when CT/MRI are inconclusive[11], while positron
f
emission tomography (PET) is not recommended.[13]
oo
Liver function is no longer stratified solely by the Child–Pugh (CP) score since 2018
pr
[6] because CP class A may still include patients with decompensation such as
ascites, variceal bleeding or prior hepatic encephalopathy—clear signs of severe
-
re
portal hypertension that reflect advanced liver disease. The incorporation of the ALBI
score refines mortality risk prediction across all BCLC stages.[14,15] For
lP
Cirrhosis status represents the first branching point in the framework (Figure 2). If
ur
cirrhosis, outcomes are mainly driven by the biology and extent of HCC. In
decompensated cirrhosis, residual liver function—alongside HCC extent and tumor
biology —takes precedence. Grading ascites refines the prognosis prediction as
subclinical ascites detected at imaging is not equivalent to have tense ascites.[19]
Some individuals develop HCC without cirrhosis, or after sustained virologic response
to hepatitis C virus (HCV) regardless of fibrosis regression. In these cases, prognosis
depends almost entirely on HCC biology.
Importantly, decompensation does not represent an absolute and irreversible
contraindication to specific therapies. Patients who achieve recompensation[20] after
treating the etiologic factor of the underlying liver disease or an adequate controlled
state through best supportive measures for cirrhosis complications (e.g., diuretics,
dietary interventions, or treatment of encephalopathy, variceal bleeding, or renal
9
dysfunction) may re-enter specific therapeutic pathways, as reflected in the CUSE
framework (Figure 2).
Cancer-related symptoms—captured objectively by the Eastern Cooperative
Oncology Group (ECOG-PS)—remain a robust, stage-independent predictor of overall
survival (OS) and are a common stratification factor in phase III trials of systemic
therapy.[21] By design, cancer-related symptoms (ECOG-PS) reflect cancer-
attributable symptoms rather than those due to comorbidities or underlying liver
disease. However, attribution can be challenging in decompensated cirrhosis and
even in compensated cirrhosis with marked fatigue. ECOG-PS is a useful tool to
estimate functional status, but in advanced cirrhosis or elderly patients its
f
interpretation is complex. Reduced activity or fatigue may not always be due to HCC,
oo
but rather cirrhosis-related complications (ascites, encephalopathy, sarcopenia) or
pr
frailty associated with aging and comorbidities. In this context, complementing ECOG-
PS with dedicated frailty assessment tools may better disentangle tumor-driven
-
re
symptoms from other contributors, while clinical judgment remains essential to
interpret patient-reported limitations.
lP
SUP-A&B
In practice, HCC staging starts with clinical assessment and imaging; prognosis is
refined by liver-function tests, ECOG-PS, and biomarkers. Decisions rest with a
multidisciplinary tumor board (MTB)— diagnostic and interventional radiology,
hepatology, medical and radiation oncology, surgery, pathology, nuclear medicine,
molecular biology, specialized nursing, palliative care, dietitian and social work——
with immune-related adverse-event (irAEs) experts added since immunotherapy has
been introduced.[26] This team maps stage to predict survival and selects therapy by
integrating clinical, imaging, pathology, laboratory, and psychosocial data. Leadership
within the MTB should be assumed by the professional best positioned to integrate
scientific knowledge with organizational skills, ensuring coordinated and efficient
decision-making. This approach reflects the BCLC’s emphasis on humanistic
10
management, prioritizing shared responsibility, transparent communication, and
patient-centered care.
Treatment
In the following paragraphs we outline the proposed treatment options for each BCLC
stage. After stage characterization, the 2025 BCLC again lists preferred options for
each stage. Treatment-stage migration (TSM) tempers the linear algorithm (Figure
3), permitting access to alternative treatments for earlier stages or even to liver
transplantation (LT) once listing criteria are met with recognition of the increased risk
of morbidity and/or the potential negative impact on expected survival. Untreatable
f
progression (UTP)—first defined for transarterial chemoembolization (TACE) [27] but
oo
now applied across all stages—signals failure of the current strategy and triggers
pr
transition to the treatment recommended for the next BCLC stage, even in the absence
of radiological progression. Major response after treatment——still lacking a
-
re
consensus definition—remains a major source of uncertainty in HCC decision-making,
as the necessity and efficacy of subsequent therapies in this setting are unknown.
lP
Although prognosis in BCLC is mainly determined by liver function and tumor burden,
additional individual factors may also influence outcomes. The BCLC 2025 model
na
makes these determinants explicit so that they can be systematically weighed together
ur
with stage and therapy. Importantly, the BCLC system was originally conceived as a
starting point at diagnosis or initiation of therapy. Over time, concepts such as TSM
Jo
and BCLC upon progression[28] were incorporated to capture patient trajectories. The
advent of immunotherapy now raises a new challenge: defining BCLC upon response.
This notion—commonly referred to as therapeutic conversion—recognises that
profound tumor regression marks not a return to a prior disease stage, but the
emergence of a distinct clinical state: a form of “treatable tumor regression” requiring
its own staging framework.
Clinical decision-making
The 2022 BCLC strategy[9] introduced a clinical decision framework, which the 2025
revision refines by integrating the CUSE approach. CUSE is a structured framework
applied when there is a gap between available evidence and its application to an
individual patient. It integrates clinical data, patient and treatment goals, and
11
contextual factors to organize complexity, align dilemmas with therapeutic priorities,
and apply the best available evidence. This process supports multidisciplinary teams
in reaching explicit, reproducible, and patient-centered decisions.
f
(RCT)—while retrospective studies remain more prone to bias, even with adjustments
oo
such as propensity score matching or inverse-probability of treatment weighting
(IPTW).[29–31] Treatment allocation in such studies often reflects center-specific
- pr
strategies, with surgery prioritized as first-line and ablation or locoregional therapy
reserved for patients unsuitable for resection. This structural selection bias cannot be
re
fully eliminated and is explicitly acknowledged within the CUSE frame work.[29–31]
lP
CUSE addresses two recurrent evidence gaps: (i) no demonstrated OS benefit and
ur
(ii) robust OS data that are not comparable across options. It draws on Mishel’s
Jo
Process (co-designed; Figure 1b) : The process involves (1) establishing the goals
of care; (2) assessing the clinical and social context, including hepatic reserve,
performance status, frailty, comorbidities, deliverability, affordability, and caregiver
capacity; (3) evaluating the evidence by level of certainty and applicability, translating
population data into person-level choices, and communicating options in plain
language with absolute numbers; (4) deliberating within the multidisciplinary team; (5)
12
making a shared decision documented with rationale and consent; and (6) iterating
the plan to reflect changing goals, clinical status, and evolving evidence, with interim
decisions in ambiguous scenarios (e.g., time-limited therapy, as used in downstaging)
and predefined review points.
Purpose & scope: align treatment with the patient’s goals, values, and life context
[35–38] while staying evidence-anchored and safety-first. This includes clarifying
goals of care (cure, survival, symptom relief, function), acceptance of risk/toxicity, day-
to-day realities (work, caregiving, travel, costs), and preferred involvement in
decisions.
f
oo
Dimensions include: Complexity (multifactorial comorbidities and competing
pr
treatments with equal or absent evidence); Uncertainty (individual prognostic
ambiguity, evidence gaps, rapidly evolving standards); Subjectivity (variability in how
-
re
individuals and clinicians interpret information and express preferences); and
Emotion (the psychological impact of stress, prior experiences, hopes, beliefs, or
lP
Decision thresholds (with patient agreement): GO— clear goal, robust safety
ur
When to invoke CUSE: CUSE is typically invoked when therapeutic ambiguity arises
as result of the following scenarios:
Multiple RCTs reporting improved OS — When more than one RCT demonstrates a
survival benefit, but no head-to-head comparisons are available. Important
considerations regarding the interpretation of median survival times are provided in
13
SUP-A. Exploratory methods such as restricted mean survival time (RMST), milestone
survival rates, progression-free survival (PFS), landmarks, time to treatment failure
(TTF) or duration of response (DoR), quality-adjusted survival (QALY/QALD), post-
progression survival (PPS), or the cumulative incidence of clinically relevant events
(e.g., liver failure, transplantation, variceal bleeding) may provide complementary
insights. While network meta-analyses (NMA) may provide additional insights in the
absence of head-to-head comparisons, their interpretation requires caution.
Differences in trial populations, study design, and the temporal context in which trials
were conducted may introduce heterogeneity and bias, potentially limiting the validity
of indirect comparisons. Therefore, although such analyses can support clinicians
f
when carefully performed, they should not be taken as evidence of clear superiority of
oo
one systemic option over another. In line with this principle, the BCLC update has
pr
refrained from presenting cross-trial hazard ratios or survival curve metrics, so as not
to imply a hierarchy unsupported by direct evidence.
-
re
The BCLC 2025 framework considers these approaches potentially useful for
lP
RCT but with immature OS — data due to short follow-up and few events, especially
late deaths— yet curative intent (e.g., neoadjuvant or combination therapy in early to
intermediate HCC) may be at stake. Teams must balance the statistical power of an
interim readout from a phase III RCT against the smaller yet fully mature, signal from
a phase II study. Consistent with this principle, BCLC 2025 recommends waiting for
the final phase III results and, in the meantime—while no OS benefit is proven—
basing treatment recommendation on mature phase II data (safety, response, and
survival), prospective projects, and individual-patient meta-analyses that provide
granular OS estimates in well-characterized cohorts. When event numbers are low,
stating that a therapy shows “no detrimental effect on OS” is clinically meaningless:
the absence of observed harm does not imply benefit—or even neutrality.
14
Favorable PFS in the absence of valid OS data— PFS is informative if the trigger for
it is fully defined—tumor growth, liver decompensation, death, or a composite—and
the outcome for each event is clearly described, with competing risks in people with
cirrhosis reported separately (Figure 5). Development of liver decompensation that
prevents treatment maintenance is a clinically relevant event that may result in the
‘failure of treatment strategy’ endpoint as introduced in the ABC-HCC trial comparing
systemic therapy vs chemoembolization (NCT04803994). In clinical trials, its
underlying cause should be described in detail. The clinical team should accept that a
PFS gain may never translate into an OS advantage, but it can justify a temporary
decision to attempt a potential delay of tumor progression, during which the person
f
may reach the next-line therapy —ideally one with proven curative potential or stronger
oo
survival data. The benefit, however, remains provisional and should be framed within
pr
the inherent uncertainty. PFS remains a vulnerable endpoint and is not considered a
robust surrogate for OS in major guidelines for intermediate to advanced
-
re
stages.[11,40]
lP
Absence of RCT — When RCT are lacking, interpretive flexibility depends on the type
of available evidence (e.g., phase I–II non-randomized studies, individual patient data
na
benefits and risks in situations of evidence scarcity. In this context, prospective and
retrospective evidence must be clearly distinguished when informing treatment
strategies. Prospective single-arm phase I/II trials may justify clinical adoption when
they represent the highest or most applicable evidence available, as seen with highly
selective molecular therapies in other cancers. By contrast, single retrospective
studies or aggregate meta-analyses of retrospective data are more prone to bias and
should be regarded as hypothesis-generating unless externally validated. In addition,
the growing role of real-world evidence requires careful appraisal, and its quality
should be assessed using established frameworks such as the ESMO-GROW
guidelines before informing therapeutic recommendations.[41]
15
Under the BCLC framework, the expected median OS after the first therapy is >5 years
for individuals at very-early and early stages (BCLC 0/A), more than 2.5 years for those
at the intermediate stage (BCLC B), roughly 2 years for the advanced stage (BCLC
C), and <1 year for end-stage disease (BCLC D). These estimates represent best-
case outcomes with effective first-line therapy, but individual prognosis varies widely.
When sequential treatment is deployed—moving promptly to the next, stage-
appropriate option once treatable progression emerges—survival can extend far
beyond these medians. However, candidacy for sequential treatments itself is a
prognostic marker and does not confirm survival improvement from any particular
sequencing strategy.
f
oo
First treatment option and CUSE framework per stage
BCLC0/A section.
ur
remains the recommended first step. [45–50] Because of the risk of recurrence, the
potential for Ab initio LT should always be considered within local policy[51,52] in
selected cases or reserve LT as rescue therapy if recurrence occurs. Regulations and
prioritization rules, however, differ worldwide and may prevent Ab initio LT and delay
timely LT. Indeed, for very early HCC an alternative approach is to “ablate and wait”
until recurrence takes place and at that time consider LT if still within criteria.[37,53,54]
AFP levels > 1,000 ng/mL signals an unacceptably high risk of post-transplant
recurrence.[22,23] If LT is not an option, ablation with percutaneous thermal
techniques (radiofrequency [RFA] or microwave [MW]) has historically been the
standard. Increasing evidence indicates that TACE, transarterial radioembolization
(TARE) and external-beam radiotherapy (EBRT) may achieve comparable outcomes.
Thus, so current practice considers these modalities as valid alternatives, with the final
choice largely driven by tumor features, expertise, and local resources.
16
In selected patients, minimally invasive resection (laparoscopic/robotic) may also be
considered, particularly for peripherally located tumors where ablation carries higher
risks such as tract seeding or damage to adjacent organs.[9]
f
definitive superiority can be claimed.[60,62] MW provides more reliable local control
oo
than RFA for tumors up to 3-4 cm and is therefore the preferred percutaneous
technique when feasible.[63,64]
pr
For radiation-based approaches, TARE achieves high objective responses, including
-
re
frequent complete responses in small lesions.[63,65] In the recurrent setting post
resection/RFA, EBRT shows survival and toxicity comparable to repeat RFA. [66]
lP
This is defined as a single intrahepatic HCC >2 cm, or up to three HCCs none larger
than 3 cm, without macrovascular invasion, extrahepatic spread, and ECOG PS 0 in
Jo
a patient with preserved liver function. In patients with liver decompensation, the
management principles are commented in CUSE 0/A section.
In BCLC-A, treatment selection is guided by HCC-related factors such as tumor
burden and location as well as presence of CSPH, and should also consider available
expertise and technology.
17
in steatotic liver disease.[71,72] HVPG may decrease after eradication of HCV
infection, but this has to be carefully assessed.[73]
In the absence of CSPH, resection (preferably minimally invasive) is the first option.
In patients with mildly increased CSPH, minimally invasive surgery remains an option
for single peripheral HCC with adequate remnant liver volume.
Surgery is preferred in potential LT candidates, as pathology provides key prognostic
data. The presence of microvascular invasion and satellites, predictors of
recurrence,[45–50] may support Ab initio LT.[51,52]
The macrotrabecular-massive (MTM) pattern is a morphologically distinct and
aggressive subtype of HCC but only microvascular invasion and satellite nodules are
f
validated predictors of aggressive recurrence and mortality [47] The vessels-
oo
encapsulating tumor clusters (VETC) pattern—endothelial vessels encasing tumor
pr
clusters, typically highlighted by CD34 immunostaining—is another poor-prognosis
phenotype [74] that often overlaps with MTM. Other parameters have been proposed
-
re
such as liquid biopsy[75,76] to stratify recurrence risk, but only conventional
pathological predictors are validated, and no molecular profile is yet reliable for
lP
survival. However, when LT is not an option and ablation is suboptimal (e.g., large
ur
f
oo
Patients with single HCC and severe liver dysfunction or decompensation should
pr
proceed directly to LT if they meet listing criteria (age, comorbidities, and other
relevant factors). When LT is contraindicated for non-HCC reasons, these individuals
-
re
are classified as BCLC stage D because survival is limited chiefly by liver failure.
However, some contraindications may be transient and thus, the multidisciplinary
lP
team must assess whether the exclusion factor can be corrected, allowing LT
candidacy to be reconsidered if the condition improves and the HCC has not
na
When ablation is selected for a BCLC 0/A patient, choosing the specific modality
Jo
f
define parameters for ablative EBRT[92], with <5% risk of severe toxicity.[93] A phase
oo
II trial of SBRT for single HCC ≤4 cm unsuitable for resection or RFA reported a 3-
pr
year OS of 65.4% (95% CI, 54.5–75).[86] Pooled analyses including 484 BCLC-0 and
1,123 BCLC-A patients (134 and 468 treatment-naïve, respectively) showed favorable
-
re
long-term outcomes: in naïve BCLC-0, overall median OS was not reached and by it
was 6.4 years with photons and 8.6 years with photons and protons, respectively. In
lP
treatment-naïve BCLC-A, median OS was 5.4 years and 4.3 and 4.9 years with
protons, respectively. The TRENDY trial,[85] a multicenter phase II study of patients
na
with single naive HCC ≤6 cm (median 3.4 cm), reported a median OS of 36.8 months
ur
with Drug-eluting beads (DEB)-TACE and 44.1 months with SBRT (HR 0.58; 95% CI
0.18–1.85; P = 0.36). However, most RCTs enrolled patients with recurrent HCC after
Jo
prior therapy, and several failed to meet accrual targets,[94,95] limiting the strength of
the evidence for first-line use.
These results suggest encouraging outcomes in early disease, particularly BCLC-0/A
with well-preserved liver function, although heterogeneity and selection bias remain
important limitations.[84] For these reasons, BCLC 2025 incorporates EBRT as an
ablative option in BCLC-0 patients.
Complete pathological response (CPR) is frequently observed in small solitary tumors
after selective or super-selective TACE,[96–100] but radiologic complete response
does not consistently predict histologic necrosis. [101]
A retrospective analysis of UNOS liver transplant candidates with an HCC exception
(2019–2024) reported that first-line TARE was associated with a higher rate of
complete pathologic necrosis on explant compared to TACE (~34% vs ~20%,
20
respectively) and a lower risk of waitlist dropout (IPTW-adjusted HR 0.78 [95% CI
0.68–0.89]).[102]
While higher CPR rates have been reported with certain locoregional therapies—
including TARE - SBRT achieved a 25% overall rate in a series of 88 patients used for
bridging or downstaging (30% vs. 12%, respectively).[103] Thus, CPR may serve as
a useful tool for assessing treatment efficacy, but it is not yet a definitive surrogate for
OS. Although TACE is not strictly ablative, outcomes from TRENDY and retrospective
cohorts suggest OS comparable to SBRT and TARE, and cohort data indicate
favorable long-term results in early stages, with apparent advantages in BCLC-0 over
BCLC-A as expected. [104,105]
f
While some LRTs are associated with higher rates of CPR, long-term survival
oo
differences across modalities remain modest, highlighting the importance of
pr
individualized patient selection and multidisciplinary evaluation.[106–108]
Retrospective cohort comparisons between bland embolization (TAE) and DEB-TACE
-
re
are confounded by selection and center-strategy biases. These results should be
interpreted descriptively, not as definitive head-to-head evidence.
lP
The BCLC 2025 supports the same recommendation of BCLC 2022 on the role of
ur
ablation but also consider the following data for the tumor board discussion : (a) Vitale
et al. and Yu et al. report favorable outcomes after resection in selected multifocal
Jo
21
with BCLC-A multifocal disease, no single treatment pathway can be established, as
therapeutic choice must be tailored to disease characteristics and patient profile.
While resection may appear favorable for early multifocal HCC in terms of absolute
survival, comparisons with locoregional therapies remain limited by strong selection
bias—including age, number of lesions, liver function, and portal hypertension —
despite propensity-score analyses. Moreover, Wang et al.[114] also reported that MW
ablation was associated with fewer complications and shorter hospital stay than
resection, underscoring that minimally invasive resection should not be the default
first-line option in multifocal BCLC-A but rather weighed through a CUSE evaluation
f
that balances feasibility, safety, oncologic risk, and patient priorities.
oo
Minimally invasive resection/ablation for Milan-IN disease is practiced in well-
pr
compensated patients, as reflected in the EASL Guidelines.[11] However, the
supporting evidence is limited—RCTs included very few multifocal cases (e.g., 15 per
-
re
arm in SURF,[115] 24 vs 31 in IMbrave050,[116] and <10% in Yuan et al.[112]), while
retrospective series involved highly selected Child–Pugh A patients with low tumor
lP
burden (mostly two nodules, platelet count >134 × 10³/µL, and <7% requiring major
resection).
na
Phase II data for EBRT and TARE remain small and heterogeneous. Hence, the BCLC
ur
2025 figure includes a CUSE note indicating that resection, TARE and EBRT may be
considered in selected cases, ensuring alignment with the EASL Guidelines while
Jo
f
strategy.[125–130]
oo
By contrast, in a cohort of 225 patients treated with RFA/MW at the BCLC group,
pr
suboptimal locations (subcapsular, dome, proximity to heart, hilum, gastrointestinal
tract, or major vessels) were linked to shorter time to recurrence (13.1 vs. 20.7 months)
-
re
and earlier need for treatment change (17.6 vs. 25.1 months) compared with optimal
sites.[124] Taken together, these findings suggest that while tumor location may not
lP
In the intermediate stage, tumor burden is defined as more than 3 HCC nodules, or
up to 3 nodules with at least one larger than 3 cm and ECOG PS 0. Preserved liver
function is required to be considered for locoregional therapies or downstaging
strategies. Patients with liver decompensation enter the pathway of decompensated
cirrhosis (Figure 2). They may be evaluated for LT if they meet local listing criteria.
The 2025 update leaves the BCLC-B stratification unchanged from 2022 [9] as no new
high-quality evidence warrants modification.
23
The first subgroup includes patients with well-defined HCC nodules who may be
candidates for LT if they meet the institution’s Extended LT criteria. AFP level >1,000
ng/mL signals an unacceptably high risk of post-transplant recurrence.[22,23] Where
access to LT is limited by donor shortage, living-donor grafts may mitigate this
constraint.[132,133] Such an alternative must balance donor risk against community
benefit, underscoring the need for expert, CUSE-guided judgment.
The second subgroup comprises patient’s ineligible for LT but with preserved portal
flow and defined tumor burden, allowing selective access to feeding tumor arteries;
these individuals are candidates for intra-arterial therapies. TACE remains the
standard locoregional therapy, as no head-to-head trials have proved any alternative
f
superior in terms of OS. If tumor distribution or hepatic function precludes effective
oo
TACE, the pathway moves directly to systemic therapy with extensive considerations
pr
about potential alternatives as exposed in the CUSE section.
AFP levels, subtle gradients of liver dysfunction within CP-A, and tumor heterogeneity
-
re
all modulate prognosis, yet robust universal thresholds remain elusive. Several scores
have been proposed to predict prognosis after TACE (SUP-B). However, they do not
lP
24
control.[139] In contrast, Brown et al. found no significant differences in objective
response rate (ORR), PFS, or OS between DEB-TACE and bland embolization in an
HCC population beyond BCLC-B.[140] This broader selection may help explain the
suboptimal survival. Thus, the trial does not establish equivalence between the two
options. In a cohort of 1,461 HCC patients (730 TACE, 731 TAE), the TAE group had
poorer liver function and higher HBV prevalence,[141] propensity-matched analyses
favored TACE over TAE for OS and response. In contrast, Roth et al.[142] found that
an apparent survival advantage of TACE attenuated after propensity weighting in the
overall cohort. However, among CP-A, TACE retained a survival advantage,
suggesting that benefit is concentrated in compensated patients.
f
The STOPPER trial[143] showed high short-term survival with epirubicin-loaded
oo
TACE, and TACTICS showed that outcomes with TACE alone were more favorable
pr
within the up-to-7 criteria or six to twelve[144] than outside. Collectively, these findings
indicate that the efficacy of TACE alone depends heavily on tumor burden and BCLC
-
re
stage distribution. In summary, when liver function is preserved, TACE is generally
preferred over TAE (see SUP-A). In unselected or less compensated populations,
lP
making systemic therapy the preferred option. The transition between these
ur
subgroups is best guided by the CUSE framework, balancing feasibility, risk, and
patient priorities. However, available evidence lacks granular data on systemic therapy
Jo
outcomes in this subgroup, as trial baseline characteristics have not reported such
detail; further clarification is awaited. The CUSE section offers further insight into
treatment selection for BCLC B patients.
Treatment decisions for people with intermediate HCC often shift when person-
specific factors rule out the default option shown in Fig. 4. The literature supporting
this proposal is also detailed in SUP-A.
While the MC[79] are the standard benchmark, growing evidence supports the
inclusion of certain BCLC-B patients beyond MC —particularly those successfully
downstaged and with AFP levels below a specific cutoff that varies across
jurisdictions— can achieve post-LT outcomes equivalent to those within MC.[145–147]
25
This has led to the development of extended and biomarker-integrated selection
models. In contrast, AFP levels exceeding 1,000 ng/mL consistently predict poor
survival and higher recurrence risk, and are widely considered a contraindication to
LT, regardless of tumor burden.[22,23]
Although AFP reduction following downstaging is common, the optimal magnitude and
duration of decline required for transplantation eligibility remain undefined. In this
setting, though it must be weighed carefully against donor risk and overall societal
benefit.[133,148]
In this context, what is often termed “neoadjuvant” therapy largely corresponds to
downstaging or bridging therapy, widely applied in patients beyond MC but within
f
extended criteria to prevent drop-out and refine selection. LT after major response to
oo
systemic treatment —particularly immunotherapy— is also emerging, though still
pr
investigational and supported only by limited prospective evidence. To avoid
ambiguity, we refer to these strategies as “bridging therapy” and “systemic treatment
-
re
prior to LT.” Both remain within the realm of clinical decision-making and, in the 2025
BCLC, systemic treatment prior to LT is categorized as alternative sequences that
lP
may be considered but have not been proven, underscoring persistent uncertainty
when LT indication is discussed.
na
should be prioritized for candidates achieving the largest net survival gain over
alternatives, without excluding those with substantial—though not maximal—benefit.
Jo
26
tumor burden, other locoregional options such as TARE or EBRT may be used to
delay systemic therapy and preserve it for progression.
Although retrospective studies showed higher tumor response and longer TTP with
TARE versus TACE in this population, no survival benefit difference has been
demonstrated.[156–158] TRACE, the only randomized trial, included patients with
ascites and ECOG-PS 1, which led to early mortality in the TACE arm and raising
concerns about its validity.[90] OS in the TACE arm was unexpectedly low—close to
the natural history of BCLC-B[159]—even though the study also included BCLC-A
cases, which limits the robustness and clinical interpretation of these findings.
More reliable benchmarks come from larger non-randomized cohorts: in 1,000
f
patients treated with TARE,[160] median OS for BCLC-B was 19 months overall, but
oo
38.5 months in ALBI-1 patients, underscoring the prognostic relevance of liver
embolization side). Encouraging results have also emerged for EBRT in BCLC-B.
ur
Moon et al. [84] reported favorable OS in an unselected cohort, with particularly long
survival in treatment-naïve patients and those with ALBI grade 1(4.5 years). However,
Jo
unclear selection criteria limit generalizability and hinder identification of patients most
likely to benefit. Sorin et al. likewise reported favorable multi-year survival with EBRT
in BCLC-B,[163] but these retrospective, small-cohort data are best considered
descriptive rather than comparative.
In summary, OS data on TAE are not robust enough to replace TACE in well-selected
BCLC-B patients. TARE, EBRT may be considered for patients who are not
candidates for TACE, but no data support its use as a first-line option based on OS. If
considering TARE, the DOSISPHERE-01 trial showed that personalized dosimetry
improves outcomes and should be standard practice.[164]
The current BCLC strategy still does not support TARE or SBRT as first line
alternatives to TACE in this context, as the available data on these therapies in non-
TACE candidates primarily involve patients with BCLC-A disease. These modalities
27
could serve to delay the initiation of systemic therapy and preserve it as a subsequent
option in the event of disease progression.
While outcomes are commonly stratified by liver function or prior therapy, few studies
address individual profiles (e.g., BCLC-B/C with diabetes or other comorbidities). This
gap underscores the need for granular data to guide therapy beyond OS-based
choices. CUSE highlights the importance of considering the reason for TSM (Figure
f
3), ensuring that subsequent decisions reflect evolving clinical status and patient
oo
goals. Figure 4 (dotted arrow) illustrates that sequences may occasionally bypass
pr
systemic therapy; however, this is not a general recommendation and reflects
temporary decisions in settings of uncertainty.
-
re
TACE-based combinations:
lP
Phase III trials have evaluated TACE plus systemic therapy in unresectable HCC.
na
TALENTACE[167]) improved PFS (HR 0.64–0.77) but OS data remain immature, with
stage-specific results reported only in TALENTACE. Three large RCTs in China—
Jo
28
Advanced stage (BCLC-C)
This stage includes patients with macrovascular invasion and/or extrahepatic spread,
regardless of intrahepatic tumor burden, which in Figure 4 is indicated as “any
intrahepatic burden” to denote its coexistence with macrovascular vascular invasion
or extrahepatic spread. They have preserved liver function and a maintained
performance status (ECOG PS 0–2) and should be considered for systemic therapy.
For first-line treatment, immune-based combinations—atezolizumab plus
bevacizumab[171], tremelimumab plus durvalumab[172] , ipilimumab plus nivolumab
[173], and camrelizumab plus rivoceranib (with only 17% of patients from non-Asian
f
countries)[174]—have all demonstrated superior OS compared to sorafenib (or
oo
sorafenib/lenvatinib in the case of ipilimumab plus nivolumab) across multiple
geographic regions (SUP-B).
pr
These trials included patients with unresectable HCC, mostly stage BCLC C but also
-
re
A and B in variable proportions, contributing to outcome. Other treatments such as
lenvatinib,[175] durvalumab[172] or tislelizumab[176] were non-inferior to
lP
29
TACE, EBRT, and sequential strategies were confounded by indication and immortal-
time bias, and survival data beyond 12 months were largely incomplete.[188]
These findings underscore that systemic therapy remains the standard for BCLC-C,
while TARE may represent an alternative in selected patients when systemic therapy
is contraindicated or not feasible, and highlight the need for stratified head-to-head
trials of it against immunotherapy. Because no head-to-head trials directly compare
the available immunotherapy-based combinations, current guidance is to select one
as initial systemic therapy, tailoring this choice through the CUSE framework.
f
oo
Among first-line therapies, only sorafenib[178,189] supports an evidence-based
lacking.
- pr
sequencing strategy since validated pathways after other first-line regimens are
re
Patients progressing to the second-line may benefit from regorafenib,[190,191]
cabozantinib regardless of prior sorafenib tolerance,[192] or ramucirumab if AFP >400
lP
Western trial of pembrolizumab versus placebo in the second line did not meet its
ur
30
As of August 5, 2025, SUP-B summarizes available data to guide first-line decisions.
In brief, five positives multiregional RCTs and more than 30 sub-analyses of pivotal
studies have characterized populations, updated OS, or described quality of life. Over
1,400 real-world data (RWD) publications are also available, although cohort overlap
cannot be reliably excluded. Four RCTs conducted in China have reported
encouraging results. However, OS benefits have not been demonstrated in
multiregional populations, precluding a general recommendation.
Regarding second-line therapy, RCTs are available only for those patients who
received sorafenib as first-line treatment. [190,192,193] No phase III multiregional
RCT has evaluated patients previously treated with immune-based regimens. In total,
f
6 phase II trials have analyzed outcomes with cabozantinib[197,198],
oo
regorafenib[199], regorafenib-nivolumab[200], regorafenib-pembrolizumab, [201] and
pr
lenvatinib.[202] Finally, >30 RWD studies describe outcomes in this population.See
SUP-B for RWD details by country, specific condition, and treatment line.
-
re
First-line therapy selection according to CUSE 2025
lP
Selecting first-line therapy requires balancing multiple elements: evidence from pivotal
na
trials (eligibility criteria and survival benefits);[171,173,174,203] safety data from trials
ur
31
guided discussion, ensuring that the final plan reflects both clinical judgment and the
goals and values of the person living with HCC.
A profile of special interest within BCLC-C is vascular invasion. Isolated cases are
extremely rare[188,216,217] the typical presentation is intrahepatic HCC with
concomitant macrovascular invasion. Although immunotherapy is the general
recommendation for BCLC-C, current evidence in patients with vascular invasion
comes mainly from subgroup analyses of pivotal trials[218] and observational
cohorts,[216,217] particularly Japanese series with atezolizumab–
bevacizumab.[217,219] These studies confirm the prognostic impact of liver function
and show heterogeneous survival depending on invasion extent, but cohort
f
heterogeneity and high rates of missing long-term data limit generalizability.
oo
Comparisons with TARE suggest lower 12-month survival, yet wide confidence
pr
intervals and divergent populations preclude indirect inference.[188]
-
re
The DOSISPHERE-01 phase II trial further informs this setting. Patients were
randomized (1:1) to personalized dosimetry (PDA; ≥205 Gy to the index lesion, up to
lP
(95% CI, 10.3–36.5) with PDA (n=18) versus 9.4 months (95% CI, 5.3–17.6) with SDA
ur
(n=21) (HR 0.52; 95% CI, 0.26–1.01; p=0.058). Post hoc analyses confirmed a dose–
response effect in this subgroup of patients median OS was 22.9 months (95% CI,
Jo
11.0–48.1) with tumor dose ≥205 Gy versus 10.3 months (95% CI, 5.9–17.6) with <205
Gy (HR 0.42; 95% CI, 0.22–0.81; p=0.010).[220]
There is increasing interest in EBRT in patients with vascular invasion in combination
with systemic therapy. A seminal RCT phase III study (RTOG 1112) compared
sorafenib to sorafenib with SBRT, patients were stratified by the presence and degree
of vascular invasion.[221] Of the 177 patients eligible for analysis, 74% had
macrovascular invasion and results showed that the vascular HCC response was 9%
(95% CI, 2-16) with sorafenib alone compared to 38% (95% CI, 26-49) with sorafenib
plus SBRT (P < 0.001). This data refers exclusively to vascular response and is
presented for descriptive purposes. Of note, OS and PFS remain the main endpoints
of the trial.
32
These favorable results from prospective trials suggest EBRT can be considered in
appropriate patients with macrovascular invasion and should be studied in future trials
that incorporate modern systemic therapies.
In addition to the Fortuny et al metanalysis[188] on the role of surgery in this
population, observational series consistently reflect treatment-selection bias[217,219]:
apparent advantages of resection largely disappear once disease burden, liver
function, and propensity-adjusted methods are accounted for. Overall, in the absence
of RCT evidence, survival differences across modalities must be interpreted with
caution, acknowledging residual confounding and the non-exchangeability of
treatment cohorts. Systemic therapy is the preferred first-line option for patients with
f
macrovascular invasion (Vp1–Vp4). If contraindicated or not feasible, TARE
oo
(DOSISPHERE-01)[164,220] or EBRT (RTOG 1112)[221] may be considered within
pr
the CUSE framework, however, evidence in patients with portal vein invasion remains
limited, with only 18 such cases included in DOSISPHERE-01, and these options are
-
re
therefore not yet robustly proven.
lP
33
230] candidates for LT,[133,231–235] or patients on dialysis.[236,237] Given the
limited evidence base, universal recommendations cannot be made.
In the second-line setting and beyond, clinical trial enrollment remains the preferred
option for patients with HCC. When trial participation is not feasible, treatment
selection must be guided by the CUSE framework. Robust evidence for sequential
therapy exists primarily after sorafenib.[190,192,193] Recommendations after
f
immunotherapy or lenvatinib are based on phase I/II trials, cohort studies, and expert
oo
consensus. Alternative strategies—including immunotherapy rechallenge, tyrosine
pr
kinase inhibitors, locoregional therapies (TARE, EBRT), or phase II–based
combinations—may be considered, but are not validated as standard second-line
-
re
options (see SUP-C).
Key uncertainties arise when discontinuing first-line therapy without a defined
lP
Decisions should weigh clinical status, tolerance, and patient goals (see SUP-C). In
ur
evidence.
Effective treatments can reduce tumor burden, yielding complete or partial responses
according to different criteria (RECIST 1.1[239] EASL,[240]
RECICL,[241]mRECIST,[242] Choi criteria[243], iRECIST,[244] imRECIST[245] and
LIRADS[246]). In such instances, follow-up staging may show that observed
downsizing (reduced tumor burden) translates into a downstaging that permits the
consideration of the potential benefit of treatments that have been discarded at
baseline. The downstaging via locoregional approaches (TACE and TARE) has been
developed and validated for LT.[108,147] Thereby, patients who would exceed listing
34
criteria but respond to therapy and fulfill such criteria may benefit from transplantation
and the outcome is optimal. The UNOS‑DS criteria are the most validated (single 5–
8 cm, 2–3 lesions ≤5 cm with total ≤8 cm, or 4–5 lesions ≤3 cm with total ≤8 cm, plus
AFP <1000 ng/mL).[108,247] The XXL RCT showed superior 5‑year OS with LT
versus best alternative therapy after successful downstaging (77.5% vs 31.2%;
HR,0.32; p=.035).[147] The key issues to explore are the upper tumor burden limits to
enter a downstaging protocol to reach effective transplantation and the post-treatment
criteria (Milan, UCSF) to fulfill for listing.[248] Optimal tumor/AFP thresholds, response
duration, and success definitions remain unsettled and preclude universal
recommendations.[249]
f
oo
Treatable Tumor Regression: Downsizing, Downstaging, and Conversion
Approach with Systemic Treatment
pr
Beyond locoregional strategies, systemic therapies are being investigated as tools to
-
re
achieve tumor regression and potential downstaging. Downstaging with
immunotherapy has recently been explored, and early data are suggestive.[250,251]
lP
Individuals with HCC who present with major cancer-related symptoms (ECOG PS
>2) or severe liver dysfunction and are not candidates for LT due to tumor burden or
non-oncologic contraindications, they have poor short-term survival.[2,261]
35
In selected scenarios, particularly when decompensation is potentially reversible—
such as HBV-related cirrhosis or alcohol-induced cirrhosis with sustained
abstinence—patients may achieve hepatic recompensation,[262–264] defined per
Baveno VII criteria.[44] Studies have shown that alcohol-related cirrhosis achieves
recompensation in approximately 15–20% of patients with sustained abstinence,
translating into a marked reduction in liver-related mortality.[265,266] In HBV-related
disease, antiviral therapy can induce recompensation in a substantial proportion of
patients.[267]
Recent data in HCV patients report recompensation rates ranging from 36% to 64%
f
across cohorts.[268,269] Achieving recompensation is associated with improved
oo
prognosis and, in some cases, may allow re‑evaluation for systemic therapy or liver
had HCC,[20] and for them tumor-directed therapy provides no meaningful survival
ur
36
Conclusion
The 2025 BCLC update reflects the challenge of managing HCC within an expanding
yet uneven evidence base. CUSE addresses this need by providing open-access
staging and treatment support that adapts to evolving data. This version preserves the
simplicity of BCLC staging while embedding CUSE to guide individualized care in
today’s complex therapeutic landscape. As a conceptual framework, CUSE
implementation may vary across settings with different resources. Its strength is in
making uncertainty explicit and creating a shared language, while future studies
should assess its impact on clinical practice and outcomes. To support
implementation, the open-access platform (www.bclccuse.ai) has been developed as
f
a decision-support tool, grounded in the CUSE framework and designed to
oo
standardize the decision-making process.
Abbreviations:
- pr
re
HCC: Hepatocellular Carcinoma; BCLC: Barcelona Clínic Liver Cancer; TACE:
Transarterial Chemoembolization; TARE: Transarterial Radioembolization; EBRT:
lP
for End-Stage Liver Disease; ECOG PS: Eastern Cooperative Oncology Group
ur
References:
f
[1] Iserte G, Llarch N, Moon AM, Reig M. Optimizing Liver Cancer Care Through
oo
BCLC principles. Submitted n.d.
[2]
pr
Llovet J, Brú C, Bruix J. Prognosis of Hepatocellular Carcinoma: The BCLC
Staging Classification. Semin Liver Dis 1999;19:329–38. doi:10.1055/s-2007-
-
re
1007122.
[3] Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet
lP
2003;362:1907–17. doi:10.1016/S0140-6736(03)14964-1.
[4] Forner A, Reig ME, Rodriguez de Lope C, Bruix J. Current Strategy for Staging
na
and Treatment: The BCLC Update and Future Prospects. Semin Liver Dis
ur
2010;30:061–74. doi:10.1055/s-0030-1247133.
[5] Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet
Jo
2012;379:1245–55. doi:10.1016/S0140-6736(11)61347-0.
[6] Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet 2018;391:1301–
14. doi:10.1016/S0140-6736(18)30010-2.
[7] Reig M, Darnell A, Forner A, Rimola J, Ayuso C, Bruix J. Systemic Therapy for
Hepatocellular Carcinoma: The Issue of Treatment Stage Migration and
Registration of Progression Using the BCLC-Refined RECIST. Semin Liver Dis
2014;34:444–55. doi:10.1055/s-0034-1394143.
[8] Bruix J, Reig M, Sherman M. Evidence-Based Diagnosis, Staging, and
Treatment of Patients With Hepatocellular Carcinoma. Gastroenterology
2016;150:835–53. doi:10.1053/j.gastro.2015.12.041.
[9] Reig M, Forner A, Rimola J, Ferrer-Fàbrega J, Burrel M, Garcia-Criado Á, et
al. BCLC strategy for prognosis prediction and treatment recommendation:
The 2022 update. J Hepatol 2022;76:681–93.
38
doi:10.1016/J.JHEP.2021.11.018.
[10] Orlandi F, Christensen E. A consensus conference on prognostic studies in
hepatology. J Hepatol 1999;30:171–2. doi:10.1016/S0168-8278(99)80023-3.
[11] Sangro B, Argemi J, Ronot M, Paradis V, Meyer T, Mazzaferro V, et al. EASL
Clinical Practice Guidelines on the management of hepatocellular carcinoma. J
Hepatol 2025;82:315–74. doi:10.1016/j.jhep.2024.08.028.
[12] Singal AG, Llovet JM, Yarchoan M, Mehta N, Heimbach JK, Dawson LA, et al.
AASLD practice guidance on prevention, diagnosis, and treatment of
hepatocellular carcinoma. vol. Publish Ah. 2023.
doi:10.1097/hep.0000000000000466.
f
[13] Nault J-C, Boubaya M, Wartski M, Dohan A, Pol S, Pop G, et al.
oo
[18F]fluorodeoxyglucose and [18F]fluorocholine PET-CT for staging
pr
optimisation and treatment modification in hepatocellular carcinoma (PET-
HCC01): a prospective multicentre study. Lancet Gastroenterol Hepatol
-
re
2025;10:306–14. doi:10.1016/S2468-1253(25)00011-1.
[14] Johnson PJ, Berhane S, Kagebayashi C, Satomura S, Teng M, Reeves HL, et
lP
8. doi:10.1200/JCO.2014.57.9151.
ur
[15] Pinato DJ, Sharma R, Allara E, Yen C, Arizumi T, Kubota K, et al. The ALBI
grade provides objective hepatic reserve estimation across each BCLC stage
Jo
f
al. Liver Transplantation for Hepatocellular Carcinoma: A Model Including α-
oo
Fetoprotein Improves the Performance of Milan Criteria. Gastroenterology
pr
2012;143:986-994.e3. doi:10.1053/j.gastro.2012.05.052.
[23] Mazzaferro V, Sposito C, Zhou J, Pinna AD, De Carlis L, Fan J, et al.
-
re
Metroticket 2.0 Model for Analysis of Competing Risks of Death After Liver
Transplantation for Hepatocellular Carcinoma. Gastroenterology
lP
2018;154:128–39. doi:10.1053/j.gastro.2017.09.025.
[24] Singal AG, Hoshida Y, Pinato DJ, Marrero J, Nault J-C, Paradis V, et al.
na
f
doi:10.1002/sim.8659.
oo
[32] Mishel MH. Reconceptualization of the Uncertainty in Illness Theory. Image J
pr
Nurs Scholarsh 1990;22:256–62. doi:10.1111/j.1547-5069.1990.tb00225.x.
[33] McCormick KM. A Concept Analysis of Uncertainty in Illness. J Nurs Scholarsh
-
re
2002;34:127–31. doi:10.1111/j.1547-5069.2002.00127.x.
[34] Griffiths F, Green E, Tsouroufli M. The nature of medical evidence and its
lP
[35] Beumer BR, Polak WG, de Man RA, Metselaar HJ, van Klaveren D, Labrecque
ur
f
evidence (GROW). Ann Oncol 2023;34:1097–112.
oo
doi:10.1016/j.annonc.2023.10.001.
pr
[42] Margini C, Berzigotti A. Portal vein thrombosis: The role of imaging in the
clinical setting. Dig Liver Dis 2017;49:113–20. doi:10.1016/j.dld.2016.11.013.
-
re
[43] Vilana R, Bru C, Bruix J, Castells A, Sole M, Rodes J. Fine-needle aspiration
biopsy of portal vein thrombus: value in detecting malignant thrombosis. Am J
lP
2022;76:959–74. doi:10.1016/j.jhep.2021.12.022.
[45] Adachi E, Maeda T, Matsumata T, Shirabe K, Kinukawa N, Sugimachi K, et al.
Jo
f
pathological risk of recurrence after surgical resection for hepatocellular
oo
carcinoma: An indication for salvage liver transplantation. Liver Transplant
2004;10:1294–300. doi:10.1002/lt.20202.
pr
[52] Ferrer‑Fàbrega J, Forner A, Liccioni A, Miquel R, Molina V, Navasa M, et al.
-
re
Prospective validation of ab initio liver transplantation in hepatocellular
carcinoma upon detection of risk factors for recurrence after resection.
lP
[54] Roberts JP, Venook A, Kerlan R, Yao F. Hepatocellular carcinoma: Ablate and
wait versus rapid transplantation. Liver Transplant 2010;16:925–9.
doi:10.1002/lt.22103.
[55] Takayama T, Hasegawa K, Izumi N, Kudo M, Shimada M, Yamanaka N, et al.
Surgery versus Radiofrequency Ablation for Small Hepatocellular Carcinoma:
A Randomized Controlled Trial (SURF Trial). Liver Cancer 2022;11:209–18.
doi:10.1159/000521665.
[56] Cucchetti A, Trevisani F, Pecorelli A, Erroi V, Farinati F, Ciccarese F, et al.
Estimation of lead-time bias and its impact on the outcome of surveillance for
the early diagnosis of hepatocellular carcinoma. J Hepatol 2014;61:333–41.
doi:10.1016/J.JHEP.2014.03.037.
[57] Feng K, Yan J, Li X, Xia F, Ma K, Wang S, et al. A randomized controlled trial
43
of radiofrequency ablation and surgical resection in the treatment of small
hepatocellular carcinoma. J Hepatol 2012;57:794–802.
doi:10.1016/j.jhep.2012.05.007.
[58] Cho YK, Kim JK, Kim WT, Chung JW. Hepatic resection versus radiofrequency
ablation for very early stage hepatocellular carcinoma. Hepatology
2010;51:1284–90. doi:10.1002/hep.23466.
[59] Doyle A, Gorgen A, Muaddi H, Aravinthan AD, Issachar A, Mironov O, et al.
Outcomes of radiofrequency ablation as first-line therapy for hepatocellular
carcinoma less than 3 cm in potentially transplantable patients. J Hepatol
2019;70:866–73. doi:10.1016/j.jhep.2018.12.027.
f
[60] Giuliante F, Famularo S, Grasselli S, Sangiovanni A, Vitale A, Cabibbo G, et
oo
al. Minimally invasive hepatectomy vs. thermoablation for single small (≤3 cm)
pr
hepatocellular carcinoma: A weighted real-life national comparison. JHEP
Reports 2025;7:101420. doi:10.1016/j.jhepr.2025.101420.
-
re
[61] Li Z, Pfister M, Raptis DA, Ma Y, Yang G, Li L, et al. Novel Benchmark for
Robotic Liver Resection – Bridging Tradition with Innovation. Ann Surg 2025.
lP
doi:10.1097/SLA.0000000000006759.
[62] Kabir T, Tan ZZ, Syn NL, Wu E, Lin JD, Zhao JJ, et al. Laparoscopic versus
na
f
[69] Citterio D, Facciorusso A, Sposito C, Rota R, Bhoori S, Mazzaferro V.
oo
Hierarchic Interaction of Factors Associated With Liver Decompensation After
doi:10.1001/jamasurg.2016.1121.
- pr
Resection for Hepatocellular Carcinoma. JAMA Surg 2016;151:846.
re
[70] Berzigotti A, Tsochatzis E, Boursier J, Castera L, Cazzagon N, Friedrich-Rust
M, et al. EASL Clinical Practice Guidelines on non-invasive tests for evaluation
lP
2021;74:811–8. doi:10.1016/j.jhep.2020.10.003.
[72] Baffy G, Bosch J. Overlooked subclinical portal hypertension in non-cirrhotic
NAFLD: Is it real and how to measure it? J Hepatol 2022;76:458–63.
doi:10.1016/j.jhep.2021.09.029.
[73] Lens S, Baiges A, Alvarado-Tapias E, LLop E, Martinez J, Fortea JI, et al.
Clinical outcome and hemodynamic changes following HCV eradication with
oral antiviral therapy in patients with clinically significant portal hypertension. J
Hepatol 2020;73:1415–24. doi:10.1016/j.jhep.2020.05.050.
[74] Fang J, Zhou H, Zhang C, Shang L, Zhang L, Xu J, et al. A novel vascular
pattern promotes metastasis of hepatocellular carcinoma in an epithelial–
mesenchymal transition–independent manner. Hepatology 2015;62:452–65.
doi:10.1002/hep.27760.
45
[75] Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies:
Role of tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol
2024;80:515–30. doi:10.1016/j.jhep.2023.11.030.
[76] Campani C, Zucman-Rossi J, Nault J-C. Genetics of Hepatocellular
Carcinoma: From Tumor to Circulating DNA. Cancers (Basel) 2023;15:817.
doi:10.3390/cancers15030817.
[77] Berardi G, Morise Z, Sposito C, Igarashi K, Panetta V, Simonelli I, et al.
Development of a nomogram to predict outcome after liver resection for
hepatocellular carcinoma in Child-Pugh B cirrhosis. J Hepatol 2020;72:75–84.
doi:10.1016/j.jhep.2019.08.032.
f
[78] Molina V, Sampson-Dávila J, Ferrer J, Fondevila C, Díaz del Gobbo R,
oo
Calatayud D, et al. Benefits of laparoscopic liver resection in patients with
pr
hepatocellular carcinoma and portal hypertension: a case-matched study. Surg
Endosc 2018;32:2345–54. doi:10.1007/s00464-017-5930-1.
-
re
[79] Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, et al.
Liver Transplantation for the Treatment of Small Hepatocellular Carcinomas in
lP
doi:10.1053/j.gastro.2008.02.091.
[81] Shiina S, Tateishi R, Arano T, Uchino K, Enooku K, Nakagawa H, et al.
Radiofrequency Ablation for Hepatocellular Carcinoma: 10-Year Outcome and
Prognostic Factors. Am J Gastroenterol 2012;107:569–77.
doi:10.1038/ajg.2011.425.
[82] Shiina S, Teratani T, Obi S, Sato S, Tateishi R, Fujishima T, et al. A
Randomized Controlled Trial of Radiofrequency Ablation With Ethanol Injection
for Small Hepatocellular Carcinoma. Gastroenterology 2005;129:122–30.
doi:10.1053/j.gastro.2005.04.009.
[83] Germani G, Pleguezuelo M, Gurusamy K, Meyer T, Isgrò G, Burroughs AK.
Clinical outcomes of radiofrequency ablation, percutaneous alcohol and acetic
acid injection for hepatocelullar carcinoma: A meta-analysis. J Hepatol
2010;52:380–8. doi:10.1016/j.jhep.2009.12.004.
46
[84] Moon MA, Yanagihara TK, Dawson LAJ, Il Yu J, Lawrence T, Kim TH, et al.
Overall survival among patients with hepatocellular carcinoma treated with
external beam radiation therapy: individual patient data outcomes from a
multinational cohort. ESMO 2025 2025.
[85] Méndez Romero A, van der Holt B, Willemssen FEJA, de Man RA, Heijmen
BJM, Habraken S, et al. Transarterial Chemoembolization With Drug-Eluting
Beads Versus Stereotactic Body Radiation Therapy for Hepatocellular
Carcinoma: Outcomes From a Multicenter, Randomized, Phase 2 Trial (the
TRENDY Trial). Int J Radiat Oncol 2023;117:45–52.
doi:10.1016/j.ijrobp.2023.03.064.
f
[86] Takeda A, Sanuki N, Tsurugai Y, Iwabuchi S, Matsunaga K, Ebinuma H, et al.
oo
Phase 2 study of stereotactic body radiotherapy and optional transarterial
pr
chemoembolization for solitary hepatocellular carcinoma not amenable to
resection and radiofrequency ablation. Cancer 2016;122:2041–9.
-
re
doi:10.1002/cncr.30008.
[87] Sapisochin G, Barry A, Doherty M, Fischer S, Goldaracena N, Rosales R, et
lP
[88] Salem R, Padia SA, Lam M, Chiesa C, Haste P, Sangro B, et al. Clinical,
dosimetric, and reporting considerations for Y-90 glass microspheres in
Jo
f
[94] Bush DA, Volk M, Smith JC, Reeves ME, Sanghvi S, Slater JD, et al. Proton
oo
beam radiotherapy versus transarterial chemoembolization for hepatocellular
doi:10.1002/cncr.34965.
- pr
carcinoma: Results of a randomized clinical trial. Cancer 2023;129:3554–63.
re
[95] Comito T, Loi M, Franzese C, Clerici E, Franceschini D, Badalamenti M, et al.
Stereotactic Radiotherapy after Incomplete Transarterial (Chemo-)
lP
13. doi:10.3390/curroncol29110692.
ur
f
Radiologic-Pathologic Correlation in 81 Tumors. J Vasc Interv Radiol
oo
2025;36:961–70. doi:10.1016/j.jvir.2025.02.016.
pr
[102] Kim NG, Yao FY, Kwong AJ, Mehta N. Yttrium-90 radioembolization for
hepatocellular carcinoma is associated with a lower risk of liver transplant
-
re
waitlist dropout compared to chemoembolization. J Hepatol 2025.
[103] Li Z, Yan M, Claasen MPAW, Rajendran L, Magyar CTJ, Saha S, et al. SBRT
lP
f
doi:10.1001/jamasurg.2024.1184.
oo
[110] Yu S, Jo H-S, Yu Y-D, Choi Y jin, Kim D-S. Clinical outcome of surgical
pr
resection for multifocal T2-T3 hepatocellular carcinoma up to 3 nodules: a
comparative analysis with a single nodule. J Liver Cancer 2023;23:377–88.
-
re
doi:10.17998/jlc.2023.08.24.
[111] Zhang Y-J, Chen J, Zhou Z, Hu D, Wang J, Pan Y, et al. Transarterial
lP
doi:10.1148/radiol.241096.
ur
50
[115] Kawaguchi Y, Hasegawa K, Kashiwabara K, Okamura Y, Kurosaki M, Kudo M,
et al. Surgery Versus Ablation for Hepatocellular Carcinoma: A Randomized
Controlled Trial (SURF-RCT Trial) and a Nonrandomized Prospective
Observational Trial (SURF-Cohort Trial). J Clin Oncol 2025;43:2628–38.
doi:10.1200/JCO-24-02030.
[116] Qin S, Chen M, Cheng A-L, Kaseb AO, Kudo M, Lee HC, et al. Atezolizumab
plus bevacizumab versus active surveillance in patients with resected or
ablated high-risk hepatocellular carcinoma (IMbrave050): a randomised, open-
label, multicentre, phase 3 trial. Lancet 2023;402:1835–47.
doi:10.1016/S0140-6736(23)01796-8.
f
[117] Frühling P, Nilsson A, Duraj F, Haglund U, Norén A. Single-center
oo
nonrandomized clinical trial to assess the safety and efficacy of irreversible
pr
electroporation (IRE) ablation of liver tumors in humans: Short to mid-term
results. Eur J Surg Oncol 2017;43:751–7. doi:10.1016/j.ejso.2016.12.004.
-
re
[118] Rog CJ, Alassas M, Ong E. Histotripsy—A Novel and Intriguing Technique of
Liver Ablation. JAMA Surg 2025;160:100. doi:10.1001/jamasurg.2024.2339.
lP
[119] Wehrle CJ, Burns K, Ong E, Couillard A, Parikh ND, Caoili E, et al. The first
international experience with histotripsy: a safety analysis of 230 cases. J
na
f
[126] Hori T, Nagata K, Hasuike S, Onaga M, Motoda M, Moriuchi A, et al. Risk
oo
factors for the local recurrence of hepatocellular carcinoma after a single
pr
session of percutaneous radiofrequency ablation. J Gastroenterol
2003;38:977–81. doi:10.1007/s00535-003-1181-0.
-
re
[127] Yang B, Zou J, Xia J, Ren Z, Gan Y, Wang Y, et al. Risk factors for recurrence
of small hepatocellular carcinoma after long-term follow-up of percutaneous
lP
Risk factors for local recurrence of small hepatocellular carcinoma tumors after
a single session, single application of percutaneous radiofrequency ablation.
Jo
f
[135] Lammer J, Malagari K, Vogl T, Pilleul F, Denys A, Watkinson A, et al.
oo
Prospective Randomized Study of Doxorubicin-Eluting-Bead Embolization in
pr
the Treatment of Hepatocellular Carcinoma: Results of the PRECISION V
Study. Cardiovasc Intervent Radiol 2010;33:41–52. doi:10.1007/s00270-009-
-
re
9711-7.
[136] Golfieri R, Giampalma E, Renzulli M, Cioni R, Bargellini I, Bartolozzi C, et al.
lP
64. doi:10.1038/bjc.2014.199.
ur
f
Embolization for the Treatment of Hepatocellular Carcinoma: A Propensity
oo
Score Analysis. Cancers (Basel) 2021;13:812. doi:10.3390/cancers13040812.
pr
[143] Zhu H-D, Li X, Sun J-H, Zhu X, Liu Z-Y, Li H-L, et al. Transarterial
Chemoembolization with Epirubicin-Loaded Microspheres for Hepatocellular
-
re
Carcinoma: A Prospective, Single-Arm, Multicenter, Phase 2 Study
(STOPPER Trial). Cardiovasc Intervent Radiol 2024;47:325–36.
lP
doi:10.1007/s00270-024-03666-4.
[144] Wang Q, Xia D, Bai W, Wang E, Sun J, Huang M, et al. Development of a
na
54
[148] Sapisochin G, Bruix J. Liver transplantation for hepatocellular carcinoma:
outcomes and novel surgical approaches. Nat Rev Gastroenterol Hepatol
2017;14:203–17. doi:10.1038/nrgastro.2016.193.
[149] Navasa M, Bruix J. Multifaceted Perspective of the Waiting List for Liver
Transplantation: the Value of Pharmacokinetic Models. Hepatology
2010;51:12–5. doi:10.1002/hep.23332.
[150] Lammer J, Malagari K, Vogl T, Pilleul F, Denys A, Watkinson A, 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 2010;33:41–52. doi:10.1007/s00270-009-
f
9711-7.
oo
[151] Llovet JM, Real MI, Montaña X, Planas R, Coll S, Aponte J, et al. Arterial
pr
embolisation or chemoembolisation versus symptomatic treatment in patients
with unresectable hepatocellular carcinoma: a randomised controlled trial.
-
re
Lancet 2002;359:1734–9. doi:10.1016/S0140-6736(02)08649-X.
[152] Burrel M, Reig M, Forner A, Barrufet M, Lope CR de, Tremosini S, et al.
lP
doi:10.1016/j.jhep.2012.01.008.
[153] Takayasu K, Arii S, Kudo M, Ichida T, Matsui O, Izumi N, et al. Superselective
Jo
f
[158] Carr BI, Kondragunta V, Buch SC, Branch RA. Therapeutic equivalence in
oo
survival for hepatic arterial chemoembolization and yttrium 90 microsphere
14. doi:10.1002/cncr.24884.
- pr
treatments in unresectable hepatocellular carcinoma. Cancer 2010;116:1305–
re
[159] Cabibbo G, Enea M, Attanasio M, Bruix J, Craxì A, Cammà C. A meta-analysis
of survival rates of untreated patients in randomized clinical trials of
lP
[160] Antkowiak M, Gabr A, Das A, Ali R, Kulik L, Ganger D, et al. Prognostic Role
ur
2019;11:879. doi:10.3390/cancers11060879.
[161] Ranganathan S, Gabr A, Entezari P, Gordon AC, Riaz A, Desai K, et al.
Radioembolization for Intermediate-Stage Hepatocellular Carcinoma Maintains
Liver Function and Permits Systemic Therapy at Progression. J Vasc Interv
Radiol 2023;34:968–75. doi:10.1016/j.jvir.2022.11.036.
[162] Kuehnle RA, Tchelebi LT, Ludmir EB, Escorcia FE, Shrestha S, Sanford N, et
al. Meta‑analysis of randomized controlled trials of external‑beam radiation
versus transarterial chemoembolization for hepatocellular carcinoma. Cancer
2025;131. doi:10.1002/cncr.35720.
[163] Sorin Y, Ikeda K, Kawamura Y, Fujiyama S, Kobayashi M, Hosaka T, et al.
Effectiveness of Particle Radiotherapy in Various Stages of Hepatocellular
Carcinoma: A Pilot Study. Liver Cancer 2018;7:323–34.
56
doi:10.1159/000487311.
[164] Garin E, Tselikas L, Guiu B, Chalaye J, Edeline J, de Baere T, et al.
Personalised versus standard dosimetry approach of selective internal
radiation therapy in patients with locally advanced hepatocellular carcinoma
(DOSISPHERE-01): a randomised, multicentre, open-label phase 2 trial.
Lancet Gastroenterol Hepatol 2021;6:17–29. doi:10.1016/S2468-
1253(20)30290-9.
[165] Sangro B, Kudo M, Erinjeri JP, Qin S, Ren Z, Chan SL, et al. Durvalumab with
or without bevacizumab with transarterial chemoembolisation in hepatocellular
carcinoma (EMERALD-1): a multiregional, randomised, double-blind, placebo-
f
controlled, phase 3 study. Lancet 2025;405:216–32. doi:10.1016/S0140-
oo
6736(24)02551-0.
pr
[166] Kudo M, Ren Z, Guo Y, Han G, Lin H, Zheng J, et al. Transarterial
chemoembolisation combined with lenvatinib plus pembrolizumab versus dual
-
re
placebo for unresectable, non-metastatic hepatocellular carcinoma (LEAP-
012): a multicentre, randomised, double-blind, phase 3 study. Lancet
lP
2025;405:203–15. doi:10.1016/S0140-6736(24)02575-3.
[167] Dong J, Han G, Ogasawara S, Liu R, Gu S, Liu F, et al. LBA2 TALENTACE: A
na
f
Nivolumab plus ipilimumab versus lenvatinib or sorafenib as first-line treatment
oo
for unresectable hepatocellular carcinoma (CheckMate 9DW): an open-label,
6736(25)00403-9.
- pr
randomised, phase 3 trial. Lancet 2025;405:1851–64. doi:10.1016/S0140-
re
[174] Qin S, Chan SL, Gu S, Bai Y, Ren Z, Lin X, et al. Camrelizumab plus
rivoceranib versus sorafenib as first-line therapy for unresectable
lP
6736(23)00961-3.
ur
[175] Kudo M, Finn RS, Qin S, Han K-H, Ikeda K, Piscaglia F, et al. Lenvatinib
versus sorafenib in first-line treatment of patients with unresectable
Jo
f
Arterial Infusion of Oxaliplatin, Fluorouracil, and Leucovorin vs Sorafenib Alone
oo
for Hepatocellular Carcinoma With Portal Vein Invasion. JAMA Oncol
pr
2019;5:953. doi:10.1001/jamaoncol.2019.0250.
[182] Qin S, Bi F, Gu S, Bai Y, Chen Z, Wang Z, et al. Donafenib Versus Sorafenib
-
re
in First-Line Treatment of Unresectable or Metastatic Hepatocellular
Carcinoma: A Randomized, Open-Label, Parallel-Controlled Phase II-III Trial. J
lP
[184] Shi Y, Han G, Zhou J, Shi X, Jia W, Cheng Y, et al. Toripalimab plus
bevacizumab versus sorafenib as first-line treatment for advanced
hepatocellular carcinoma (HEPATORCH): a randomised, open-label, phase 3
trial. Lancet Gastroenterol Hepatol 2025;10:658–70. doi:10.1016/S2468-
1253(25)00059-7.
[185] Vilgrain V, Pereira H, Assenat E, Guiu B, Ilonca AD, Pageaux G-P, 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 2017;18:1624–36. doi:10.1016/S1470-
2045(17)30683-6.
[186] Chow PKH, Gandhi M, Tan S-B, Khin MW, Khasbazar A, Ong J, et al.
SIRveNIB: Selective Internal Radiation Therapy Versus Sorafenib in Asia-
59
Pacific Patients With Hepatocellular Carcinoma. J Clin Oncol 2018;36:1913–
21. doi:10.1200/JCO.2017.76.0892.
[187] Ricke, J.Bulla K, Walecki J, Schott E, Sangro B, Kolligs F, Amthauer H, et al.
Safety and toxicity of the combination of Y90-radioembolization and sorafenib
in advanced HCC: an interim analysis of the European multicenter trial
SORAMIC. J Hepatol 2013;58:S114. doi:10.1016/S0168-8278(13)60270-6.
[188] Fortuny M, De Rosa A, Roca I, Ouchi D, Suárez E, Cadiz A, et al.
Hepatocellular carcinoma with macrovascular invasion: review and survival
meta-analysis with minimal prognostic criteria. Submitted n.d.
[189] Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J-F, et al.
f
Sorafenib in Advanced Hepatocellular Carcinoma. N Engl J Med
oo
2008;359:378–90. doi:10.1056/NEJMoa0708857.
pr
[190] Bruix J, Qin S, Merle P, Granito A, Huang Y-H, Bodoky G, et al. Regorafenib
for patients with hepatocellular carcinoma who progressed on sorafenib
-
re
treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase
3 trial. Lancet 2017;389:56–66. doi:10.1016/S0140-6736(16)32453-9.
lP
[191] Finn RS, Merle P, Granito A, Huang Y-H, Bodoky G, Pracht M, et al.
Outcomes of sequential treatment with sorafenib followed by regorafenib for
na
HCC: Additional analyses from the phase III RESORCE trial. J Hepatol
ur
2018;69:353–8. doi:10.1016/j.jhep.2018.04.010.
[192] Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, et
Jo
f
[198] Sanduzzi-Zamparelli M, Martinez SM, Calvo M, Varela M, Llarch N, Iserte G,
oo
et al. Interim analysis of the ACTION trial: Cabozantinib for hepatocellular
pr
carcinoma patients who discontinued first line treatment other than sorafenib
or due to sorafenib intolerance. J Hepatol 2023;78:S579–80.
-
re
doi:10.1016/S0168-8278(23)01381-8.
[199] Cheon J, Ryoo B-Y, Chon HJ, Kim H-D, Ryu M-H, Kim K-P, et al. Multicenter
lP
doi:10.1159/000543666.
[200] Sanduzzi Zamparelli M, Matilla A, Lledó JL, Martínez SM, Varela M,
Jo
f
doi:10.1002/hep.27729.
oo
[206] Ogasawara S, Chiba T, Ooka Y, Suzuki E, Kanogawa N, Saito T, et al. Post-
pr
progression survival in patients with advanced hepatocellular carcinoma
resistant to sorafenib. Invest New Drugs 2016;34:255–60. doi:10.1007/s10637-
-
re
016-0323-1.
[207] Reig M, Galle PR, Kudo M, Finn R, Llovet JM, Metti AL, et al. Pattern of
lP
[208] Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenib for
ur
f
Experience of Hepatocellular Carcinoma and Their Treatment Goals: An
oo
International Qualitative Study and Patient Journey Map. Adv Ther
pr
2024;41:3598–614. doi:10.1007/s12325-024-02939-1.
[215] Lo SH, Sharma R, Costentin CE, Aggio D, Shergill S, Colaone F, et al. Patient
-
re
Preferences for Advanced Hepatocellular Carcinoma Treatment: A
Multicountry Stated Preference Study. Futur Oncol 2021;17:4275–87.
lP
doi:10.2217/fon-2021-0697.
[216] Famularo S, Donadon M, Cipriani F, Giuliante F, Ferri S, Celsa C, et al.
na
f
P, et al. Immune checkpoint inhibitors and the liver: balancing therapeutic
oo
benefit and adverse events. Gut 2025;74:1165–77. doi:10.1136/gutjnl-2024-
332125.
pr
[224] Fortuny M, da Fonseca LG, Allaire M, Sánchez R, Nault J, Iavarone M, et al.
-
re
Breaking Barriers for Intensive Care Admission in Patients With Advanced
HCC on Immunotherapy. Liver Int 2025;45. doi:10.1111/liv.70264.
lP
f
[232] Marino R, Hassan AT, Fagenson A, Tabrizian P. Liver transplantation for
oo
hepatocellular carcinoma following immunotherapy. Curr Opin Organ
pr
Transplant 2025;30:242–50. doi:10.1097/MOT.0000000000001228.
[233] Ye Z, Song M, AMA A-K, Zhuang L, Zhang Y, Ye S, et al. Prognostic impact of
-
re
tace combined with sorafenib or lenvatinib followed by regorafenib in recurrent
hepatocellular carcinoma after liver transplantation: a retrospective study. BMC
lP
f
doi:10.1016/S0168-8278(01)00130-1.
oo
[241] Kudo M, Ueshima K, Kubo S, Sakamoto M, Tanaka M, Ikai I, et al. Response
pr
Evaluation Criteria in Cancer of the Liver (RECICL) (2015 Revised version).
Hepatol Res 2016;46:3–9. doi:10.1111/hepr.12542.
-
re
[242] Lencioni R, Llovet J. Modified RECIST (mRECIST) Assessment for
Hepatocellular Carcinoma. Semin Liver Dis 2010;30:052–60. doi:10.1055/s-
lP
0030-1247132.
[243] Choi H, Charnsangavej C, Faria SC, Macapinlac HA, Burgess MA, Patel SR,
na
66
[247] Yao FY, Ferrell L, Bass NM, Watson JJ, Bacchetti P, Venook A, et al. Liver
Transplantation for Hepatocellular Carcinoma: Expansion of the Tumor Size
Limits Does Not Adversely Impact Survival. Hepatology 2001;33:1394–403.
doi:10.1053/jhep.2001.24563.
[248] Xu E, Tabrizian P, Gutierrez J, Hoteit M, Ghaziani T, Zhou K, et al.
Downstaging of hepatocellular carcinoma before liver transplantation: Results
from a national multicenter prospective cohort study. Hepatology 2025.
doi:10.1097/HEP.0000000000001231.
[249] Tran NH, Muñoz S, Thompson S, Hallemeier CL, Bruix J. Hepatocellular
carcinoma downstaging for liver transplantation in the era of systemic
f
combined therapy with anti‑VEGF/TKI and immunotherapy. Hepatology
oo
2022;76:1203–18. doi:10.1002/hep.32613.
pr
[250] Guo Z, Liu Y, Ling Q, Xu L, Wang T, Zhu J, et al. Pretransplant use of immune
checkpoint inhibitors for hepatocellular carcinoma: A multicenter, retrospective
-
re
cohort study. Am J Transplant 2024;24:1837–56.
doi:10.1016/j.ajt.2024.04.007.
lP
[251] Tabrizian P, Holzner ML, Ajmera V, Kim AK, Zhou K, Schnickel GT, et al.
Intention-to-treat outcomes of patients with hepatocellular carcinoma receiving
na
67
[255] Chen Y, Zhang J, Hu W, Li X, Sun K, Shen Y, et al. Envafolimab plus
lenvatinib and transcatheter arterial chemoembolization for unresectable
hepatocellular carcinoma: a prospective, single-arm, phase II study. Signal
Transduct Target Ther 2024;9:280. doi:10.1038/s41392-024-01991-1.
[256] Liu D, Mu H, Liu C, Zhang W, Cui Y, Wu Q, et al. Sintilimab, bevacizumab
biosimilar, and HAIC for unresectable hepatocellular carcinoma conversion
therapy: a prospective, single-arm phase II trial. Neoplasma 2024;70:811–8.
doi:10.4149/neo_2023_230806N413.
[257] Zhang W, Tong S, Hu B, Wan T, Tang H, Zhao F, et al. Lenvatinib plus anti-
PD-1 antibodies as conversion therapy for patients with unresectable
f
intermediate-advanced hepatocellular carcinoma: a single-arm, phase II trial. J
oo
Immunother Cancer 2023;11:e007366. doi:10.1136/jitc-2023-007366.
pr
[258] GOTO Y, NIIZEKI T, FUKUTOMI S, SHIRONO T, SHIMOSE S, IWAMOTO H,
et al. Preoperative Chemotherapy Followed by Hepatectomy for Potentially
-
re
Resectable UICC7 Stage IIIA, IIIB Hepatocellular Carcinoma; A Phase II
Clinical Trial. Kurume Med J 2021;68:MS6834010.
lP
doi:10.2739/kurumemedj.MS6834010.
[259] Chiang CL, Chiu KWH, Chan KSK, Lee FAS, Li JCB, Wan CWS, et al.
na
f
[266] Hofer BS, Simbrunner B, Hartl L, Jachs M, Bauer DJM, Balcar L, et al. Alcohol
oo
Abstinence Improves Prognosis Across All Stages of Portal Hypertension in
doi:10.1016/j.cgh.2022.11.033.
- pr
Alcohol-Related Cirrhosis. Clin Gastroenterol Hepatol 2023;21:2308-2317.e7.
re
[267] Marcellin P, Gane E, Buti M, Afdhal N, Sievert W, Jacobson IM, et al.
Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for
lP
Figure legends
f
safety at every level. CUSE advocates defining choices as ‘optimal’ rather than ‘best’
oo
when evidence is lacking—avoiding false certainty—and promotes explicit acceptance
pr
of uncertainty through time-bound, reviewable decisions with clear objectives. This
ensures that each person’s unique profile guides therapy beyond trial data alone.
-
re
Figure 1b. CUSE framework process
lP
The CUSE process (co-designed) provides a structured pathway for clinical decision-
making in hepatocellular carcinoma. It involves: (1) establishing the goals of care; (2)
na
assessing the clinical and social context, including hepatic reserve, performance
ur
data into person-level choices, and communicating options in plain language using
absolute numbers; (4) deliberating within the multidisciplinary team; (5) making a
shared decision documented with rationale and consent; and (6) iterating the plan to
reflect changing goals, clinical status, and evolving evidence, incorporating interim
decisions in ambiguous scenarios (e.g., time-limited therapy used in downstaging)
and predefined review points.
f
Figure 3. Treatment-Stage Migration and CUSE-Defined Clinical Dilemmas
oo
Treatment-stage migration in HCC occurs when patients move between therapeutic
pr
categories due to factors such as comorbidities or age, feasibility constraints,
untreatable tumor progression, major treatment response, or patient decisions and
-
re
values. These shifts give rise to clinical dilemmas that the CUSE framework seeks to
define and address. Comorbidities/Age, competing health conditions or advanced age;
lP
f
general recommendation and highlights the need for temporary decisions in the face
oo
of uncertainty and limited evidence.
pr
Figure 5. Limitations of Progression-Free Survival as a Surrogate for Overall
-
re
Survival
The same PFS interval can herald very different outcomes depending on its defining
lP
event. In panel A PFS also ends with progression, but of an indolent pattern that does
not impact long-term survival (OS remains long), In panel B, PFS ends with disease
na
progression that directly drives mortality, resulting in short OS. The hatched area in
ur
72
f
oo
r
BCLC figures
-p
re
lP
VERSION 2025
na
ur
Jo
22th October
FIGURE 1A
f
oo
Patient profile • RCT phase III - end point OS
r
www.bclccuse.ai
-p
• RCT phase III - end point others
re
• RCT phase II - end point OS
lP
• RCT phase II end point others
Values/Culture
Health system
na
• Individual Data Metanalysis with OS
• Meta analysis with I2 < =50%
ur
Safety
•
Jo
Prospective cohorts with OS
• Individual Data Metanalysis without OS
• RCT phase I or Prospective cohorts without OS
• Retrospective cohorts
• Meta analysis with I2 > 50%
• Case reports, case series
• No publication
Regulatory
RCT, Randomized Clinical Trials; i2, I-squared; OS, overall surival
CUSE, Complexity, Uncertainty, Subjectivity, and Emotion factors involved in Clinical-decision making process
FIGURE 1B
Jo
ur
na
lP
re Patient profile
-p
r oo
f
BCLCCUSE.AI
Values/Culture
FIGURE 2 HCC Diagnosis
No
f
oo
BCLC 0/A-D
r
-p
Clinical Decision-Making
No Meets the LT criteria?
re
CUSE framework
(Complexity, Uncertainty, Subjectivity, and Emotion)
lP
PROGNOSIS DELINEATED
By LIVER FUNCTION
Yes
na
Treatment Stage Migration
(Comorbidities, Feasibility,
ur
Untreatable progression, Major Response,
BCLC -D
Patient Decision)
Jo
BSC Yes COMORBIDITIES that contraindicate LT?
No BCLC 0/A-B
BSC, Best Supportive Care; LT, Liver Transplantation, HCC, Hepatocellular Carcinoma
CUSE, Complexity, Uncertainty, Subjectivity, and Emotion are factors involved in Clinical-decision making process
Figure 3
f
oo
Treatment stage migration due to
r
-p
re
lP
na
Comorbidities/Age/Frailty Feasibility/Availability Untreatable progression Major Response Patient Decision/Values
ur
Jo
Clinical Dilemmas defined by CUSE approach
FIGURE 4 HCC
Based on tumor burden, liver
Prognosis function and Very early stage (0) Early stage (A) Intermediate stage (B) Advanced stage (C) End stage (D)
physical status
Single ≤ 2cm nodule Single or ≤ 3 nodules, each ≤ 3 cm >3 nodules, or up to 3 nodules, with at least 1 > 3 cm Vascular invasion and/or Extrahepatic spread Any tumor burden
Refined by AFP, ALBI score, Preserved liver function*, PS 0 Preserved liver function*, PS 0 Preserved liver function*, PS 0 Any intrahepatic burden End stage liver function, PS 3-4
Preserved liver function, PS 0-2
Child-Pugh, MELD
Potential candidate for LT Single ≤ 3 nodules, Extended Well defined nodules, Diffuse, infiltrative,
Patient characterization
f
oo
To decide individualized
treatment approach Normal Increased^ Contraindication to LT
r
-p
Yes^ No
re
lP
na
1st Treatment Option Ablation Resection Ablation Transplant TACE Systemic Treatment BSC
ur
Expected Survival > 5 years > 2.5 years > 2 years < 1 year
Jo
CUSE approach MW/RFA 1st Line Atezolizumab-Bevacizumab/Tremelimumab-Durvalumab/
www.bclccuse.ai TARE/EBRT Successful Camrelizumab-Rivoceranib/ Ipilimumab-Nivolumab
Downstaging/ If not feasible Sorafenib /Lenvatinib /Durvalumab/Tislelizumab
Clinical Decision-Making
TACE
Not feasible or failure Bridging therapy
(Complexity, Uncertainty, Regorafenib
Subjectivity, and Emotions) 2nd Line
feasible
(sorafenib-tolerant)
Not
feasible
3rd Line
Not
Cabozantinib
(Comorbidities, Age, Frailty,
Feasibility, Availability
Untreatable progression,
Major Response, Patient Decision)
Alternative sequences upon major response or failure may be considered according to CUSE framework but have not been robustly proven#
denotes temporary decisions * Except when tumor burden is within LT criteria ^ Resection for single peripheral HCC with adequate remnant volume should, when feasible, be performed minimally invasively
under uncertainty
& EBRT or resection may be considered prior to systemic treatment in BCLC-A #TACE, TARE, EBRT, resection, or LT after major response fall in this category
FIGURE 5
f
oo
NO RTP or Response
B PFS
r
-p
Mild-moderate liver dysfunction
re
Death
lP
na
ur
Symptomatic Progression C PFS
Jo
D PFS
Treatment-related death
Death