Hepatocellular carcinoma: updates on epidemiology, surveillance, diagnosis and treatment

Article information

Clin Mol Hepatol. 2025;31(Suppl):S228-S254
Publication date (electronic) : 2024 December 26
doi : https://doi.org/10.3350/cmh.2024.0824
1Department of Internal Medicine, University of Maryland Medical Center, Midtown Campus, Baltimore, Maryland, USA
2Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
3Dumont-UCLA Transplant and Liver Cancer Centers, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
4Division of Gastroenterology, Department of Medicine, University of California, San Francisco, California, USA
5Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan, USA
6Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
7Department of Medicine, Weill Medical College at Cornell University, New York, USA
8Trinity College Dublin, Dublin, Ireland
9Division of Digestive and Liver Diseases, UT Southwestern Medical Center, Dallas, Texas, USA
10Karsh Division of Gastroenterology and Hepatology, Comprehensive Transplant Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
Corresponding author : Ju Dong Yang Karsh Division of Gastroenterology and Hepatology, Comprehensive Transplant Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8900 Beverly Boulevard, Los Angeles, CA 90048, USA Tel: +1-310-423-1971, Fax: +1-310-423-2356, E-mail: judong.yang@cshs.org
Editor: Ju Hyun Shim, University of Ulsan, Korea
Received 2024 September 22; Revised 2024 November 8; Accepted 2024 December 20.

Abstract

Hepatocellular carcinoma (HCC) is a major global burden, ranking as the third leading cause of cancer-related mortality. HCC due to chronic hepatitis B virus (HBV) or C virus (HCV) infection has decreased due to universal vaccination for HBV and effective antiviral therapy for both HBV and HCV, but HCC related to metabolic dysfunction-associated steatotic liver disease and alcohol-associated liver disease is increasing. Biannual liver ultrasonography and serum α-fetoprotein are the primary surveillance tools for early HCC detection among high-risk patients (e.g., cirrhosis, chronic HBV). Alternative surveillance tools such as blood-based biomarker panels and abbreviated magnetic resonance imaging (MRI) are being investigated. Multiphasic computed tomography or MRI is the standard for HCC diagnosis, but histological confirmation should be considered, especially when inconclusive findings are seen on cross-sectional imaging. Staging and treatment decisions are complex and should be made in multidisciplinary settings, incorporating multiple factors including tumor burden, degree of liver dysfunction, patient performance status, available expertise, and patient preferences. Early-stage HCC is best treated with curative options such as resection, ablation, or transplantation. For intermediate-stage disease, locoregional therapies are primarily recommended although systemic therapies may be preferred for patients with large intrahepatic tumor burden. In advanced-stage disease, immune checkpoint inhibitor-based therapy is the preferred treatment regimen. In this review article, we discuss the recent global epidemiology, risk factors, and HCC care continuum encompassing surveillance, diagnosis, staging, and treatments.

INTRODUCTION

Liver cancer is a global challenge, remaining among the leading cause of cancer-related death, with increasing mortality rates in many countries [1]. Hepatocellular carcinoma (HCC) is the most common form of liver cancer, accounting for around 90% of the cases [2]. The change in epidemiology has important implications for the optimal HCC prevention, screening, and treatment strategies. These implications are particularly important to consider in the context of emerging strategies for HCC screening (e.g., blood-based biomarker panels and abbreviated magnetic resonance imaging [MRI] protocols) and treatment (e.g., immune checkpoint inhibitors [ICIs] combinations). This review article aims to comprehensively review the current global epidemiology and state-of-the-art evidence in the surveillance, diagnosis, and treatment of HCC.

GLOBAL INCIDENCE AND MORTALITY OF HCC

Liver cancer ranks as the sixth most commonly diagnosed cancer and the third leading cause of cancer-related deaths worldwide [1]. According to the Global Burden of Disease (GBD) Study 2021, there were over 529,000 new cases and 483,800 deaths attributed to liver cancer in 2021 [3]. In the past two decades, incidences of liver cancer increased by 53.7%, whereas death increased by 48.0% (Fig. 1A, 1B). The age-standardized incidence and death rates vary significantly by country (Fig. 1C, 1D) [4], with the highest incidence and mortality rates reported in Africa and the Western Pacific region. Liver cancer is predominantly diagnosed in middle-aged to older adults, with a median age of 62 at diagnosis; however, 14.7% of incident cases also occur in younger adults aged 15–49 years [5-7]. Despite improvements over time, the prognosis for liver cancer remains poor, with 5-year overall survival rates below 20% [2,8].

Figure 1.

(A) Incidences attributable to liver cancer from 2000 to 2021, by etiology. (B) Deaths attributable to liver cancer from 2000 to 2021, by etiology. (C) Age-standardized incidence rates attributable to liver cancer in 2021 per 100,000 population by country. (D) Agestandardized death rates attributable to liver cancer in 2021 per 100,000 population by country. (E) Proportion of incidences of liver cancer attributable to different etiologies from 2000 to 2021. (F) Proportion of deaths from liver cancer attributable to different etiologies from 2000 to 2021. ALD, alcohol-associated liver disease; HBV, hepatitis B virus; HCV, hepatitis C virus; MASH, metabolic dysfunction-associated steatohepatitis.

The incidence of HCC varies globally due to the differing distribution of these risk factors [9-12]. According to GLOBOCAN 2020, over 70% of HCC cases occur in Asia, while Africa accounts for 7.8% of the global liver cancer burden, with hepatitis B virus (HBV) being the predominant risk factor in both regions. Europe contributes nearly 10% of the global HCC incidence, North America accounts for 5.1%, and South America represents 4.4% [13,14]. Despite these regional variations, there has been a notable shift in the etiology of HCC across most regions, transitioning from viral to non-viral etiologies [15-18].

ETIOLOGY

Most patients diagnosed with HCC have underlying cirrhosis. Compared to patients without cirrhosis, those with cirrhosis face an over 30-fold increased risk of developing HCC [19]. The most common causes of liver cancer include metabolic dysfunction-associated steatotic liver disease (MASLD), alcohol-associated liver disease (ALD), chronic HBV infection, and chronic hepatitis C virus (HCV) infection, therefore making most HCC cases potentially preventable [20]. Less common causes include hereditary hemochromatosis, primary biliary cholangitis, primary sclerosing cholangitis, autoimmune hepatitis, and aflatoxin exposure [21-24]. Over the past two decades, MASLD and ALD have been the only liver cancer etiologies with rising incidence rates [3,4].

MASLD

According to GBD 2021 data, metabolic dysfunction-associated steatohepatitis (MASH)-associated liver cancer made up 8% of all liver cancer cases and 9% of liver cancer-related deaths (Fig. 1E, 1F). Due to the rising prevalence of metabolic syndrome, MASH has become the fastest-growing cause of HCC in many regions of the world [5,6,25,26], with disproportionate increases seen in low and low-middle-income countries [27,28]. MASLD is the most commonly occurring chronic liver disease, affecting one-fourth to one-third of the global population [29-32]. In addition to being a risk factor for chronic liver disease, MASLD can accelerate cardiovascular risk even in the absence of metabolic comorbidities [33,34]. Over time, hepatic steatosis causes a disruption in hepatic metabolism, increasing lipotoxicity and oxidative stress, and triggering immune system activation [35]. MASH, the inflammatory subtype of MASLD, involves both steatosis and signs of hepatocyte injury and inflammation [36]. It is currently estimated that approximately 25% of patients with MASLD progress to MASH [37-39]. Approximately 40% of MASH patients develop fibrosis, with 15% progressing to cirrhosis [35]. Interestingly, MASH-related HCC is more frequently observed in patients without cirrhosis compared to HCC from other causes [40]. Emerging data also suggest that MASH-HCC patients may have distinct characteristics compared to those with HCC from other etiologies. Patients with MASH-HCC tend to be female and older and has more metabolic comorbidities [41,42]. Additionally, patients with MASH-HCC are often diagnosed at more advanced stages compared to HCC from other causes [43,44].

Alcohol

In 2021, ALD-associated liver cancer represented 19% of both total liver cancer incidence and liver cancer-related deaths (Fig. 1E, 1F). The increase in global per-capita alcohol consumption has resulted in a surge in ALD and associated complications, particularly during the COVID-19 pandemic [45-49]. Another meta-analysis by the World Cancer Research Fund found that liver cancer risk increased by 4% for each 10 g of alcohol consumption per day [50]. A population-based study in Sweden, which utilized liver biopsy-proven ALD, found that patients with ALD had a significantly elevated risk of HCC, with a hazard ratio (HR) of 12.8 (vs. general population).51 Another systematic review and meta-analysis of over 148,000 patients determined that the cumulative incidence of HCC in patients with ALD cirrhosis at 5- and 10-year follow-ups was 3% and 9%, respectively [52]. Alcohol-associated HCC is linked to more advanced Barcelona Clinic Liver Cancer (BCLC) staging at diagnosis, reduced chances of receiving curative treatment, and worse survival outcomes [53]. Chronic alcohol consumption also increases the risk of HCC in patients with other etiologies of chronic liver diseases [54-57]. Therefore, strict abstinence from alcohol is recommended in patients with chronic liver diseases to prevent the progression of liver diseases and ultimately, HCC development.

HBV

Chronic HBV infection was responsible for 39% of total liver cancer cases and 37% of liver cancer-related deaths (Fig. 1E, 1F). Patients with chronic HBV infection are at increased risk of developing HCC [58], with risk significantly elevated by factors such as cirrhosis, high HBV DNA levels, specific genotypes, and co-infections (e.g., hepatitis D) [59-62]. Although HBV remains the leading etiology of HCC globally, expanded vaccination coverage has led to a gradual decline in HBV-related HCC [63]. A recent study revealed a nonlinear parabolic association between HBV DNA titer and HCC risk, with HBV DNA around 6 log10 IU/mL (moderate viral loads) corresponded to the highest non-invasive test scores for hepatic inflammation and fibrosis [64,65]. More recently, a large retrospective multinational study confirmed the nonlinear parabolic association between HBV DNA and HCC risk in patients with normal alanine aminotransferase (ALT) [65]. Among patients with HBV infection, antiviral therapy, particularly with nucleos(t)ide analogs, significantly reduces HCC risk by 40–60% by suppressing viral replication and reducing inflammation, contributing to the global reduction of HBV-related HCC [66-69].

HCV

Chronic HCV infection accounted for 29% of total liver cancer cases and 30% of liver cancer-related deaths (Fig. 1E, 1F). Chronic HCV infection is a well-established risk factor for HCC, with the highest risk observed in patients who have developed cirrhosis [70]. Increased implementation of HCV treatment programs using direct-acting antivirals has significantly reduced HCV-related HCC, although increased efforts are needed to meet World Health Organization (WHO) viral hepatitis elimination goals [71-74]. Further, HCC risk is significantly reduced but not eliminated after viral eradication, with studies showing persistent annual HCC incidence rates exceeding 1% more than 5 years after sustained virologic response, underscoring the importance of ongoing surveillance [75-77].

Aflatoxin

Aflatoxins are potent carcinogenic mycotoxins that commonly contaminate foods such as maize and groundnuts, exposing approximately 4.5 billion people globally [78,79]. The majority of liver cancer cases linked to aflatoxin exposure are concentrated in Sub-Saharan Africa, Southeast Asia, and China [80,81]. The primary aflatoxin involved in liver carcinogenesis is aflatoxin B1 (AFB1), produced by Aspergillus species. AFB1 leads to the formation of DNA adducts, which interact with guanine bases in hepatocyte DNA, causing a mutation in the P53 tumor suppressor gene at the codon 249 hotspot in exon 7, ultimately resulting in HCC [82,83]. The risk of HCC from aflatoxin is not evenly distributed, with a higher risk observed in children and individuals with HBV infection [80,84]. The increased risk observed with aflatoxin and HBV may be due to chronic HBV infection-inducing cytochrome P450 enzymes, which convert inactive AFB1 into the mutagenic AFB1–8,9-epoxide, thereby causing mutations in hepatocytes and ultimately hepatocarcinogenesis [85].

SURVEILLANCE

Target populations

Current guidelines recommend semi-annual HCC surveillance in at-risk populations [86-88]. Traditionally, surveillance was deemed cost-effective in cirrhosis patients with an annual HCC incidence rate of 1.5% or greater [89]. More recent cost-effectiveness analyses demonstrated an HCC incidence of more than 0.4% per year and surveillance adherence of more than 19.5% biannually for ultrasound (US) with alpha-fetoprotein (AFP) to be cost-effective in patients with compensated cirrhosis [90]. Cirrhosis patients with Child-Pugh class A or B, regardless of etiology, should undergo surveillance, while this should be restricted to transplant-eligible patients in the setting of Child-Pugh class C cirrhosis [86,91]. Non-cirrhotic, chronic hepatitis B patients are also at high risk to warrant surveillance based on their risk factors such as age, sex, endemic region, family history, HBV DNA titer, or the PAGE-B score [86]. Although 25–30% of HCC cases in patients with MASLD occur in the absence of cirrhosis, the annual risk of HCC in noncirrhotic MASLD is low. Universal HCC surveillance is not cost-effective, underscoring the need for risk stratification in this patient population [40]. Precision surveillance based on risk group using four patient factors (sex, etiology of liver disease, Child-Pugh class, and body mass index) was able to detect a higher proportion of HCC at an early stage and was more cost-effective than the universal approach, although it requires further validation before implementation in clinical practice [92].

Benefits and risks of HCC surveillance

The best data for HCC surveillance comes from a large randomized controlled trial (RCT) in patients with chronic HBV infection [93]. Although there is not a similar RCT among patients with cirrhosis, several cohort studies show associations between HCC surveillance and early-stage HCC detection, curative treatment receipt, and prolonged survival [94]. The benefits of HCC surveillance must be weighed against potential physical, psychological, and financial harms. There are fewer studies enumerating surveillance-related harms, although available data suggest these are of mild severity in most patients [94-98]. The balance of benefits vs. harms may be particularly relevant when expanding surveillance to patients with advanced fibrosis but no cirrhosis, in whom the annual incidence of HCC is lower, and the number needed to screen will inevitably become larger, further escalating the extent of unnecessary harm [99,100].

Implementation and disparity of HCC surveillance

Despite the advancement in surveillance modalities for early HCC detection, HCC surveillance in clinical practice is underused. Among 1,014 patients with cirrhosis with HCC, only 37.2% of the patients had regular outpatient care the year before HCC presentation, and only 24.7% of those under regular outpatient care were under surveillance. Nearly half lacked surveillance orders despite knowing the patient had cirrhosis [101]. Underuse of HCC surveillance is related to a combination of patient and provider-reported barriers [102,103]. Common patient-reported barriers include cost, difficulty scheduling, uncertainty of where to receive an ultrasound exam, or difficulty with transportation [102]. Underuse of surveillance is particularly high in patients with non-viral liver diseases, including those with ALD or MASLD, related to a combination of difficulty recognizing at-risk patients as well as increased social and financial barriers [53,104]. HCC surveillance is also underused in racial and ethnic minorities, populations disproportionately impacted by HCC mortality [105]. Several interventions are efficacious in increasing surveillance, but studies are needed to see their effectiveness when implemented in routine clinical practice [106].

Modality of surveillance

Biannual surveillance for HCC using abdominal ultrasound with/without AFP is recommended by multiple professional society guidelines with few notable similarities and differences (Table 1) [86-88,107]. Biannual ultrasound has been the standard imaging of choice due to its cost-effectiveness, low risk of adverse events, and detection of other complications of cirrhosis [87]. However, the sensitivity of ultrasound for early-stage HCC is operator-dependent as well as varies widely based on the patient’s body habitus, liver disease etiology, and presence of cirrhosis (Table 2) [108-110]. Therefore, there are concerns that ultrasound performance will worsen over time given the shift from viral-related liver disease to increasing proportions due to MASLD and ALD. Although ultrasound in combination with AFP increases sensitivity for early-stage HCC detection, this strategy still misses over one-third of HCC at an early stage [90].

Summary of surveillance tools recommended for hepatocellular carcinoma based on international society guidelines

Summary of the accuracy of surveillance tools for hepatocellular carcinoma

There are multiple emerging blood- and imaging-based strategies for HCC surveillance (Table 2). AFP-L3, which measures a subfraction of the AFP, and des gamma carboxy prothrombin (DCP), also known as protein induced by vitamin K absence/antagonist-II (PIVKA-II) are two serum protein biomarkers extensively studied and currently approved by the Food and Drug Administration (FDA) for risk stratification [111]. Although each biomarker has insufficient sensitivity for early-stage when used alone, biomarker panels including multiple biomarkers appear to have higher accuracy. The original hepatocellular carcinoma early detection screening (HES) score combining AFP with age, ALT, and platelet count has shown to be superior to AFP alone for detection of early HCC, and a more recent version of HES V2.0 including AFP-L3 and des-gamma-carboxy prothrombin was proposed with excellent performance [112,113]. The GALAD (gender, age, AFP-L3, AFP, des-gamma-carboxy prothrombin) model combined the three biomarkers and was recently evaluated in a national phase 3 biomarker study demonstrating an increase in accuracy: GALAD had the highest true positive rate (63.6%, 73.8%, and 71.4% for all HCC, and 53.8%, 63.3%, and 61.8% for early HCC within 6, 12, and 24 months, respectively) [111,114,115]. Another phase 3 study revealed that longitudinal GALAD further improved the sensitivity up to 66.7% (vs. 54.8% for single point GALAD) [116]. Given these promising results, GALAD is now undergoing prospective validation compared to ultrasound in a large RCT in the US enrolling over 5,000 patients with cirrhosis (NCT06084234).

Liquid biopsy is also emerging as a supplementary approach to imaging for the early detection of HCC, which consists of analysis of circulating tumor DNA, tumor cells, or tumor-derived extracellular vesicles (EV) [117]. Methylation profiling of circulating tumor DNA has evolved as a promising surveillance tool for detecting early HCC in at-risk populations, although phase 3 biomarker validation studies are yet to be done [117,118]. A recent phase 2 study on a six-marker methylated DNA marker panel (HOXA1, EMX1, AK055957, ECE1, PFKP, CLEC11A) yielded an area under the curve (AUC) of 0.96 with a sensitivity of 95% and specificity of 92% [119]. An algorithm incorporating sex and AFP levels with the methylation biomarkers (HOXA1, TSPYL5, and B3GALT6) was also developed, showing a higher early-stage sensitivity compared to AFP level (≥20 ng/mL) and GALAD (≥–0.63), although specificity was lower [118]. In addition, the HCC EV surface protein assay based on four HCC-associated surface protein markers and two EV markers is also a promising circulating biomarker for early-stage HCC detection and was validated in a phase 2 study [120]. These strategies are promising, although most data are limited to case-control studies, which can overestimate performance, and prospective validation in cohort studies is needed prior to adoption.

An abbreviated MRI has been a suggested alternative imaging modality to improve sensitivity since a complete MRI is not feasible as a surveillance tool due to prolonged scanning time. A pooled estimate of 15 studies demonstrated 86% sensitivity and 94% specificity per patient, comparable in sensitivity and specificity between non-contrast and contrast-enhanced MRI [121]. Abbreviated MRI is undergoing prospective validation in large trials in the US, France, and South Korea (NCT05486572, NCT05095714, NCT06312826) [122]. Although these studies show high test performance, issues including radiological capacity, costs, and patient acceptance would need to be considered for implementation.

Follow-up after surveillance

Follow-up of surveillance test results for subsequent procedures or diagnostic testing is recommended based on the LI-RADS (Liver Imaging Reporting and Data System) visualization score, size of the lesion, and AFP levels [86]. Patients with well-visualized sonography (visualization A) with no lesions and normal AFP levels can repeat surveillance in 6 months.

For patients with subcentimeter liver lesions in ultrasound, a short-interval repeat test in 3–4 months is reasonable when the initial ultrasound demonstrated optimal visualization [123]. However, patients with suboptimal ultrasound visualization or increased odds of poor visualization, such as obesity, alcohol-related liver disease, or MASLD cirrhosis, should be considered for further computed tomography (CT) or MRI [124]. For patients with lesions that are more than 1 cm on the ultrasound, AFP levels over 20 ng/mL, or increasing AFP levels, diagnostic contrast-enhanced multiphase MRI or CT should be performed.

CT/MRI LI‐RADS is used to further characterize abnormal imaging findings [125]. For LR‐1 (definitely benign) and LR‐2 (probably benign) lesions, return to ultrasound surveillance at a routine 6‐month interval is recommended for LR‐1 observations, while follow‐up CT or MRI in 6 months or less may be considered for LR‐2 observations. For LR‐3 (intermediate probability of HCC) lesions, close monitoring with follow‐up CT or MRI in 3 to 6 months is warranted because approximately 30% to 40% of LR‐3 observations may represent HCC. For LR-4 (probably HCC) lesions, multidisciplinary discussion for tailored workup is recommended with options including a biopsy or repeated imaging in a short interval of around 3 months. LR‐5 is diagnostic for HCC, as discussed further in the following section.

DIAGNOSIS OF HCC

HCC, unlike most cancers, can be diagnosed in at-risk patients such as patients with cirrhosis or chronic HBV infection using specific non-invasive imaging criteria, without the need for histologic confirmation [86]. This unique feature is due to the specific pattern of angiogenesis during hepatocarcinogenesis in which HCC is dominantly supplied by the hepatic artery whereas the liver parenchyma is supplied by both the portal vein (75%) and hepatic artery (25%) [126,127]. The LI-RADS categorizes liver nodules based on 1) the presence of arterial phase hyperenhancement, 2) the size of the nodule, and 3) the number of additional major features including enhancing “capsule”, non-peripheral “washout”, threshold growth and LR-5, is diagnostic for HCC [128]. An important caveat of the LI-RADS criteria is that it is only applicable in at-risk populations (i.e., cirrhosis, chronic hepatitis B, history of HCC), and liver biopsy is still required otherwise for histologic confirmation [128].

Pathological diagnosis is required for indeterminate focal lesions, which have atypical imaging features, and for patients without cirrhosis or HBV infection, in whom the LI-RADS criteria do not apply [86,87]. Routine biopsy could be considered before initiation of systemic therapy, particularly considering emerging data suggesting that up to 10% of patients with larger tumor burden can have other diagnoses (e.g., mixed HCC-CCA, CCA, or neuroendocrine), which would change the choice of systemic therapy [129]. Pathological diagnosis of HCC should be based on the criteria of the WHO classification and International Consensus recommendations using the required histological and immunohistological analyses [87].

Tissue and/or ctDNA profiling using the next generation sequencing (NGS) method is a novel oncological diagnostic with identified common mutations such as TERT, TP53, CTNNB1, and AXIN1 [130]. With the emergence of various systemic therapies, NGS has the potential to establish a foundation for making decisions regarding personalized therapy. HCC has demonstrated significant intertumoral genomic heterogeneity, with different patterns of HCC evolution and high molecular heterogeneity in multiple regions of the same tumor nodule. Thus, a liquid biopsy might have a role in the comprehensive monitoring of evolution of the molecular landscape of HCC [131].

TREATMENT

Staging is the first step to determining the treatment plan for HCC but remains challenging as it should incorporate not only tumor burden, but the underlying liver dysfunction, other medical comorbidities, performance status, and ultimately reflecting prognosis. The BCLC staging system is the most widely validated and incorporated into most society guidelines. The BCLC staging system takes into account the tumor burden (including the number and size of nodules, vascular invasion, extrahepatic spread), liver dysfunction, and performance status and is classified into 5 stages; very early stage (0), early stage (A), intermediate stage (B), advanced stage (C), and terminal stage (D) [132]. However, there is a recognition of heterogeneity within BCLC stages, which can impact recommended treatment strategies, so alternative staging systems have been proposed [133,134].

There has been a rapid revolution in HCC treatment over the past decade (Fig. 2). Patients with very early-stage and early-stage tumors are potential candidates for curative treatment, including liver transplantation (LT), local ablation, or surgical resection. Intermediate-stage tumors, i.e., localized to the liver but beyond early stage, are often considered for locoregional therapies such as transarterial chemoembolization (TACE), transarterial radioembolization (TARE) with yttrium-90 (Y90), although systemic therapy is considered for some patients with large tumor burden. The mainstay of treatment for patients with advanced-stage tumors is systemic therapy, although some patients with locally advanced tumors (e.g., branch tumor thrombus) can be treated with surgical treatment or local therapies such as TARE [132]. Patients often transition from one treatment to the other with disease progression or response over time [135].

Figure 2.

Recent advances in the management of hepatocellular carcinoma. Created in BioRender. Hwang S (2024) https://BioRender. com/u75r972. EBRT, external beam radiation therapy; HCC, hepatocellular carcinoma; IRE, irreversible electroporatio; MWA, microwave ablation; PBT, proton beam therapy; RFA, radiofrequency ablation; SBRT, stereotactic body radiation therapy; TACE, transarterial chemoembolization; TARE, transarterial radioembolization.

A multidisciplinary approach involving the collaboration of healthcare professionals from various fields is recommended for all HCC patients due to the complexity of HCC staging and management, as well as rapidly emerging treatment options. This has been associated with improved overall survival and an increased chance of patients receiving curative treatment [136]. The role of the multidisciplinary team is to confirm the diagnosis and staging of HCC and formulate a treatment plan such as identifying candidates for surgical resection for early-stage HCC, as well as promptly identifying those eligible for liver transplant [137]. Multidisciplinary care is important not only at the initial presentation but also throughout a patient’s treatment course.

Surgical resection

Patients with localized HCC without evidence of vascular invasion or extrahepatic metastases are suitable candidates for liver resection. The underlying liver function and presence of portal hypertension (defined as HVPG ≤10 mmHg) play a major role in determining resectability [138]. Although most data are limited to BCLC 0 or A stages, studies on extending resection criteria to select patients with limited multifocal HCC or those with locally advanced HCC have suggested survival benefits compared to TACE or radiofrequency ablation (RFA). It suggests that, for early multinodular HCC patients ineligible for transplant, resection should be prioritized [139-145]. Most of the data for these extended indications are derived from Asian cohorts, with fewer data from Western cohorts in non-HBV populations. Minimally invasive liver resection including laparoscopic and robotic approaches is applied more to reduce morbidity and mortality [146]. Recurrence occurs in more than half of the patients undergoing surgical resection, mostly within 2 years. Nevertheless, surgical resection can achieve long-term survival by detecting and treating recurrent tumor early [147-149].

Local ablation

Local ablation is highly effective for tumors up to 3 cm in diameter. RFA and microwave ablation (MWA) are commonly used ablation treatments, although other ablative modalities are available with similar outcomes [150]. RFA induces radiofrequency-generated heat to destroy tumoral tissue and its efficacy has been demonstrated in early HCC patients [151]. MWA uses high-frequency electromagnetic energy to induce tissue death with large tumor ablation volumes, faster ablation time, and lower risk of heat-sink effect [152,153]. Although the local recurrence rate and disease-free survival rate were similar between RFA versus MWA, MWA had a lower distant recurrence rate, potentially decreasing the rate of long-term recurrences [154]. Both are a valuable option for patients who are not a candidate for surgery due to tumor size, number, central location, impaired liver function, or comorbidities and are a cost-effective option compared to surgery [155,156]. Depending on the location of the tumor, RFA can be adopted as first-line due to excellent local control of small HCC [87,157]. It has also been proven effective as downstaging and bridging therapy in the pre-transplant setting [158]. However, its efficacy diminishes in patients with large lesions (more than 3 cm) or lesions adjacent to large vessels [86]. Alternative treatment should be considered in patients with HCC adjacent to other critical structures, including central bile ducts, or if the area is not accessible via a percutaneous method. Similar to surgical resection, the high rate of tumor recurrence and/or distant metastasis remains a challenge [159]. Furthermore, there are alternative ablative modalities such as cryoablation which demonstrated lower local tumor progression and equivalent efficacy and safety compared to RFA in a multicenter RCT [160], or irreversible electroporation which utilizes short pulses of direct current at high voltage through multiple electrodes and recently demonstrated efficacy in a clinical setting on HCC patients [161].

Neoadjuvant and adjuvant treatment

Systemic therapies in the adjuvant and neoadjuvant settings for early-stage HCC have been investigated recently. In a phase 3 trial of adjuvant treatment with sorafenib after HCC surgical resection or local ablation, there was no benefit compared to the placebo in recurrence-free survival [162]. The IMbrave 050 study was the first phase 3 study to report positive results. There was a significantly improved recurrence-free survival (HR 0.72, 95% confidence interval [CI] 0.53–0.98) in the adjuvant therapy group (atezolizumab plus bevacizumab) vs. control group of high-risk HCC patients after curative-intent surgical resection or ablation [163]. However, more recently, longer follow-up results suggested a lack of improvement in progression-free survival [164]. Neoadjuvant therapies are also promising in priming the immune system to prevent recurrence and downstaging of locally advanced tumors for surgical resection or liver transplants [165]. HCC develops in a setting of chronic inflammation of the liver in which the microenvironment is altered and immune cells are altered; thus, several synergistic mechanisms for the efficacy of neoadjuvant immunotherapy have been proposed [165]. Neoadjuvant cabozantinib and nivolumab converted locally advanced HCC into resectable disease (80% with margin negative resection, 42% with major pathologic response) in a total of 15 patients [166]. A phase 2 study on perioperative nivolumab and nivolumab plus ipilimumab was safe and tolerable. 33% of the patients achieved major pathological response (MPR) and 5/6 MPR patients achieved complete response [167]. Multiple phase 1–2 clinical trials are ongoing to investigate the role of neoadjuvant immunotherapy for HCC (NCT05908786, NCT03299946, NCT05471674, NCT04224480, NCT03337841, NCT05440864, NCT03682276, NCT03510871, NCT04658147, NCT04123379).

LT

LT is an optimal treatment option for early-stage, unresectable HCC in patients with cirrhosis as it can not only remove intrahepatic tumors but also cure the underlying cirrhosis. About 15% of patients experience HCC recurrence after LT with a mortality rate of 19.8% at 5 years and 35.7% at 10 years in patients receiving deceased donor LT (DDLT). There has been a significant improvement in transplant outcomes over the past few decades [168,169]. The selection of patients for LT has been historically guided by the Milan criteria (one tumor that is less than 5 cm in diameter; up to three tumors, each less than 3 cm in diameter) while there were ongoing efforts on extending the patient population eligible for LT [170]. Additional criteria have also been proposed such as the UCSF criteria where the overall size limitation has expanded (one tumor ≤6.5 cm in diameter or up to three tumors ≤4.5 cm with total tumor diameter ≤8 cm) with no significant difference in 5-year follow-up [171-173]. Successful downstaging, defined as a reduction of viable tumor burden by locoregional therapy within the Milan criteria, demonstrates good post-LT outcomes and is currently adopted in the United Network for Organ Sharing (UNOS) in the U.S [174,175]. Over 80% of the cases had a successful down-staging rate with HCC within the UNOS-DS criteria (a single lesion greater than 5 cm and up to 8 cm; two to three lesions, with at least one lesion less than 3 cm and each 5 cm, with a total tumor diameter of 8 cm; four to five lesions, each less than 3 cm, with a total tumor diameter of 8 cm) [176]. The outcome of “all-comers,” HCC exceeding UNOS-DS criteria in regard to primary tumor burden, had a slightly lower successful downstaging but acceptable rate (67%), suggesting that it is a realistic goal in selected all-comer patients [177].

Salvage LT is also a strategy for patients with HCC who have undergone resection and developed tumor recurrence within the LT criteria or liver decompensation [178,179]. It has shown better disease-free survival compared to second resection for recurrent HCC [180]. A major limitation of LT has been organ shortage, and living donor LT (LDLT) has been a viable option, especially in Asia [181]. LDLT recipients showed comparable long-term outcomes compared to DDLT recipients. LDLT may provide better survival benefits, especially in regions that had low deceased organ availability [182].

TACE and TARE

TACE is the standard of care treatment for intermediate-stage HCC with well-defined nodules, preserved portal flow, and selective access [86,183]. It is a method of injecting chemotherapy into tumor-feeding arteries while hindering the arterial blood supply to the tumor, inducing tumor necrosis [184]. Two RCTs confirmed the survival benefit of TACE for unresectable HCC [185,186]. It is often used as a bridging or downstaging therapy for the patient to be a candidate for LT or resection [184]. However, challenges remain as patients may develop TACE-refractory tumors or experience failure to achieve objective response after two or more treatments [187]. Furthermore, transarterial embolization using microspheres alone when compared to TACE demonstrated no difference in RECIST response in a randomized trial in a single tertiary center, challenging the role of chemotherapy [188].

TARE delivers radioactive microspheres loaded with Y90 leading to tumor destruction with excellent efficacy in solitary HCC [189]. A phase 2 study randomized BCLC stage B or C, Child-Pugh stage A patients with unablatable/unresectable HCC to either Y90 therapy or TACE and was able to demonstrate that the Y90 radioembolization group had a significantly longer median time to progression (TTP) (>26 versus 6.8 months) with better tumor control and increased likelihood to receive transplantation and slightly favorable side effect profile [190]. The superiority of TARE in terms of TTP was insufficient to prolong the overall survival compared to TACE [191]. However, prolonging TTP through TARE can provide higher rates of successful bridging to transplantation, especially as Y90 is an intra-arterial therapy that eliminates the risk of tract seeding. TARE is better tolerated than TACE [190]. The TRACE phase 2 RCT compared Y90 glass TARE with doxorubicin drug-eluting bead TACE and demonstrated superiority in tumor control and overall survival [192]. In addition, a phase 2 trial (DOSISPHERE-01) reported a personalized dosimetry approach delivering at least 205 Gy to the index lesion improved overall survival, especially in patients downstaged for resection compared to the prior standardized approach, highlighting the importance of using personalized dosimetry [193].

External beam radiation treatment

Stereotactic body radiation therapy (SBRT) is an emerging treatment option across early, intermediate and advanced stages of HCC [194]. It is a modality that delivers high doses of conformal radiation to the tumors in 1–5 fractions, enabling a precise steep dose gradient without involving the surrounding structures including the liver [194-196]. It is a promising alternative to RFA or TACE in terms of local control but also demonstrated superior outcomes when combined with TACE plus RT compared to TACE alone [197,198]. Patients with advanced HCC and macroscopic vascular invasion may also benefit from first-line treatment with TACE plus RT vs sorafenib, although superiority of this combination approach to systemic treatment needs to be revisited given the improved efficacy of systemic treatment with the introduction of ICI [199].

Proton beam therapy is also an emerging option that is non-invasive and used in locations that are unsuitable for other local treatments such as RFA or surgery [200]. In a phase 3 clinical trial on recurrent/residual HCC (size <3 cm, number ≤2), patients were randomized to proton beam and RFA. Proton beam therapy was non-inferior to RFA with 2-year local progression-free survival being 92.8% vs. 83.2% [201]. Of note, crossover was permitted and PBT demonstrated greater feasibility than RFA, with a notably lower crossover rate (8.3 versus 26.4%) [201]. Overall, external beam radiation treatment is a viable option for early-to-intermediate stage HCC in achieving local control and is promising in combination therapies with TACE in intermediate-stage, and combination with systemic therapy in advanced HCC, especially with vascular invasion [199,200].

Systemic treatment

Systemic treatment is indicated for unresectable advanced-stage HCC, intermediate-stage HCC with extensive, infiltrative liver involvement, and those who had disease progression despite locoregional therapy or are poor candidates for locoregional treatment [86]. Tyrosine kinase inhibitors (TKI) and ICI with or without anti-vascular endothelial growth factor (VEGF) inhibitors have become the mainstay of systemic treatment for HCC (Table 3) [163,202-213]. When deciding on the appropriate systemic therapy, several factors need to be considered. For example, history of autoimmune disease or prior adverse events needs to be considered when starting an ICI, whereas a history of cardiac comorbidities or risk of bleeding/thrombosis should be accounted for before initiation of an anti-angiogenic treatment [214]. Screening for esophageal varices is recommended before initiating therapy with bevacizumab due to the potential adverse effects of gastrointestinal bleeding associated with the treatment [215].

Summary of the efficacy of systemic treatment with proven benefit in phase 3 RCTs

First-line treatment

Atezolizumab (anti-PDL1) plus bevacizumab, an antiangiogenic agent, has become the standard of care and the first-line regimen for patients with advanced HCC after the IMbrave 150 clinical trial in 2020 [202,215]. This has also been validated by several studies regarding its efficacy and safety profile/tolerance in Child-Pugh class B patients, although the presence of main portal vein tumor thrombus and a higher albumin-bilirubin (ALBI) grade was associated with a poorer prognosis [216,217]. The combination treatment of durvalumab and tremelimumab is also currently approved as the first-line regimen for patients with advanced HCC [214]. A phase 3 trial demonstrated that STRIDE (Single Tremelimumab Regular Interval Durvalumab) significantly improved overall survival (HR 0.78, 95% CI 0.65–0.93) compared to sorafenib, and durvalumab monotherapy was non-inferior to sorafenib for patients with unresectable HCC [203]. The main factor to consider in selecting first-line combination regimens between the two is whether the patient can tolerate an anti-VEGF inhibitor [218]. While nivolumab plus ipilimumab was tested before as second-line (CheckMate-040), a recent phase 3 study (CheckMate-9DW) on first-line nivolumab plus ipilimumab compared to sorafenib or lenvatinib reported improved median overall survival (23.7 versus 20.6 months, HR 0.79, 95% CI 0.65–0.96) suggesting that this combination ICI therapy will likely be approved as a first-line regimen for unresectable HCC soon [204,205].

Sorafenib is the first TKI that showed improved survival and TTP in untreated advanced HCC with Child-Pugh class A [206]. It is also the treatment of choice for advanced HCC recurrence after LT, although drug-to-drug interactions with immunosuppressants should be closely monitored [219]. Lenvatinib, another TKI, has subsequently demonstrated non-inferiority (vs. sorafenib) in overall survival in the REFLECT trial [207].

Subsequent line treatment

Regorafenib was shown to improve overall survival in BCLC stage B or C HCC patients not eligible for local treatment who previously progressed on sorafenib in the RESOURCE study [208] and cabozantinib demonstrated improved survival in HCC not amenable to curative treatment with up to 2 lines of prior treatment including sorafenib [209]. Ramucirumab, a monoclonal antibody against VEGF2, was studied in BCLC stage B or C HCC patients previously treated with sorafenib and with an AFP above 400 ng/mL and showed improved median overall survival compared to placebo [210]. Pembrolizumab showed efficacy in a phase 3 trial in advanced HCC, previously treated with sorafenib compared to placebo [211]. These treatments were U.S. FDA-approved as subsequent line treatments for advanced HCC.

Combination of systemic and locoregional treatment

Combining systemic treatment with other modalities such as ablation, TACE, TARE or SBRT is expected to improve outcomes and is under active investigation in intermediate and advanced-stage HCC. For example, local ablation in combination with tremelimumab showed additional increased immunogenicity effects, leading to increased intratumoral CD8+ cells and reduction in HCV viral load [220]. The EMERALD-1 is a phase 3 trial that demonstrated improvement in PFS with the combination of ICI plus VEGF inhibitor and TACE compared to only TACE in patients with embolization-eligible unresectable HCC, Child-Pugh A to B7 liver function, ECOG 0–1, and no evidence of extrahepatic disease, though there needs to be caution as further studies need to explore overall survival and the safety profile [221]. There was also a recent study comparing the addition of a locoregional therapy to systemic therapy in advanced HCC. TACE-ICI-VEGF treatment, versus ICI-VEGF, exhibited improved median OS (22.6 months versus 15.9 months) and higher ORR (47.3% versus 29.7%) [222].

An important caveat is that these phase 3 clinical trials were performed in select populations with preserved liver function (e.g., Child-Pugh class A liver disease) and good performance (ECOG PS 0 or 1) status, limiting generalizability of the efficacy and safety of treatment in patients with more comorbid conditions [223]. ICI treatment should be considered and further studied in patients with advanced liver dysfunction given its tolerability; a meta-analysis found that Child-Pugh class B HCC patients had a comparable safety profile from ICI treatment and response rate compared to Child-Pugh class A patients but poorer prognosis in overall survival due to higher competing risk of death from liver failure [224]. Furthermore, although various combination therapies are being studied, the identification of biomarkers to predict the response to systemic therapies such as ICI or TKI is limited with few relevant studies to date [225]. Lastly, the financial burden of systemic treatment is substantial and should be better understood and taken into consideration in selecting a treatment strategy for HCC [226].

BEST SUPPORTIVE CARE (BSC)

Palliative care can be provided alongside active treatment, but it can also be chosen as the only treatment option when disease-modifying therapies are unavailable, referred to as BSC. Symptom management especially in the terminal stage includes managing complications of liver failure such as ascites and encephalopathy, pain or dyspnea from metastatic disease, and fatigue or anorexia from extrahepatic symptoms [227]. Furthermore, the palliative care team can partake in advance care planning discussions before treatment, and provide support around treatment decisions as well as end-of-life preferences or hospice [228].

CONCLUSION

The landscape of HCC care continuum has transformed dynamically in recent years. The increasing burden of MASLD has brought a challenge in the selection of the population that is eligible for HCC surveillance which subsequently affects the surveillance implementation, policy and appropriate modality for surveillance. Diagnostics are shifting towards precision medicine along with the various treatment options that have emerged. With the advance of locoregional therapy and downstaging HCC, the candidates for LT have been broadened with promising outcomes. Systemic treatment with ICIs and TKIs has become the backbone of advanced HCC with its applications broadening in the neoadjuvant and adjuvant settings. Studies combining different modalities such as locoregional therapy and systemic therapy in intermediate and advanced HCC are also being actively performed with promising results.

Notes

Authors’ contribution

Concept and design: SYH, PD, JDY. Administrative Support: JDY. Writing, original draft: SYH, PD. Writing, review, and editing: VA, NM, NDP, GKA, AGS, JDY. Final Approval and Agreement: All Authors.

Acknowledgements

Dr. Singal’s research is supported by NCI R01 MD012565 and R01 CA256977. Dr. Yang’s research is supported by NCI K08CA259534.

Conflicts of Interest

Ju Dong Yang provides a consulting service for Astra-Zeneca, Eisai, Exact Sciences, Exelixis, Fujifilm Medical Sciences. Neehar Parikh has served as a consultant or advisor for Genentech, Fujifilm Medical, Eisai, Exelixis, Merck, Exact Sciences, Freenome, and Gilead. Ghassan K. Abou-Alfa reports research support from Agenus, Arcus, Astra Zeneca, BioNtech, BMS, Elicio, Genentech/Roche, Helsinn, Parker Institute, Pertzye, Puma, QED, Servier, Yiviva and consulting support from Abbvie, Alligator Astra Zeneca, Autem, Berry Genomics, BioNtech, Boehringer Ingelheim, BMS, Eisai, Exelixis, Genentech/Roche, Incyte, Ipsen, J-Pharma, Merck, Merus, Neogene, Novartis, Servier, Tango, Tempus, Vector, Yiviva.

Amit Singal has served as a consultant or on advisory boards for Genentech, AztraZeneca, Eisai, Exelixis, Bayer, Merck, Elevar, Boston Scientific, Sirtex, Histosonics, FujiFilm Medical Sciences, Exact Sciences, Roche, Glycotest, Abbott, and GRAIL.

Abbreviations

AFB1

aflatoxin B1

AFP

alpha-fetoprotein

ALD

alcohol-associated liver disease

ALT

alanine aminotransferase

AUC

area under the curve

BCLC

Barcelona Clinic Liver Cancer

BSC

best supportive care

CI

confidence interval

CT

computed tomography

DCP

des gamma carboxy prothrombin

DDLT

deceased donor liver transplantation

EV

extracellular vesicles

FDA

Food and Drug Administration

GBD

Global Burden of Disease

GALAD

gender

HBV

hepatitis B virus

HCC

hepatocellular carcinoma

HCV

hepatitis C virus

HES

hepatocellular carcinoma early detection screening

HR

hazard ratio

ICI

immune checkpoint inhibitor

LDLT

living donor liver transplantation

LI-RADS

Liver Imaging Reporting and Data System

LT

liver transplantation

MASH

metabolic dysfunction-associated steatohepatitis

MASLD

metabolic dysfunction-associated steatotic liver disease

MPR

major pathological response

MRI

magnetic resonance imaging

MWA

microwave ablation

NGS

next generation sequencing

RCT

randomized controlled trial

RFA

radiofrequency ablation

SBRT

stereotactic body radiation therapy

TACE

transarterial chemoembolization

TARE

transarterial radioembolization

TKI

tyrosine kinase inhibitors

TTP

time to progression

UNOS

United Network for Organ Sharing

WHO

World Health Organization

Y90

yttrium-90

References

1. Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol 2022;77:1598–1606.
2. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:6. Erratum in: Nat Rev Dis Primers 2024;10:10.
3. Danpanichkul P, Suparan K, Tothanarungroj P, Dejvajara D, Rakwong K, Pang Y, et al. Epidemiology of gastrointestinal cancers: a systematic analysis from the Global Burden of Disease Study 2021. Gut 2024;74:26–34.
4. GBD 2021 Causes of Death Collaborators. Global burden of 288 causes of death and life expectancy decomposition in 204 countries and territories and 811 subnational locations, 1990-2021: a systematic analysis for the Global Burden of Disease Study 2021. Lancet 2024;403:2100–2132. Erratum in: Lancet 2024;403:1988.
5. Danpanichkul P, Aboona MB, Sukphutanan B, Kongarin S, Duangsonk K, Ng CH, et al. Incidence of liver cancer in young adults according to the Global Burden of Disease database 2019. Hepatology 2024;80:828–843.
6. Huang DQ, Singal AG, Kono Y, Tan DJH, El-Serag HB, Loomba R. Changing global epidemiology of liver cancer from 2010 to 2019: NASH is the fastest growing cause of liver cancer. Cell Metab 2022;34:969–977. e2.
7. Yang JD, Altekruse SF, Nguyen MH, Gores GJ, Roberts LR. Impact of country of birth on age at the time of diagnosis of hepatocellular carcinoma in the United States. Cancer 2017;123:81–89.
8. Huang DQ, Singal AG, Kanwal F, Lampertico P, Buti M, Sirlin CB, et al. Hepatocellular carcinoma surveillance - utilization, barriers and the impact of changing aetiology. Nat Rev Gastroenterol Hepatol 2023;20:797–809.
9. Danpanichkul P, Suparan K, Sukphutanan B, Kaeosri C, Tothanarungroj P, Sirimangklanurak S, et al. Changes in the epidemiological trends of primary liver cancer in the Asia-Pacific region. Sci Rep 2024;14:19544.
10. Ganne-Carrié N, Nahon P. Differences between hepatocellular carcinoma caused by alcohol and other aetiologies. J Hepatol 2024 Dec 20. doi: 10.1016/j.jhep.2024.12.030.
11. Muthiah MD, Sukphutanan B, Sim B, Huang DQ, Seko Y, Lee BP, et al. Socio-economic association of alcohol use disorder and cardiovascular and alcohol-associated liver disease from 2010 to 2019. Aliment Pharmacol Ther 2024;60:340–349.
12. Pimpin L, Cortez-Pinto H, Negro F, Corbould E, Lazarus JV, Webber L, et al, ; EASL HEPAHEALTH Steering Committee. Burden of liver disease in Europe: Epidemiology and analysis of risk factors to identify prevention policies. J Hepatol 2018;69:718–735.
13. Singal AG, Kanwal F, Llovet JM. Global trends in hepatocellular carcinoma epidemiology: implications for screening, prevention and therapy. Nat Rev Clin Oncol 2023;20:864–884.
14. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–249.
15. Mak LY, Liu K, Chirapongsathorn S, Yew KC, Tamaki N, Rajaram RB, et al. Liver diseases and hepatocellular carcinoma in the Asia-Pacific region: burden, trends, challenges and future directions. Nat Rev Gastroenterol Hepatol 2024;21:834–851.
16. Kondili LA, Lazarus JV, Jepsen P, Murray F, Schattenberg JM, Korenjak M, et al. Inequities in primary liver cancer in Europe: the state of play. J Hepatol 2024;80:645–660.
17. Kedar Mukthinuthalapati VVP, Sewram V, Ndlovu N, Kimani S, Abdelaziz AO, Chiao EY, et al. Hepatocellular carcinoma in sub-Saharan Africa. JCO Glob Oncol 2021;7:756–766.
18. Pinheiro PS, Jones PD, Medina H, Cranford HM, Koru-Sengul T, Bungum T, et al. Incidence of etiology-specific hepatocellular carcinoma: diverging trends and significant heterogeneity by race and ethnicity. Clin Gastroenterol Hepatol 2024;22:562–571.e8.
19. Thiele M, Gluud LL, Fialla AD, Dahl EK, Krag A. Large variations in risk of hepatocellular carcinoma and mortality in treatment naïve hepatitis B patients: systematic review with meta-analyses. PLoS One 2014;9:e107177.
20. Choi S, Kim BK, Yon DK, Lee SW, Lee HG, Chang HH, et al. Global burden of primary liver cancer and its association with underlying aetiologies, sociodemographic status, and sex differences from 1990-2019: A DALY-based analysis of the Global Burden of Disease 2019 study. Clin Mol Hepatol 2023;29:433–452.
21. Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol 2019;16:589–604.
22. Muratori L, Lohse AW, Lenzi M. Diagnosis and management of autoimmune hepatitis. BMJ 2023;380:e070201. Erratum in: BMJ 2023;380:p330.
23. Wu HC, Santella R. The role of aflatoxins in hepatocellular carcinoma. Hepat Mon 2012;12(10 HCC):e7238.
24. Brissot P, Pietrangelo A, Adams PC, de Graaff B, McLaren CE, Loréal O. Haemochromatosis. Nat Rev Dis Primers 2018;4:18016.
25. Koh JH, Ng CH, Nah B, Tan DJH, Loomba R, Huang DQ, ; Nash HCC Transplant Collaborative. NASH is the leading cause of hepatocellular carcinoma in liver transplant candidates. Clin Gastroenterol Hepatol 2024;22:197–199. e3.
26. Noureddin M, Vipani A, Bresee C, Todo T, Kim IK, Alkhouri N, et al. nash leading cause of liver transplant in women: updated analysis of indications for liver transplant and ethnic and gender variances. Am J Gastroenterol 2018;113:1649–1659.
27. Danpanichkul P, Suparan K, Dutta P, Kaeosri C, Sukphutanan B, Pang Y, et al. Disparities in metabolic dysfunction-associated steatotic liver disease and cardiometabolic conditions in low and lower middle-income countries: a systematic analysis from the global burden of disease study 2019. Metabolism 2024;158:155958.
28. Danpanichkul P, Auttapracha T, Sukphutanan B, Ng CH, Wattanachayakul P, Kongarin S, et al. The burden of overweight and obesity-associated gastrointestinal cancers in low and lower-middle-income countries: a global burden of disease 2019 analysis. Am J Gastroenterol 2024;119:1177–1180.
29. Younossi ZM, Golabi P, Paik JM, Henry A, Van Dongen C, Henry L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology 2023;77:1335–1347.
30. Teng ML, Ng CH, Huang DQ, Chan KE, Tan DJ, Lim WH, et al. Global incidence and prevalence of nonalcoholic fatty liver disease. Clin Mol Hepatol 2023;29(Suppl):S32–S42.
31. Danpanichkul P, Ng CH, Muthiah MD, Duangsonk K, Yong JN, Tan DJH, et al. The silent burden of non-alcoholic fatty liver disease in the elderly: a global burden of disease analysis. Aliment Pharmacol Ther 2023;58:1062–1074.
32. Danpanichkul P, Suparan K, Kim D, Wijarnpreecha K. What is new in metabolic dysfunction-associated steatotic liver disease in lean individuals: from bench to bedside. J Clin Med 2024;13:278.
33. Ivancovsky-Wajcman D, Brennan PN, Kopka CJ, Zelber-Sagi S, Younossi ZM, Allen AM, et al. Integrating social nutrition principles into the treatment of steatotic liver disease. Commun Med (Lond) 2023;3:165.
34. Allen AM, Younossi ZM, Diehl AM, Charlton MR, Lazarus JV. Envisioning how to advance the MASH field. Nat Rev Gastroenterol Hepatol 2024;21:726–738.
35. Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M. From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol 2019;16:411–428.
36. Sheka AC, Adeyi O, Thompson J, Hameed B, Crawford PA, Ikramuddin S. Nonalcoholic steatohepatitis: a review. JAMA 2020;323:1175–1183. Erratum in: JAMA 2020;323:1619.
37. Peng C, Stewart AG, Woodman OL, Ritchie RH, Qin CX. Nonalcoholic steatohepatitis: a review of its mechanism, models and medical treatments. Front Pharmacol 2020;11:603926.
38. Armandi A, Bugianesi E. Natural history of NASH. Liver Int 2021;41 Suppl 1(Suppl 1):78–82.
39. Reddy YK, Marella HK, Jiang Y, Ganguli S, Snell P, Podila PSB, et al. Natural history of non-alcoholic fatty liver disease: a study with paired liver biopsies. J Clin Exp Hepatol 2020;10:245–254.
40. Mittal S, El-Serag HB, Sada YH, Kanwal F, Duan Z, Temple S, et al. Hepatocellular carcinoma in the absence of cirrhosis in united states veterans is associated with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2016;14:124–131.e1.
41. Reddy SK, Steel JL, Chen HW, DeMateo DJ, Cardinal J, Behari J, et al. Outcomes of curative treatment for hepatocellular cancer in nonalcoholic steatohepatitis versus hepatitis C and alcoholic liver disease. Hepatology 2012;55:1809–1819.
42. Michelotti GA, Machado MV, Diehl AM. NAFLD, NASH and liver cancer. Nat Rev Gastroenterol Hepatol 2013;10:656–665.
43. Mittal S, Sada YH, El-Serag HB, Kanwal F, Duan Z, Temple S, et al. Temporal trends of nonalcoholic fatty liver disease-related hepatocellular carcinoma in the veteran affairs population. Clin Gastroenterol Hepatol 2015;13:594–601.e1.
44. Geh D, Manas DM, Reeves HL. Hepatocellular carcinoma in non-alcoholic fatty liver disease-a review of an emerging challenge facing clinicians. Hepatobiliary Surg Nutr 2021;10:59–75.
45. Danpanichkul P, Duangsonk K, Diaz LA, Arab JP, Liangpunsakul S, Wijarnpreecha K. Editorial: Sounding the alarm-The rising global burden of adolescent and young adult alcohol-related liver disease. Author’s reply. Aliment Pharmacol Ther 2024;60:521–522.
46. Narro GEC, Díaz LA, Ortega EK, Garín MFB, Reyes EC, Delfin PSM, et al. Alcohol-related liver disease: a global perspective. Ann Hepatol 2024;29:101499.
47. Murthy P, Narasimha VL. Effects of the COVID-19 pandemic and lockdown on alcohol use disorders and complications. Curr Opin Psychiatry 2021;34:376–385.
48. Kim JU, Majid A, Judge R, Crook P, Nathwani R, Selvapatt N, et al. Effect of COVID-19 lockdown on alcohol consumption in patients with pre-existing alcohol use disorder. Lancet Gastroenterol Hepatol 2020;5:886–887.
49. Singal AK, Leggio L, DiMartini A. Alcohol use disorder in alcohol-associated liver disease: two sides of the same coin. Liver Transpl 2024;30:200–212.
50. Meza V, Arnold J, Díaz LA, Ayala Valverde M, Idalsoaga F, Ayares G, et al. Alcohol consumption: medical implications, the liver and beyond. Alcohol Alcohol 2022;57:283–291.
51. Hagström H, Thiele M, Sharma R, Simon TG, Roelstraete B, Söderling J, et al. Risk of cancer in biopsy-proven alcohol-related liver disease: a population-based cohort study of 3410 persons. Clin Gastroenterol Hepatol 2022;20:918–929. e8.
52. Huang DQ, Tan DJH, Ng CH, Amangurbanova M, Sutter N, Lin Tay PW, et al. Hepatocellular carcinoma incidence in alcohol-associated cirrhosis: systematic review and metaanalysis. Clin Gastroenterol Hepatol 2023;21:1169–1177.
53. Zeng RW, Ong CEY, Ong EYH, Chung CH, Lim WH, Xiao J, et al. Global prevalence, clinical characteristics, surveillance, treatment allocation, and outcomes of alcohol-associated hepatocellular carcinoma. Clin Gastroenterol Hepatol 2024;22:2394–2402.e15.
54. Szabo G, Saha B, Bukong TN. Alcohol and HCV: implications for liver cancer. Adv Exp Med Biol 2015;815:197–216.
55. Lin CW, Lin CC, Mo LR, Chang CY, Perng DS, Hsu CC, et al. Heavy alcohol consumption increases the incidence of hepatocellular carcinoma in hepatitis B virus-related cirrhosis. J Hepatol 2013;58:730–735.
56. Díaz LA, Arab JP, Louvet A, Bataller R, Arrese M. The intersection between alcohol-related liver disease and nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2023;20:764–783.
57. Aboona MB, Danpanichkul P, Chen VL, Rangan P, Kim D, Alkhouri N, et al. Mortality outcomes in individuals with MASLD versus MASLD and increased alcohol intake. J Gastroenterol Hepatol 2024;39:2456–2463.
58. Levrero M, Zucman-Rossi J. Mechanisms of HBV-induced hepatocellular carcinoma. J Hepatol 2016;64(1 Suppl):S84–S101.
59. Kamal H, Fornes R, Simin J, Stål P, Duberg AS, Brusselaers N, et al. Risk of hepatocellular carcinoma in hepatitis B and D virus co-infected patients: a systematic review and meta-analysis of longitudinal studies. J Viral Hepat 2021;28:1431–1442.
60. Negro F, Lok AS. Hepatitis D: a review. JAMA 2023;330:2376–2387.
61. Tseng TC, Liu CJ, Hsu CY, Hong CM, Su TH, Yang WT, et al. High Level of hepatitis B core-related antigen associated with increased risk of hepatocellular carcinoma in patients with chronic HBV infection of intermediate viral load. Gastroenterology 2019;157:1518–1529.e3.
62. Yang HI, Yeh SH, Chen PJ, Iloeje UH, Jen CL, Su J, et al, ; REVEAL-HBV Study Group. Associations between hepatitis B virus genotype and mutants and the risk of hepatocellular carcinoma. J Natl Cancer Inst 2008;100:1134–1143.
63. Suk-Fong Lok A. Hepatitis B treatment: what we know now and what remains to be researched. Hepatol Commun 2018;3:8–19.
64. Choi WM, Kim GA, Choi J, Choi GH, Lee YB, Sinn DH, et al. Non-linear association of baseline viral load with on-treatment hepatocellular carcinoma risk in chronic hepatitis B. Gut 2024;73:649–658.
65. Kim GA, Lim YS, Han S, Choi GH, Choi WM, Choi J, et al. Viral load-based prediction of hepatocellular carcinoma risk in noncirrhotic patients with chronic hepatitis B: a multinational study for the development and external validation of a new prognostic model. Ann Intern Med 2024;177:1308–1318.
66. Hoang JK, Yang HI, Le A, Nguyen NH, Lin D, Vu VD, et al. Lower liver cancer risk with antiviral therapy in chronic hepatitis B patients with normal to minimally elevated ALT and no cirrhosis. Medicine (Baltimore) 2016;95:e4433.
67. Huang ZH, Lu GY, Qiu LX, Zhong GH, Huang Y, Yao XM, et al. Risk of hepatocellular carcinoma in antiviral treatment-naïve chronic hepatitis B patients treated with entecavir or tenofovir disoproxil fumarate: a network meta-analysis. BMC Cancer 2022;22:287.
68. Liang YJ, Chiou YW, Chiu AP, Shiao MS, Teng W, Lin CW, et al. Antiviral therapy reduces hepatocellular carcinoma through suppressing hepatitis B virus replication may improve ER stress, mitochondrial and metabolic dysfunctions and decrease p62 in hybridized mice with single HBV transgene and miR-122. J Med Virol 2023;95:e29325.
69. Lin CL, Kao JH. Development of hepatocellular carcinoma in treated and untreated patients with chronic hepatitis B virus infection. Clin Mol Hepatol 2023;29:605–622.
70. Lin MV, King LY, Chung RT. Hepatitis C virus-associated cancer. Annu Rev Pathol 2015;10:345–370.
71. Lazarus JV, Picchio CA, Guy D, Aleman S, James C, Nava FA, et al. Hepatitis C standards of care: a review of good practices since the advent of direct-acting antiviral therapy. Clin Res Hepatol Gastroenterol 2021;45:101564.
72. Lazarus JV, Picchio CA, Colombo M. Hepatocellular carcinoma prevention in the era of hepatitis C elimination. Int J Mol Sci 2023;24:14404.
73. Lazarus JV, Pericàs JM, Picchio C, Cernosa J, Hoekstra M, Luhmann N, et al. We know DAAs work, so now what? Simplifying models of care to enhance the hepatitis C cascade. J Intern Med 2019;286:503–525. Erratum in: J Intern Med 2020;287:455-456.
74. Marshall AD, Willing AR, Kairouz A, Cunningham EB, Wheeler A, O’Brien N, et al, ; Global HCV and HIV Treatment Restrictions Group. Direct-acting antiviral therapies for hepatitis C infection: global registration, reimbursement, and restrictions. Lancet Gastroenterol Hepatol 2024;9:366–382.
75. Kanwal F, Kramer J, Asch SM, Chayanupatkul M, Cao Y, El-Serag HB. Risk of hepatocellular cancer in HCV patients treated with direct-acting antiviral agents. Gastroenterology 2017;153:996–1005.e1.
76. Chen Q, Ayer T, Adee MG, Wang X, Kanwal F, Chhatwal J. Assessment of incidence of and surveillance burden for hepatocellular carcinoma among patients with hepatitis C in the era of direct-acting antiviral agents. JAMA Netw Open 2020;3:e2021173.
77. Luna-Cuadros MA, Chen HW, Hanif H, Ali MJ, Khan MM, Lau DT. Risk of hepatocellular carcinoma after hepatitis C virus cure. World J Gastroenterol 2022;28:96–107.
78. Groopman JD, Smith JW, Rivera-Andrade A, Alvarez CS, Kroker-Lobos MF, Egner PA, et al. Aflatoxin and the etiology of liver cancer and its implications for guatemala. World Mycotoxin J 2021;14:305–317.
79. Valencia-Quintana R, Milić M, Jakšić D, Šegvić Klarić M, Tenorio-Arvide MG, Pérez-Flores GA, et al. Environment changes, aflatoxins, and health issues, a review. Int J Environ Res Public Health 2020;17:7850.
80. Magnussen A, Parsi MA. Aflatoxins, hepatocellular carcinoma and public health. World J Gastroenterol 2013;19:1508–1512.
81. Kew MC. Aflatoxins as a cause of hepatocellular carcinoma. J Gastrointestin Liver Dis 2013;22:305–310.
82. Hsu IC, Metcalf RA, Sun T, Welsh JA, Wang NJ, Harris CC. Mutational hotspot in the p53 gene in human hepatocellular carcinomas. Nature 1991;350:427–428.
83. Soini Y, Chia SC, Bennett WP, Groopman JD, Wang JS, DeBenedetti VM, et al. An aflatoxin-associated mutational hotspot at codon 249 in the p53 tumor suppressor gene occurs in hepatocellular carcinomas from Mexico. Carcinogenesis 1996;17:1007–1012.
84. Liu Y, Chang CC, Marsh GM, Wu F. Population attributable risk of aflatoxin-related liver cancer: systematic review and meta-analysis. Eur J Cancer 2012;48:2125–2136.
85. Kew MC. Synergistic interaction between aflatoxin B1 and hepatitis B virus in hepatocarcinogenesis. Liver Int 2003;23:405–409.
86. 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. Hepatology 2023;78:1922–1965. Erratum in: Hepatology 2023;78:E105.
87. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: management of hepatocellular carcinoma. J Hepatol 2018;69:182–236. Erratum in: J Hepatol 2019;70:817.
88. Omata M, Cheng AL, Kokudo N, Kudo M, Lee JM, Jia J, et al. Asia-Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update. Hepatol Int 2017;11:317–370.
89. Marrero JA, Kulik LM, Sirlin CB, Zhu AX, Finn RS, Abecassis MM, et al. Diagnosis, staging, and management of hepatocellular carcinoma: 2018 practice guidance by the American Association for the Study of Liver Diseases. Hepatology 2018;68:723–750.
90. Parikh ND, Singal AG, Hutton DW, Tapper EB. Cost-effectiveness of hepatocellular carcinoma surveillance: an assessment of benefits and harms. Am J Gastroenterol 2020;115:1642–1649.
91. Trevisani F, Santi V, Gramenzi A, Di Nolfo MA, Del Poggio P, Benvegnù L, et al, ; Italian Liver Cancer Group. Surveillance for early diagnosis of hepatocellular carcinoma: is it effective in intermediate/advanced cirrhosis? Am J Gastroenterol 2007;102:2448–2457. quiz 2458.
92. Kao SZ, Sangha K, Fujiwara N, Hoshida Y, Parikh ND, Singal AG. Cost-effectiveness of a precision hepatocellular carcinoma surveillance strategy in patients with cirrhosis. EClinicalMedicine 2024;75:102755.
93. Zhang BH, Yang BH, Tang ZY. Randomized controlled trial of screening for hepatocellular carcinoma. J Cancer Res Clin Oncol 2004;130:417–422.
94. Singal AG, Zhang E, Narasimman M, Rich NE, Waljee AK, Hoshida Y, et al. HCC surveillance improves early detection, curative treatment receipt, and survival in patients with cirrhosis: a meta-analysis. J Hepatol 2022;77:128–139.
95. Yang J, Yang Z, Zeng X, Yu S, Gao L, Jiang Y, et al. Benefits and harms of screening for hepatocellular carcinoma in high-risk populations: systematic review and meta-analysis. J Natl Cancer Cent 2023;3:175–185.
96. Daher D, Seif El Dahan K, Rich NE, Tayob N, Merrill V, et al. Hepatocellular carcinoma screening in a contemporary cohort of at-risk patients. JAMA Netw Open 2024;7:e248755.
97. Narasimman M, Hernaez R, Cerda V, Lee M, Sood A, Yekkaluri S, et al. Hepatocellular carcinoma surveillance may be associated with potential psychological harms in patients with cirrhosis. Hepatology 2024;79:107–117.
98. Narasimman M, Hernaez R, Cerda V, Lee M, Yekkaluri S, Khan A, et al. Financial burden of hepatocellular carcinoma screening in patients with cirrhosis. Clin Gastroenterol Hepatol 2024;22:760–767. e1.
99. Lee YT, Fujiwara N, Yang JD, Hoshida Y. Risk stratification and early detection biomarkers for precision HCC screening. Hepatology 2023;78:319–362.
100. Liu J, Ayada I, Zhang X, Wang L, Li Y, Wen T, et al. Estimating global prevalence of metabolic dysfunction-associated fatty liver disease in overweight or obese adults. Clin Gastroenterol Hepatol 2022;20:e573–e582.
101. Marquardt P, Liu PH, Immergluck J, Olivares J, Arroyo A, Rich NE, et al. Hepatocellular carcinoma screening process failures in patients with cirrhosis. Hepatol Commun 2021;5:1481–1489.
102. Singal AG, Tiro JA, Murphy CC, Blackwell JM, Kramer JR, Khan A, et al. Patient-reported barriers are associated with receipt of hepatocellular carcinoma surveillance in a multicenter cohort of patients with cirrhosis. Clin Gastroenterol Hepatol 2021;19:987–995. e1.
103. Simmons OL, Feng Y, Parikh ND, Singal AG. Primary care provider practice patterns and barriers to hepatocellular carcinoma surveillance. Clin Gastroenterol Hepatol 2019;17:766–773.
104. Lee YT, Karim MA, Kum HC, Park S, Rich NE, Noureddin M, et al. Factors associated with unrecognized cirrhosis in patients with hepatocellular carcinoma. Clin Mol Hepatol 2023;29:453–464.
105. Schoenberger H, Rich NE, Jones P, Yekkaluri S, Yopp A, Singal AG; Multi-Ethnic HCC Cohort Investigators. Racial and ethnic disparities in barriers to care in patients with hepatocellular carcinoma. Clin Gastroenterol Hepatol 2023;21:1094–1096. e2.
106. Wolf E, Rich NE, Marrero JA, Parikh ND, Singal AG. Use of hepatocellular carcinoma surveillance in patients with cirrhosis: a systematic review and meta-analysis. Hepatology 2021;73:713–725.
107. Seif El Dahan K, Daher D, Singal AG. Hepatocellular carcinoma surveillance in patients with non-alcoholic fatty liver disease. Clin Mol Hepatol 2023;29(Suppl):S207–S219.
108. Singal A, Volk ML, Waljee A, Salgia R, Higgins P, Rogers MA, et al. Meta-analysis: surveillance with ultrasound for early-stage hepatocellular carcinoma in patients with cirrhosis. Aliment Pharmacol Ther 2009;30:37–47.
109. Wong LL, Reyes RJ, Kwee SA, Hernandez BY, Kalathil SC, Tsai NC. Pitfalls in surveillance for hepatocellular carcinoma: how successful is it in the real world? Clin Mol Hepatol 2017;23:239–248.
110. Tzartzeva K, Obi J, Rich NE, Parikh ND, Marrero JA, Yopp A, et al. Surveillance imaging and alpha fetoprotein for early detection of hepatocellular carcinoma in patients with cirrhosis: a meta-analysis. Gastroenterology 2018;154:1706–1718.e1.
111. Tayob N, Kanwal F, Alsarraj A, Hernaez R, El-Serag HB. The performance of AFP, AFP-3, DCP as biomarkers for detection of hepatocellular carcinoma (HCC): a phase 3 biomarker study in the United States. Clin Gastroenterol Hepatol 2023;21:415–423.e4.
112. El-Serag HB, Jin Q, Tayob N, Salem E, Luster M, Alsarraj A, et al. HES V2.0 outperforms GALAD for detection of HCC: A phase 3 biomarker study in the United States. Hepatology 2025;81:465–475.
113. Tayob N, Corley DA, Christie I, Almers L, Rahal AK, Richardson P, et al. Validation of the updated hepatocellular carcinoma early detection screening algorithm in a community-based cohort of patients with cirrhosis of multiple etiologies. Clin Gastroenterol Hepatol 2021;19:1443–1450.e6. Erratum in: Clin Gastroenterol Hepatol 2021;19:2459.
114. Yang JD, Addissie BD, Mara KC, Harmsen WS, Dai J, Zhang N, et al. GALAD score for hepatocellular carcinoma detection in comparison with liver ultrasound and proposal of GALADUS score. Cancer Epidemiol Biomarkers Prev 2019;28:531–538.
115. Berhane S, Toyoda H, Tada T, Kumada T, Kagebayashi C, Satomura S, et al. Role of the GALAD and BALAD-2 serologic models in diagnosis of hepatocellular carcinoma and prediction of survival in patients. Clin Gastroenterol Hepatol 2016;14:875–886.e6.
116. Singal AG, Tayob N, Mehta A, Marrero JA, El-Serag H, Jin Q, et al. GALAD demonstrates high sensitivity for HCC surveillance in a cohort of patients with cirrhosis. Hepatology 2022;75:541–549.
117. von Felden J, Garcia-Lezana T, Schulze K, Losic B, Villanueva A. Liquid biopsy in the clinical management of hepatocellular carcinoma. Gut 2020;69:2025–2034.
118. Chalasani NP, Porter K, Bhattacharya A, Book AJ, Neis BM, Xiong KM, et al. Validation of a novel multitarget blood test shows high sensitivity to detect early stage hepatocellular carcinoma. Clin Gastroenterol Hepatol 2022;20:173–182.e7.
119. Kisiel JB, Dukek BA, V S R Kanipakam R, Ghoz HM, Yab TC, Berger CK, et al. Hepatocellular carcinoma detection by plasma methylated DNA: discovery, phase I pilot, and phase II clinical validation. Hepatology 2019;69:1180–1192.
120. Sun N, Zhang C, Lee YT, Tran BV, Wang J, Kim H, et al. HCC EV ECG score: an extracellular vesicle-based protein assay for detection of early-stage hepatocellular carcinoma. Hepatology 2023;77:774–788.
121. Gupta P, Soundararajan R, Patel A, Kumar-M P, Sharma V, Kalra N. Abbreviated MRI for hepatocellular carcinoma screening: a systematic review and meta-analysis. J Hepatol 2021;75:108–119.
122. Nahon P, Ronot M, Sutter O, Natella PA, Baloul S, DurandZaleski I, et al. Study protocol for FASTRAK: a randomised controlled trial evaluating the cost impact and effectiveness of FAST-MRI for HCC suRveillance in pAtients with high risK of liver cancer. BMJ Open 2024;14:e083701.
123. Singal AG, Ghaziani TT, Mehta N, Zhou K, Grinspan LT, Benhammou JN, et al. Recall patterns and risk of primary liver cancer for subcentimeter ultrasound liver observations: a multicenter study. Hepatol Commun 2023;7:e0073.
124. Schoenberger H, Chong N, Fetzer DT, Rich NE, Yokoo T, Khatri G, et al. Dynamic changes in ultrasound quality for hepatocellular carcinoma screening in patients with cirrhosis. Clin Gastroenterol Hepatol 2022;20:1561–1569.e4.
125. Lee YT, Wang JJ, Zhu Y, Agopian VG, Tseng HR, Yang JD. Diagnostic criteria and LI-RADS for hepatocellular carcinoma. Clin Liver Dis (Hoboken) 2021;17:409–413.
126. Matsui O, Kobayashi S, Sanada J, Kouda W, Ryu Y, Kozaka K, et al. Hepatocelluar nodules in liver cirrhosis: hemodynamic evaluation (angiography-assisted CT) with special reference to multi-step hepatocarcinogenesis. Abdom Imaging 2011;36:264–272.
127. Sutphin PD, Lamus D, Kalva SP, Li J, Corbin IR. Interventional radiologic therapies for hepatocellular carcinoma: from where we began to where we are going. In: Hoshida Y, ed. Hepatocellular Carcinoma: Translational Precision Medicine Approaches. Cham (CH): Humana Press, 2019.
128. American College of Radiology. CT/MRI LI-RADS® v2018 CORE. American College of Radiology web site, <https://www.acr.org/-/media/ACR/Files/RADS/LI-RADS/LI-RADS2018-Core.pdf>. Accessed 27 Nov 2024.
129. Childs A, Zakeri N, Ma YT, O’Rourke J, Ross P, Hashem E, et al. Biopsy for advanced hepatocellular carcinoma: results of a multicentre UK audit. Br J Cancer 2021;125:1350–1355.
130. Kopystecka A, Patryn R, Leśniewska M, Budzyńska J, Kozioł I. The use of ctDNA in the diagnosis and monitoring of hepatocellular carcinoma-literature review. Int J Mol Sci 2023;24:9342.
131. Craig AJ, von Felden J, Garcia-Lezana T, Sarcognato S, Villanueva A. Tumour evolution in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2020;17:139–152.
132. 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;76:681–693.
133. Parikh ND, Singal AG. The ITA.LI.CA staging system: a novel staging system for hepatocellular carcinoma. PLoS Med 2016;13:e1002005.
134. Yau T, Tang VY, Yao TJ, Fan ST, Lo CM, Poon RT. Development of Hong Kong Liver Cancer staging system with treatment stratification for patients with hepatocellular carcinoma. Gastroenterology 2014;146:1691–1700.e3.
135. 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;34:444–455.
136. Oh JH, Sinn DH. Multidisciplinary approach for hepatocellular carcinoma patients: current evidence and future perspectives. J Liver Cancer 2024;24:47–56.
137. Siddique O, Yoo ER, Perumpail RB, Perumpail BJ, Liu A, Cholankeril G, et al. The importance of a multidisciplinary approach to hepatocellular carcinoma. J Multidiscip Healthc 2017;10:95–100.
138. Yang JD, Heimbach JK. New advances in the diagnosis and management of hepatocellular carcinoma. BMJ 2020;371:m3544.
139. Hyun MH, Lee YS, Kim JH, Lee CU, Jung YK, Seo YS, et al. Hepatic resection compared to chemoembolization in intermediate- to advanced-stage hepatocellular carcinoma: a meta-analysis of high-quality studies. Hepatology 2018;68:977–993.
140. Tsilimigras DI, Mehta R, Paredes AZ, Moris D, Sahara K, Bagante F, et al. Overall tumor burden dictates outcomes for patients undergoing resection of multinodular hepatocellular carcinoma beyond the milan criteria. Ann Surg 2020;272:574–581.
141. Lim M, Kim J, Rhu J, Choi GS, Joh JW. Liver resection in selective hepatocellular carcinoma with Vp3 or Vp4 portal vein tumor thrombosis improves prognosis. J Liver Cancer 2024;24:102–112.
142. Kokudo T, Hasegawa K, Matsuyama Y, Takayama T, Izumi N, Kadoya M, et al, ; Liver Cancer Study Group of Japan. Survival benefit of liver resection for hepatocellular carcinoma associated with portal vein invasion. J Hepatol 2016;65:938–943.
143. Liang L, Chen TH, Li C, Xing H, Han J, Wang MD, et al. A systematic review comparing outcomes of surgical resection and non-surgical treatments for patients with hepatocellular carcinoma and portal vein tumor thrombus. HPB (Oxford) 2018;20:1119–1129.
144. Govalan R, Lauzon M, Luu M, Ahn JC, Kosari K, Todo T, et al. Comparison of surgical resection and systemic treatment for hepatocellular carcinoma with vascular invasion: national cancer database analysis. Liver Cancer 2021;10:407–418.
145. Vitale A, Romano P, Cillo U, ; Writing Group for the HE.RC. O.LE.S Collaborative Group, Writing Group for the ITA.LI.CA Collaborative Group, HE.RC.O.LE.S and ITA.LI.CA Collaborative Groups, et al. Liver resection vs nonsurgical treatments for patients with early multinodular hepatocellular carcinoma. JAMA Surg 2024;159:881–889.
146. Di Benedetto F, Magistri P, Di Sandro S, Sposito C, Oberkofler C, Brandon E, et al, ; Robotic HPB Study Group. Safety and efficacy of robotic vs open liver resection for hepatocellular carcinoma. JAMA Surg 2023;158:46–54.
147. Zheng J, Chou JF, Gönen M, Vachharajani N, Chapman WC, Majella Doyle MB, et al. Prediction of hepatocellular carcinoma recurrence beyond milan criteria after resection: validation of a clinical risk score in an international cohort. Ann Surg 2017;266:693–701.
148. Tabrizian P, Jibara G, Shrager B, Schwartz M, Roayaie S. Recurrence of hepatocellular cancer after resection: patterns, treatments, and prognosis. Ann Surg 2015;261:947–955.
149. Shah SA, Cleary SP, Wei AC, Yang I, Taylor BR, Hemming AW, et al. Recurrence after liver resection for hepatocellular carcinoma: risk factors, treatment, and outcomes. Surgery 2007;141:330–339.
150. Shiina S, Sato K, Tateishi R, Shimizu M, Ohama H, Hatanaka T, et al. Percutaneous ablation for hepatocellular carcinoma: comparison of various ablation techniques and surgery. Can J Gastroenterol Hepatol 2018;2018:4756147.
151. Lencioni R, Cioni D, Crocetti L, Franchini C, Pina CD, Lera J, et al. Early-stage hepatocellular carcinoma in patients with cirrhosis: long-term results of percutaneous image-guided radiofrequency ablation. Radiology 2005;234:961–967.
152. Alexander ES, Wolf FJ, Machan JT, Charpentier KP, Beland MD, Iannuccilli JD, et al. Microwave ablation of focal hepatic malignancies regardless of size: a 9-year retrospective study of 64 patients. Eur J Radiol 2015;84:1083–1090.
153. Wright AS, Sampson LA, Warner TF, Mahvi DM, Lee FT Jr. Radiofrequency versus microwave ablation in a hepatic porcine model. Radiology 2005;236:132–139.
154. Facciorusso A, Abd El Aziz MA, Tartaglia N, Ramai D, Mohan BP, Cotsoglou C, et al. Microwave ablation versus radiofrequency ablation for treatment of hepatocellular carcinoma: a meta-analysis of randomized controlled trials. Cancers (Basel) 2020;12:3796.
155. Nishikawa H, Inuzuka T, Takeda H, Nakajima J, Matsuda F, Sakamoto A, et al. Comparison of percutaneous radiofrequency thermal ablation and surgical resection for small hepatocellular carcinoma. BMC Gastroenterol 2011;11:143.
156. Livraghi T, Meloni F, Di Stasi M, Rolle E, Solbiati L, Tinelli C, et al. Sustained complete response and complications rates after radiofrequency ablation of very early hepatocellular carcinoma in cirrhosis: is resection still the treatment of choice? Hepatology 2008;47:82–89.
157. Lau WY, Leung TW, Yu SC, Ho SK. Percutaneous local ablative therapy for hepatocellular carcinoma: a review and look into the future. Ann Surg 2003;237:171–179.
158. Lu DS, Yu NC, Raman SS, Lassman C, Tong MJ, Britten C, et al. Percutaneous radiofrequency ablation of hepatocellular carcinoma as a bridge to liver transplantation. Hepatology 2005;41:1130–1137.
159. Wang H, Wu Z, Cui D, Shi Y, Zhai B. Radiofrequency ablation of hepatocellular carcinoma: current status, challenges, and prospects. Liver Res 2023;7:108–115.
160. Wang C, Wang H, Yang W, Hu K, Xie H, Hu KQ, et al. Multicenter randomized controlled trial of percutaneous cryoablation versus radiofrequency ablation in hepatocellular carcinoma. Hepatology 2015;61:1579–1590.
161. Frühling P, Stillström D, Holmquist F, Nilsson A, Freedman J. Irreversible electroporation of hepatocellular carcinoma and colorectal cancer liver metastases: a nationwide multicenter study with short- and long-term follow-up. Eur J Surg Oncol 2023;49:107046.
162. Bruix J, Takayama T, Mazzaferro V, Chau GY, Yang J, Kudo M, et al, ; STORM investigators. Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol 2015;16:1344–1354.
163. Qin S, Chen M, Cheng AL, Kaseb AO, Kudo M, Lee HC, et al, ; IMbrave050 investigators. 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;402:1835–1847.
164. Yopp A, Kudo M, Chen M, Cheng AL, Kaseb AO, Lee HC, et al. Updated efficacy and safety data from IMbrave050: Phase III study of adjuvant atezolizumab (atezo) + bevacizumab (bev) vs active surveillance in patients (pts) with resected or ablated high-risk hepatocellular carcinoma (HCC). Paper presented at: ESMO Congress 2024; 2024 Sep 13-17; Barcelona, Spain.
165. Chick RC, Ruff SM, Pawlik TM. Neoadjuvant systemic therapy for hepatocellular carcinoma. Front Immunol 2024;15:1355812.
166. Ho WJ, Zhu Q, Durham J, Popovic A, Xavier S, Leatherman J, et al. Neoadjuvant cabozantinib and nivolumab converts locally advanced HCC into resectable disease with enhanced antitumor immunity. Nat Cancer 2021;2:891–903.
167. Kaseb AO, Hasanov E, Cao HST, Xiao L, Vauthey JN, Lee SS, et al. Perioperative nivolumab monotherapy versus nivolumab plus ipilimumab in resectable hepatocellular carcinoma: a randomised, open-label, phase 2 trial. Lancet Gastroenterol Hepatol 2022;7:208–218.
168. Organ Procurement and Transplantation Network (OPTN) and Scientific Registry of Transplant Recipients (SRTR). OPTN/SRTR 2022 Annual Data Report. U.S. Department of Health and Human Services, Health Resources and Services Administration web site, <http://srtr.transplant.hrsa.gov/annual_reports/Default.aspx>. Accessed 9 Aug 2024.
169. Zimmerman MA, Ghobrial RM, Tong MJ, Hiatt JR, Cameron AM, Hong J, et al. Recurrence of hepatocellular carcinoma following liver transplantation: a review of preoperative and postoperative prognostic indicators. Arch Surg 2008;143:182–188. discussion 188.
170. Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, et al. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med 1996;334:693–699.
171. Yao FY, Xiao L, Bass NM, Kerlan R, Ascher NL, Roberts JP. Liver transplantation for hepatocellular carcinoma: validation of the UCSF-expanded criteria based on preoperative imaging. Am J Transplant 2007;7:2587–2596.
172. Bento de Sousa JH, Calil IL, Tustumi F, da Cunha Khalil D, Felga GEG, de Arruda Pecora RA, et al. Comparison between Milan and UCSF criteria for liver transplantation in patients with hepatocellular carcinoma: a systematic review and meta-analysis. Transl Gastroenterol Hepatol 2021;6:11.
173. Duffy JP, Vardanian A, Benjamin E, Watson M, Farmer DG, Ghobrial RM, et al. Liver transplantation criteria for hepatocellular carcinoma should be expanded: a 22-year experience with 467 patients at UCLA. Ann Surg 2007;246:502–509. discussion 509-511.
174. Tabrizian P, Holzner ML, Mehta N, Halazun K, Agopian VG, Yao F, et al. Ten-year outcomes of liver transplant and downstaging for hepatocellular carcinoma. JAMA Surg 2022;157:779–788.
175. Organ Procurement and Transplantation Network. OPTN policy 2024. OPTN web site, <https://optn.transplant.hrsa.gov/media/eavh5bf3/optn_policies.pdf>. Accessed 12 Aug 2024.
176. Mehta N, Frenette C, Tabrizian P, Hoteit M, Guy J, Parikh N, et al. Downstaging outcomes for hepatocellular carcinoma: results from the Multicenter Evaluation of Reduction in Tumor Size before Liver Transplantation (MERITS-LT) consortium. Gastroenterology 2021;161:1502–1512.
177. Natarajan B, Tabrizian P, Hoteit M, Frenette C, Parikh N, Ghaziani T, et al. Downstaging hepatocellular carcinoma before liver transplantation: a multicenter analysis of the “all-comers” protocol in the Multicenter Evaluation of Reduction in Tumor Size before Liver Transplantation (MERITS-LT) consortium. Am J Transplant 2023;23:1771–1780.
178. Shan Y, Huang L, Xia Q. Salvage liver transplantation leads to poorer outcome in hepatocellular carcinoma compared with primary liver transplantation. Sci Rep 2017;7:44652.
179. Majno PE, Sarasin FP, Mentha G, Hadengue A. Primary liver resection and salvage transplantation or primary liver transplantation in patients with single, small hepatocellular carcinoma and preserved liver function: an outcome-oriented decision analysis. Hepatology 2000;31:899–906.
180. Lim C, Shinkawa H, Hasegawa K, Bhangui P, Salloum C, Gomez Gavara C, et al. Salvage liver transplantation or repeat hepatectomy for recurrent hepatocellular carcinoma: an intent-to-treat analysis. Liver Transpl 2017;23:1553–1563.
181. Chen CL, Kabiling CS, Concejero AM. Why does living donor liver transplantation flourish in Asia? Nat Rev Gastroenterol Hepatol 2013;10:746–751.
182. Elkomos BE, Abdo M, Mamdouh R, Abdelaal A. Can living donor liver transplantation provide similar outcomes to deceased-donor liver transplantation for hepatocellular carcinoma? A systematic review and meta-analysis. Hepatol Int 2023;17:18–37.
183. Raoul JL, Forner A, Bolondi L, Cheung TT, Kloeckner R, de Baere T. Updated use of TACE for hepatocellular carcinoma treatment: How and when to use it based on clinical evidence. Cancer Treat Rev 2019;72:28–36.
184. Young M, John S. Hepatic chemoembolization. Treasure Island (FL): StatPearls Publishing, 2023.
185. Llovet JM, Real MI, Montaña X, Planas R, Coll S, Aponte J, et al, ; Barcelona Liver Cancer Group. Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet 2002;359:1734–1739.
186. Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, Poon RT, et al. Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology 2002;35:1164–1171.
187. Kudo M, Matsui O, Izumi N, Kadoya M, Okusaka T, Miyayama S, et al, ; Liver Cancer Study Group of Japan. Transarterial chemoembolization failure/refractoriness: JSH-LCSGJ criteria 2014 update. Oncology 2014;87 Suppl 2:22–31. Erratum in: Oncology 2015;89 Suppl 2:71-72.
188. Brown KT, Do RK, Gonen M, Covey AM, Getrajdman GI, Sofocleous CT, et al. Randomized trial of hepatic artery embolization for hepatocellular carcinoma using doxorubicin-eluting microspheres compared with embolization with microspheres alone. J Clin Oncol 2016;34:2046–2053.
189. Salem R, Johnson GE, Kim E, Riaz A, Bishay V, Boucher E, et al. Yttrium-90 radioembolization for the treatment of solitary, unresectable HCC: the LEGACY study. Hepatology 2021;74:2342–2352.
190. Salem R, Gordon AC, Mouli S, Hickey R, Kallini J, Gabr A, et al. Y90 radioembolization significantly prolongs time to progression compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology 2016;151:1155–1163.e2.
191. Brown AM, Kassab I, Massani M, Townsend W, Singal AG, Soydal C, et al. TACE versus TARE for patients with hepatocellular carcinoma: overall and individual patient level meta analysis. Cancer Med 2023;12:2590–2599.
192. Dhondt E, Lambert B, Hermie L, Huyck L, Vanlangenhove P, Geerts A, et al. 90Y radioembolization versus drug-eluting bead chemoembolization for unresectable hepatocellular carcinoma: results from the TRACE phase II randomized controlled trial. Radiology 2022;303:699–710.
193. Garin E, Tselikas L, Guiu B, Chalaye J, Rolland Y, de Baere T, et al. Long-term overall survival after selective internal radiation therapy for locally advanced hepatocellular carcinomas: updated analysis of DOSISPHERE-01 trial. J Nucl Med 2024;65:264–269.
194. Lewis S, Dawson L, Barry A, Stanescu T, Mohamad I, Hosni A. Stereotactic body radiation therapy for hepatocellular carcinoma: from infancy to ongoing maturity. JHEP Rep 2022;4:100498.
195. Durand-Labrunie J, Baumann AS, Ayav A, Laurent V, Boleslawski E, Cattan S, et al. Curative irradiation treatment of hepatocellular carcinoma: a multicenter phase 2 trial. Int J Radiat Oncol Biol Phys 2020;107:116–125.
196. Yoon SM, Kim SY, Lim YS, Kim KM, Shim JH, Lee D, et al. Stereotactic body radiation therapy for small (≤5 cm) hepatocellular carcinoma not amenable to curative treatment: results of a single-arm, phase II clinical trial. Clin Mol Hepatol 2020;26:506–515.
197. Liu Y, Yan J, Wang F. Effects of TACE combined with precise RT on p53 gene expression and prognosis of HCC patients. Oncol Lett 2018;16:5733–5738.
198. Kang JK, Kim MS, Cho CK, Yang KM, Yoo HJ, Kim JH, et al. Stereotactic body radiation therapy for inoperable hepatocellular carcinoma as a local salvage treatment after incomplete transarterial chemoembolization. Cancer 2012;118:5424–5431.
199. Yoon SM, Ryoo BY, Lee SJ, Kim JH, Shin JH, An JH, et al. Efficacy and safety of transarterial chemoembolization plus external beam radiotherapy vs sorafenib in hepatocellular carcinoma with macroscopic vascular invasion: a randomized clinical trial. JAMA Oncol 2018;4:661–669.
200. Lee SU, Kim TH. Current evidence and the potential role of proton beam therapy for hepatocellular carcinoma. Clin Mol Hepatol 2023;29:958–968.
201. Kim TH, Koh YH, Kim BH, Kim MJ, Lee JH, Park B, et al. Proton beam radiotherapy vs. radiofrequency ablation for recurrent hepatocellular carcinoma: a randomized phase III trial. J Hepatol 2021;74:603–612.
202. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al, ; IMbrave150 Investigators. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894–1905.
203. Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, et al. Tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. NEJM Evid 2022;1:EVIDoa2100070.
204. Yau T, Kang YK, Kim TY, El-Khoueiry AB, Santoro A, Sangro B, et al. Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: the CheckMate 040 randomized clinical trial. JAMA Oncol 2020;6:e204564. Erratum in: JAMA Oncol 2021;7:140.
205. Galle PR, Decaens T, Kudo M, Qin S, Fonseca L, Sangro B, et al. Nivolumab (NIVO) plus ipilimumab (IPI) vs lenvatinib (LEN) or sorafenib (SOR) as first-line treatment for unresectable hepatocellular carcinoma (uHCC): first results from CheckMate 9DW. J Clin Oncol 2024;42(17 suppl):LBA4008–LBA4008.
206. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al, ; SHARP Investigators Study Group. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359:378–390.
207. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet 2018;391:1163–1173.
208. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al, ; RESORCE Investigators. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;389:56–66. Erratum in: Lancet 2017;389:36.
209. Kelley RK, Ryoo BY, Merle P, Park JW, Bolondi L, Chan SL, et al. Second-line cabozantinib after sorafenib treatment for advanced hepatocellular carcinoma: a subgroup analysis of the phase 3 CELESTIAL trial. ESMO Open 2020;5:e000714.
210. Zhu AX, Kang YK, Yen CJ, Finn RS, Galle PR, Llovet JM, et al, ; REACH-2 study investigators. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased α-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2019;20:282–296.
211. Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, et al, ; KEYNOTE-240 investigators. Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase III trial. J Clin Oncol 2020;38:193–202.
212. Qin S, Kudo M, Meyer T, Bai Y, Guo Y, Meng Z, et al. Tislelizumab vs sorafenib as first-line treatment for unresectable hepatocellular carcinoma: a phase 3 randomized clinical trial. JAMA Oncol 2023;9:1651–1659.
213. Qin S, Chan SL, Gu S, Bai Y, Ren Z, Lin X, et al, ; CARES-310 Study Group. Camrelizumab plus rivoceranib versus sorafenib as first-line therapy for unresectable hepatocellular carcinoma (CARES-310): a randomised, open-label, international phase 3 study. Lancet 2023;402:1133–1146.
214. Sankar K, Gong J, Osipov A, Miles SA, Kosari K, Nissen NN, et al. Recent advances in the management of hepatocellular carcinoma. Clin Mol Hepatol 2024;30:1–15.
215. Gordan JD, Kennedy EB, Abou-Alfa GK, Beal E, Finn RS, Gade TP, et al. Systemic therapy for advanced hepatocellular carcinoma: ASCO guideline update. J Clin Oncol 2024;42:1830–1850.
216. Fulgenzi CAM, Cheon J, D’Alessio A, Nishida N, Ang C, Marron TU, et al. Reproducible safety and efficacy of atezolizumab plus bevacizumab for HCC in clinical practice: results of the AB-real study. Eur J Cancer 2022;175:204–213.
217. Kulkarni AV, Tevethia H, Kumar K, Premkumar M, Muttaiah MD, Hiraoka A, et al. Effectiveness and safety of atezolizumab-bevacizumab in patients with unresectable hepatocellular carcinoma: a systematic review and meta-analysis. EClinicalMedicine 2023;63:102179.
218. Hatanaka T, Naganuma A, Yata Y, Kakizaki S. Atezolizumab plus bevacizumab and tremelimumab plus durvalumab: how should we choose these two immunotherapy regimens for advanced hepatocellular carcinoma? Hepatobiliary Surg Nutr 2022;11:927–930.
219. Weinmann A, Niederle IM, Koch S, Hoppe-Lotichius M, Heise M, Düber C, et al. Sorafenib for recurrence of hepatocellular carcinoma after liver transplantation. Dig Liver Dis 2012;44:432–437.
220. Duffy AG, Ulahannan SV, Makorova-Rusher O, Rahma O, Wedemeyer H, Pratt D, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol 2017;66:545–551.
221. Lencioni R, Kudo M, Erinjeri J, Qin S, Ren Z, Chan S, et al. EMERALD-1: a phase 3, randomized, placebo-controlled study of transarterial chemoembolization combined with durvalumab with or without bevacizumab in participants with unresectable hepatocellular carcinoma eligible for embolization. J Clin Oncol 2024;42(suppl 3):abstr–LBA432.
222. Jin ZC, Chen JJ, Zhu XL, Duan XH, Xin YJ, Zhong BY, et al, ; CHANCE2201 Investigators. Immune checkpoint inhibitors and anti-vascular endothelial growth factor antibody/tyrosine kinase inhibitors with or without transarterial chemoembolization as first-line treatment for advanced hepatocellular carcinoma (CHANCE2201): a target trial emulation study. EClinicalMedicine 2024;72:102622.
223. Su GL, Altayar O, O’Shea R, Shah R, Estfan B, Wenzell C, et al. AGA clinical practice guideline on systemic therapy for hepatocellular carcinoma. Gastroenterology 2022;162:920–934.
224. Xie E, Yeo YH, Scheiner B, Zhang Y, Hiraoka A, Tantai X, et al. Immune checkpoint inhibitors for Child-Pugh Class B advanced hepatocellular carcinoma: a systematic review and meta-analysis. JAMA Oncol 2023;9:1423–1431.
225. Chen X, Kou L, Xie X, Su S, Li J, Li Y. Prognostic biomarkers associated with immune checkpoint inhibitors in hepatocellular carcinoma. Immunology 2024;172:21–45.
226. Desai R, Jiang Y, VanWagner LB, Singal AG, Lieber SR. Financial burden in a US cohort of patients with HCC. Hepatol Commun 2024;8:e0453.
227. Laube R, Sabih AH, Strasser SI, Lim L, Cigolini M, Liu K. Palliative care in hepatocellular carcinoma. J Gastroenterol Hepatol 2021;36:618–628.
228. Woodrell CD, Hansen L, Schiano TD, Goldstein NE. Palliative care for people with hepatocellular carcinoma, and specific benefits for older adults. Clin Ther 2018;40:512–525.

Article information Continued

Figure 1.

(A) Incidences attributable to liver cancer from 2000 to 2021, by etiology. (B) Deaths attributable to liver cancer from 2000 to 2021, by etiology. (C) Age-standardized incidence rates attributable to liver cancer in 2021 per 100,000 population by country. (D) Agestandardized death rates attributable to liver cancer in 2021 per 100,000 population by country. (E) Proportion of incidences of liver cancer attributable to different etiologies from 2000 to 2021. (F) Proportion of deaths from liver cancer attributable to different etiologies from 2000 to 2021. ALD, alcohol-associated liver disease; HBV, hepatitis B virus; HCV, hepatitis C virus; MASH, metabolic dysfunction-associated steatohepatitis.

Figure 2.

Recent advances in the management of hepatocellular carcinoma. Created in BioRender. Hwang S (2024) https://BioRender. com/u75r972. EBRT, external beam radiation therapy; HCC, hepatocellular carcinoma; IRE, irreversible electroporatio; MWA, microwave ablation; PBT, proton beam therapy; RFA, radiofrequency ablation; SBRT, stereotactic body radiation therapy; TACE, transarterial chemoembolization; TARE, transarterial radioembolization.

Table 1.

Summary of surveillance tools recommended for hepatocellular carcinoma based on international society guidelines

Surveillance recommendation European Association for the Study of the Liver (EASL) 2018 [87] American Association for the Study of Liver Diseases (AASLD) 2023 [86] Asian Pacific Association for the Study of the Liver (APASL) 2017 [88]
Indication for surveillance - Cirrhotic patients, Child-Pugh stage A-B or Child-Pugh stage C waiting transplantation - Child-Pugh A-B cirrhosis, any etiology - Cirrhotic hepatitis patients, any etiology
- Non-cirrhotic HBV patients at intermediate (PAGE-B 10-17) or high risk of HCC (PAGE-B ≥18) (for Caucasians) - Child-Pugh C cirrhosis, transplant candidate - Chronic HBV carriers, non-cirrhotic (HBsAg positive): Asian females >50 yr Asian males >40 yr Africans aged >20 yr History of HCC in family
- Non-cirrhotic F3 patients considered for surveillance based on individual risk assessment - Non-cirrhotic chronic hepatitis B: Males aged >40 yr from endemic country
Patients on waiting list for transplantation to detect, manage tumour occurrence/ response, and help define priority policies for transplantation (Strong Recommendation) Females aged >50 yr from endemic country
Person from Africa at earlier age
Family history of HCC
Role for NAFLD without cirrhosis unclear PAGE-B score ≥10
- Need of risk stratification: hepatitis C and stage 3 fibrosis, noncirrhotic NAFLD
Patients listed for transplantation should undergo HCC surveillance (Strong Recommendation)
Surveillance interval Every 6 mo
Surveillance modality - Abdominal ultrasound by experienced personnel (Strong Recommendation) - Ultrasound and AFP (Strong Recommendation) - Combination of US and serum AFP (Weaker Recommendation)
- Multidetector CT or dynamic MRI are not cost-effective has high false-positive results in general, but might be considered in listed patients for liver transplant or inadequate US assessment (e.g., obesity, intestinal gas) - Do not recommend routine use of CT/MRI-based imaging and tumor biomarkers, outside of AFP (Weak Recommendation) - AFP alone is not recommended for routine surveillance (Strong Recommendation)
- Tumour biomarkers for accurate early detection still lacking. Biomarkers tested (i.e., AFP, AFP-L3 and DCP) are suboptimal in terms of cost-effectiveness for routine surveillance of early HCC - Alternative imaging modalities, such as contrast-enhanced MRI, may be considered in select patients in whom US-based surveillance is suboptimal (Weak Recommendation) - Cut-off value of AFP should be set at 200 ng/mL when used in combination with US. It can be set at lower value in a population with hepatitis virus suppression/eradication (Weaker Recommendation)

AFP, alpha-fetoprotein; CT, computed tomography; DCP, des gamma carboxy prothrombin; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; MRI, magnetic resonance imaging; NAFLD, nonalcoholic fatty liver disease; US, ultrasound.

Table 2.

Summary of the accuracy of surveillance tools for hepatocellular carcinoma

Surveillance tools Sensitivity Specificity Comments
Ultrasound (US) [110] 27.9–100% (all HCC) 70.7–99.6% (all HCC) · Pooled sensitivities for all HCC 84% (95% CI 76–92%)
21.4–68.2% (early-stage HCC) 89.7–96.2% (early-stage HCC) · Pooled sensitivities for early-stage HCC 45% (95% CI 30–62%)
US plus AFP [110] 32.6–100% (early-stage HCC) 82.7–88.2% (early-stage HCC) · Pooled sensitivities for early-stage HCC 63% (95% CI 48–75%)
AFP-L3% [111] 62% 90%
DCP [111] 40% 81%
GALAD model [111] 62.9–73.3% (all HCC) 90% · Including gender, age, AFP-L3, AFP, des-gamma-carboxy prothrombin
51.9–62.5% (early-stage HCC) · Phase 3 biomarker study
Hepatocellular carcinoma early detection screening (HES) algorithm V2.0 [112,113] 47.2% (any time before HCC diagnosis) 90.0% (fixed FPR of 10%) · Age, etiology, AFP, ALT, platelet included in HES V1.0 and in addition AFP-L3 and des-gamma-carboxy prothrombin included in HES V2.0
· AUROC for any HCC and early-stage HCC at any time is 0.77 and 0.77 respectively
Abbreviated MRI [121] 86% 94% · Sensitivity of AMRI for detection of HCC <2 cm versus HCC ≥2 cm (69% vs. 86%)
· Non-contrast AMRI versus contrast-enhanced AMRI (sensitivity 86% and 94% vs. specificity 87% and 94%, respectively)
Six-marker methylated DNA marker (MDM) panel [119] 95% 92% · Including HOXA1, EMX1, AK055957, ECE1, PFKP, CLEC11A normalized by B3GALT6 level
· AUC of 0.96 (95% CI, 0.93–0.99) in a phase 2 trial
Extracellular vesicle (EV) surface protein assay [120] 91% (early-stage HCC) 90% (early-stage HCC) · AUC of 0.95 (95% CI, 0.90–0.99) in early-stage HCC

AFP, alpha-fetoprotein; ALT, alanine aminotransferase; AUC, area under the curve; AUROC, Area Under the Receiver Operating Characteristic curve; CI, confidence interval; DCP, des gamma carboxy prothrombin; FPR, false positive rate; HCC, hepatocellular carcinoma; MRI, magnetic resonance imaging.

Table 3.

Summary of the efficacy of systemic treatment with proven benefit in phase 3 RCTs

First-line treatment
Monotherapy (TKIs/ICIs)
Study population Comparator Median overall survival Hazard ratio (95% CI)
Systemic treatment (Trial)
 Sorafenib (SHARP) [206] Advanced HCC patients without prior systemic therapy, ECOG PS 0-2 and Child-Pugh class A Sorafenib 400 mg twice daily versus placebo 10.7 vs. 7.9 mo 0.69 (0.55–0.87)
 Lenvatinib (REFLECT) [207] Unresectable HCC, BCLC stage B or C, Child-Pugh class A, ECOG PS 0-1 Lenvatinib 12 mg/day (for body weight ≥60 kg) or 8 mg/day (for body weights <60 kg) versus sorafenib 400 mg twice daily (non-inferiority trial) 13.6 vs. 12.3 mo 0.92 (0.79–1.06)
 Tislelizumab (RATIONALE-301) [212] HCC with disease progression following (or patient not amenable to) locoregional therapy BCLC stage B or C, Tislelizumab, 200 mg intravenously every 3 wk, or sorafenib tosylate, 400 mg orally twice daily (non-inferiority trial) 15.9 vs. 14.1 mo 0.85 (0.71–1.02)
Child-Pugh class A, ECOG 0-1
Combination therapy
 Nivolumab+Ipilimumab (CheckMate-9DW) [205] Previously untreated HCC not eligible for curative Tx, Child-Pugh score 5-6, ECOG PS 0-1 Nivolumab 1 mg/kg plus ipilimumab 3 mg/kg followed by nivolumab 480 mg every 4 wk versus lenvatinib 8 or 12 mg daily or sorafenib 400 mg twice daily 23.7 vs. 20.6 mo 0.79 (0.65–0.96)
 Atezolizumab+bevacizumab (IMBrave150) [202] Unresectable HCC, Child-Pugh class A, ECOG 0-1 Atezolizumab 1,200 mg plus bevacizumab 15 mg/kg every 3 wk versus sorafenib 400 mg twice daily NE vs. 13.2 mo 0.58 (0.42–0.79)
 Tremelimumab+durvalumab (HIMALAYA) [203] HCC patients ineligible for locoregional therapy, BCLC stage B or C, Child-Pugh class A, ECOG 0-1 STRIDE (tremelimumab 300 mg plus durvalumab 1,500 mg) versus durvalumab 1,500 mg versus sorafenib 400 mg twice daily 16.43 vs. 16.56 vs. 13.77 mo 0.78 (0.65–0.93) (STRIDE)
0.86 (0.73–1.03) (Durvalumab)
 Camrelizumab+rivoceranib (CARES-310) [213] Advanced HCC in patients not amenable to or had progressed after surgical or locoregional therapy, BCLC stage B or C, Child-Pugh class A, ECOG 0-1 Camrelizumab 200 mg IV every 2 wk plus rivoceranib 250 mg orally once daily versus sorafenib 400 mg twice daily 22.1 vs. 15.2 mo 0.62 (0.49–0.80)
Second or subsequent line treatment
 Monotherapy (TKIs/ICIs)
  Regorafenib (RESOURCE) [208] HCC patients who tolerated but progressed on sorafenib, Child-Pugh class A Regorafenib 160 mg daily versus placebo 10.6 vs. 7.8 mo 0.63 (0.50–0.79)
  Cabozantinib (CELESTIAL) [209] HCC patients who received previous treatment with sorafenib and had had disease progression after at least one systemic treatment (received up to 2), Child-Pugh class A, ECOG PS 0-1 Cabozantinib 60 mg daily versus placebo 10.2 vs. 8.0 mo 0.76 (0.63–0.92)
  Ramucirumab (REACH-2) [210] BCLC stage B or C, Child-Pugh class A, ECOG PS 0-1, AFP ≥400 ng/mL Ramucirumab 8 mg/kg versus placebo 8.5 vs. 7.3 mo 0.710 (0.531–0.949)
  Pembrolizumab (CheckMate 240) [211] HCC patients who progressed from or intolerable to sorafenib, BCLC stage B or C, Child-Pugh class A, ECOG 0-1 Pembrolizumab 200 mg every 3 wk versus placebo 13.9 vs. 10.6 mo 0.77 (0.62–0.96)
 Combination therapy
  Nivolumab+Ipilimumab (CheckMate 040) [204] Advanced HCC patients previously treated with sorafenib, Child-Pugh class A, ECOG PS 0-1 Nivolumab 1 mg/kg plus ipilimumab 3 mg/kg followed by nivolumab 240 mg every 2 wk (arm A); nivolumab 3 mg/ kg plus ipilimumab 1 mg/kg followed by nivolumab 240 mg every 2 wk (arm B); or nivolumab 3 mg/kg every 2 wk plus ipilimumab 1 mg/kg every 6 wk (arm C) 22.8 mo (arm A) vs. 12.5 mo (arm B) vs. 12.7 mo (arm C) -
 Adjuvant therapy
  Atezolizumab+bevacizumab (IMbrave050) [163] High-risk surgically resected or ablated HCC Atezolizumab 1,200 mg plus bevacizumab 15 mg/kg every 3 wk versus active surveillance (adjuvant therapy) - 0.72 (0.53–0.98) for recurrence free survival*

AFP, alpha-fetoprotein; BCLC, Barcelona Clinic Liver Cancer; CI, confidence interval; ICI, immune checkpoint inhibitor; HCC, hepatocellular carcinoma; NE, not evaluated; RCT, randomized controlled trial; TKI, tyrosine kinase inhibitors.

*

Needs to be interpreted with caution as updated efficacy and safety data does not demonstrate consistent results with longer follow-up. [162]