ArticlePDF Available

Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction

Authors:

Abstract

Hepatocellular carcinoma (HCC) is one of the most prevalent cancers worldwide, and due to substantial morbidity and mortality, has proven a significant global health-economic burden. Treatment options are broad and include surgical approaches (i.e., transplantation and resection), radiological (i.e., percutaneous ablation, and trans arterial approaches) and systemic therapies, though treatment response often remains poor. As such, clinical decision making requires a multidisciplinary approach to improve treatment strategy after consideration of the patient's tumor stage, liver function, and performance status. Current systemic cytotoxic therapies for non-surgical candidates have largely remained unchanged over the last decade. Systemic therapies have extended life expectancy by up to 3 months but not without potential notable adverse effects that often limit their use. However, even if patients have necessary access to best treatment, survival outcomes remain concerningly poor. Improved understanding of the pathogenic role of advanced liver fibrosis and wider cancer biology is spearheading the development of targeted immunogenic therapies that appear to offer real promise. Importantly, limiting progression to cirrhosis and early detection of HCC in at risk groups, alongside best use of currently accessible therapeutic options, remains key across global healthcare systems. The focus of this review is to critically assess all current published literature, encapsulating the prevalence, diagnosis, and management of HCC, whilst looking ahead to the potential future therapeutic directions in HCC management.
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
1 Volume 8; Issue 01
Review Article
Incidence, Diagnosis, and Management of
Hepatocellular Carcinoma: Current Perspectives
and Future Direction
Balasubramaniyan Vairappan1*, Gavin Wright2, T.S. Ravikumar3
1Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research,
Pondicherry-605006, India
2Mid and South Essex NHS Foundation Trust, Basildon & Thurrock University Hospitals NHS Foundation Trust, UK
3AIIMS, Mangalagiri, Andhra Pradesh, India
*Corresponding author: Balasubramaniyan Vairappan, Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal
Institute of Postgraduate Medical Education and Research, Pondicherry-605006, India.
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma:
Current Perspectives and Future Direction. J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
Received Date: 11 February 2023; Accepted Date: 21 February 2023; Published Date: 24 February 2023
Journal of Digestive Diseases and Hepatology
Vairappan B, et al. J Dig Dis Hepatol 8: 189.
www.doi.org/10.29011/2574-3511.100089
www.gavinpublishers.com
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent cancers worldwide, and due to substantial morbidity and
mortality, has proven a signicant global health-economic burden. Treatment options are broad and include surgical approaches
(i.e., transplantation and resection), radiological (i.e., percutaneous ablation, and trans arterial approaches) and systemic
therapies, though treatment response often remains poor. As such, clinical decision making requires a multidisciplinary approach
to improve treatment strategy after consideration of the patient’s tumor stage, liver function, and performance status. Current
systemic cytotoxic therapies for non-surgical candidates have largely remained unchanged over the last decade. Systemic
therapies have extended life expectancy by up to 3 months but not without potential notable adverse effects that often limit their
use. However, even if patients have necessary access to best treatment, survival outcomes remain concerningly poor. Improved
understanding of the pathogenic role of advanced liver brosis and wider cancer biology is spearheading the development
of targeted immunogenic therapies that appear to offer real promise. Importantly, limiting progression to cirrhosis and early
detection of HCC in at risk groups, alongside best use of currently accessible therapeutic options, remains key across global
healthcare systems. The focus of this review is to critically assess all current published literature, encapsulating the prevalence,
diagnosis, and management of HCC, whilst looking ahead to the potential future therapeutic directions in HCC management.
Keywords: Diagnosis; Imaging; Liver Cancer; Management;
Surveillance; Systemic Therapy.
Abbreviations: AASLD: American Association for the Study
of Liver Disease; AFP: Alpha-Fetoprotein; APASL: Asia-Pacic
Association for the Study of the Liver; BCLC: Barcelona Clinic
Liver Cancer; CT: Computed Tomography; EASL: European
Association for the Study of the Liver; ECOG: The Eastern
Cooperative Oncology Group; HCC: Hepatocellular Carcinoma;
HBV: Hepatitis B Virus; HCV: Hepatitis C Virus; LT: Liver
Transplantation; NAFLD: Non-Alcoholic Fatty Liver Disease;
NASH: Non-Alcoholic Steatohepatitis; MELD: Model for End-
Stage Liver Disease; MRI: Magnetic Resonance Imaging; TACE:
Transarterial Chemoembolization; US: Ultrasound; WHO: World
Health Organization
Introduction
Hepatocellular carcinoma (HCC) is one of the most lethal
malignancies (~830000 deaths per year) and is an important
medical problem globally. It is ranked as the fth most common
neoplasm and the second leading cause of cancer-related
mortality, with a relative ve-year survival rate of ~ 18% [1-4].
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
2Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
In Asian countries, the prevalence accounts for nearly 75-80 %
of primary liver cancers [5]. In 2020, nearly one million people
were diagnosed with liver cancer worldwide, the most dominant
form of which reported was HCC [4]. Moreover, the World Health
Organization (WHO) estimates that more than one million HCC
patients will die in 2030 [6]. The burden of HCC varies according
to demographic factors (age, gender, race/ethnicity), rarely occurs
in people before age 40, increases more than 55 years, and reaches
a peak at 70 years. Furthermore, the incidence rates among men are
three times as high as the rates among women [7]. Consequently,
due to its high prevalence, not many signicant therapeutic options
are available for advanced HCC. Therefore, improving the early
detection and prognostication of HCC patients is imperative.
Incidence
Most cases of HCC arise from cirrhosis and additional
comorbidities, and its incidence is expected to rise in the future [8,
9]. Hepatitis B virus (HBV) is a DNA virus that promotes mutation
in liver cells by inducing necroinammation and thus causes HCC
and death worldwide (33%). By contrast, hepatitis C virus (HCV)
is an RNA virus that does not integrate into the host genome and
is thus unlikely to be the primary initiator of HCC. About 90%
of HCV-associated liver cancer cases are heralded by cirrhosis,
and the annual incidence rate ranges from 0.5% to 10% [5, 10,
11]. Moreover, chronic heavy alcohol consumption (>3 drinks/
day) was associated with increased HCC risk (~16%); perhaps
no association was noticed with lower levels of consumption
(<3 drinks/day) [12]. Non-alcoholic fatty liver disease (NAFLD)
and its more severe form, non-alcoholic steatohepatitis (NASH),
is emerging as one of the leading HCC risk factors in developed
regions [13, 14]. Recent evidence has emerged that obesity-
mediated chronic inammation was also associated with an
increased risk of HCC [15, 16]. Interestingly, a diabetic individual
with obesity shows an increased risk of liver cancer [1].
Prevention
Cirrhosis and other chronic liver diseases are susceptible to
HCC, consequently, prevention may reduce the population at risk.
Prevention of HCC can be achieved with universal vaccination
against HBV infection [17]. The WHO recommends HBV vaccines
for infants and high-risk groups. A previous study has shown in
Taiwan that Nationwide HBV vaccination to infants resulted in a
36 % reduction in the incidence of HCC compared to unvaccinated
cohorts [18]. Antiviral therapies effectively reduce HCC incidence
in HBV-infected patients (HBsAg-positive), and eliminate HCV
in viremia patients, however, does not eradicate the risk of HCC
in viral hepatitis patients [7]. In this context, it has been shown
that treatment with lamivudine (100mg/day) for 5 years to chronic
HBV patients on the background of cirrhosis reduced the incidence
of HCC risk compared to placebo [19]. Interferon therapy to
HCV patients without cirrhosis had a sustained viral response
and reduced HCC risk by about 75% compared to HCV patients
with cirrhosis who do not have a sustained response to antiviral
therapy [20, 21]. Furthermore, several studies conducted in Japan
and southern Europe have shown that coffee drinking is associated
with a reduced risk of HCC [22]. Albeit the mechanism for this
protective effect remains poorly understood.
Surveillance/Screening
Surveillance and screening of HCC are tremendous
approaches to detect the disease early and reduce mortality. So far,
no high-quality randomized controlled trial has been available for
the surveillance of HCC in cirrhotic patients [2]. However, several
non-randomized studies have reported that HCC patients recruited
into a surveillance programme had a chance of early diagnosis,
more frequent curative therapy and better overall survival than
unrecruited peers [23]. The imaging and serum α fetoprotein
(AFP) measurement are the standard methods for surveillance of
HCC. However, the use of AFP is no longer recommended due
to its inadequate sensitivity (around 60%), specicity (80%) and
predictive value for surveillance testing of HCC. Recently, many
studies have identied a reliable biomarker in diagnosing AFP-
negative HCC and thus ensuring the timely initiation of treatment
(0599a pdf). Ultrasound (US) is the preferred imaging test for
HCC surveillance and has a sensitivity ranging from 60-80%
and a specicity of >90% [24, 25]. However, due to its operator
dependency and unsatisfactory diagnostic accuracy, the use of US
as a surveillance tool in clinical practice is limited [26].
Diagnosis
In general, an accurate and early diagnosis of HCC can
improve the quality of life of HCC patients. Indeed, routinely
followed clinical techniques such as imaging and histology could
only detect late-stage diagnosis of HCC [27]. Globally, AFP is used
as a conventional serum biomarker to detect HCC, albeit its levels
remain normal in 30% of advanced HCC cases [28]. Moreover,
elevated AFP is identied in benign liver diseases such as hepatitis
and cirrhosis [29]. Consequently, the American association for the
study of liver diseases (AASLD) practice guidelines no longer
recommend AFP for the early detection of HCC [10]. Currently,
many clinical and pre-clinical studies are focusing on identifying
a new biomarker for diagnosing AFP-negative HCC, which may
ensure the timely initiation of treatment. A large-scale multicenter
study shows that serum DKK1, a Wnt/β-catenin signaling pathway
inhibitor, could complement the diagnostic accuracy of AFP and
improve the identication of patients with AFP-negative HCC and
HCC from other chronic liver diseases [30]. In cirrhotic patients,
HCC can be diagnosed based on validated imaging techniques or
tissue biopsy. Multiphasic computed tomography (CT) or MRI
is the commonly used imaging technique for HCC diagnosis if
the diameter of the nodule is>1cm. However, these modalities
represent a major clinical challenge if the nodule diameter is <1cm
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
3Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
[8, 2, 4]. Biopsy in advanced liver disease is safe and overcomes
the limitations of non-invasive criteria since diagnostic certainty is
needed to ensure the appropriate use of systemic therapy [31, 4].
Childs et al. also conrmed that biopsy in advanced liver disease
predicts positivity in ~ 91% of HCC cases which proves about
9% of patients would receive inappropriate therapy in the absence
of biopsy [31, 4]. However, the sensitivity of these noninvasive
criteria is only 33% since a negative biopsy does not rule out
HCC [32]. According to the AASLD and EASL guidelines, CD34,
cytokeratin (CK) 7&19, GS, HSP70 and glypican 3 staining to
improve diagnostic accuracy. EASL guidelines also supplement
gene expression proles of glypican 3 and survivin for HCC
diagnosis. Arginase is a hepatocellular differentiation marker
shown to differentiate less well-differentiated tumor from other
liver tumors [4].
Clinical and biochemical markers
Vascular endothelial growth factor (VEGF) A and
Angiotensin II are biomarkers of angiogenesis and have been
associated with poor prognosis in HCC, albeit these markers
failed to predict response to treatment [5, 11, 4]. Furthermore,
biomarkers to predict treatment outcomes are lacking in HCC
patients undergoing immunotherapy. Of note, an inammatory
marker such as CRAFITY (CRP and AFP in ImmunoTherapY)
score is associated with survival and radiological response in
HCC patients receiving anti-programmed death (ligand) (PD-
L)1 immunotherapy but requires prospective validation [33].
The other inammatory marker, neutrophil to lymphocyte ratio
(NLR), is considered a prognostic predictor of HCC patients
undergoing transarterial chemoembolization (TACE). Heat shock
protein 90 alpha (Hsp90α), a molecular chaperone, is increased
in HCC patients and positively correlated with tumor malignancy
[34]. Compelling evidence indicates that micro RNAs (MiRs)
are aberrantly expressed in HCC. In particular, MiRs are highly
stable in circulation and can be used as a biomarker to test early-
stage HCC [35]. In addition, osteopontin, glypican-3 and protein
induced by vitamin K deciency or antagonist-II (PIVKA-II),
also known as Des-γ -carboxy-prothrombin (DCP), have been
identied as serum biomarkers for early detection of HCC [36,
37]. Recently, we identied in HCC patients that tight junction
protein zonula occludens (ZO) 1 blood levels were elevated and
correlated well with serum hsCRP levels [38]. A recent study has
shown that lens culinary agglutinin-reactive fraction of fetoprotein
(AFP-L3), a subtype of AFP, is derived from cancerous hepatocytes
used to diagnose early HCC [39]. However, AFP-L3 has not been
recognized as a conventional diagnostic indicator of HCC. Saad et
al. reported in HCV-related HCC patients (n=30) that serum levels
of annexin A4 (ANXA4) might be a promising biomarker for the
early diagnosis of HCC [40].
Prognosis assessment of HCC
Prognostic prediction is central in the management of HCC. For
HCC patients with concurrent liver disease, the benets of treating
the tumor must be balanced against the potential harms of medical
interventions already recommended to cirrhotic patients [2]. Thus,
the complexity of managing HCC appeals for a multidisciplinary
approach with expertise in hepatology, hepatobiliary surgery,
radiology, pathology, oncology, and specialized nursing [10, 11,
2]. The prognostic assessment incorporated several measures,
which include tumor burden (quantied based on the number and
size), presence of macrovascular invasion, extrahepatic metastasis,
degree of hepatic dysfunction (assessed by Child-Turcotte-Pugh
score, MELD score, ascites, portal hypertension, albumin and
bilirubin), and the Eastern cooperative oncology group (ECOG)
performance status [11, 3]. Among the serological markers, elevated
AFP level was correlated with poor prognosis and associated
with the risk of tumor reoccurrence after surgical resection and
liver transplant. Furthermore, a high DNA copy number of HBV
was associated with poor prognosis and tumor reoccurrence [11,
3]. Several staging systems have been developed to assess the
prognosis of HCC patients. The Barcelona-Clinic Liver Cancer
(BCLC) staging system has been extensively validated and is the
most widely applied staging system for HCC [5, 11, 2]. The other
externally evaluated staging systems are the Cancer of the Liver
Italian Program (CLIP), the French classication, Japan Integrated
Staging (JIS), tumor, node, metastasis (TNM), the Hong-Kong
Liver Cancer (HKLC) staging system, the Chinese University
Prognostic Index (CUIP) and the Taipei integrated scoring system.
According to the BCLC algorithm, HCC patients can be classied
into ve clinical stages, 0, A, B, C, and D, for a better treatment
approach (Figure 1) [5, 8, 11, 2, 4].
BCLC 0: a very early stage of HCC with solitary nodule ≤ 2 cm
without vascular invasion, Child-Pugh A, ECOG-PS 0.
BCLC A: early-stage HCC with solitary (>2 cm) or 2-3 nodules,
all ≤ 3 cm, Child-Pugh A-B, ECOG-PS 0.
BCLC B: intermediate stage HCC with multinodular unresectable
(>3 nodules or ≥2 nodules if any > 3cm), Child-Pugh A-B, ECOG-
PS 0.
BCLC C: advanced HCC with symptomatic tumor, unresectable,
segmental, or portal vein invasion, extrahepatic metastasis, Child-
Pugh A-B, ECOG-PS 1-2.
BCLC D: end-stage liver function with non-transplantable HCC,
Child-Pugh C, ECOG-PS 3-4.
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
4Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
Figure 1: BCLC staging and treatment approach. According to the BCLC system, HCC can be categorized into ve different stages
of prognosis that are concurrent to rst-line treatment recommendation. Indeed, to achieve the best clinical outcome, multidisciplinary
team should meet up and carefully discuss the treatment plans. End-stage liver cirrhotic patients should be considered for LT due to
precipitated liver function (high MELD and Child-Pugh class C or early stages with predictor of poor prognosis) [4,8,11]. Sorafenib
followed by regorafenib as second-line therapy are effective in HCC patients. Lenvatinib has been shown to be non-inferior to sorafenib,
however no second line therapy has been developed [4,8,11]. Moreover, Cabozantinib has been shown to be effective than placebo in
2nd and 3rd line with an improvement of OS [4,8,11]. Note: ECOG PS- Eastern cooperative oncology group performance status; HCC-
hepatocellular carcinoma; LT- liver transplantation; OS- overall survival. 1st line treatment: Sorafenib and Levatinib; 2nd line treatment:
Regorafenib, Cabozantinib and Ramucirumab.
Clinical Management
Several randomized controlled trials and cohort studies have
revealed that numerous therapeutic approaches have exhibited
survival benets for HCC [5, 8, 11, 2, 4]. It arises from multiple
etiologies, and almost 80-90% of HCC cases have underlying
cirrhosis; therefore, the therapeutic option is limited due to the
overall health status of the patients. The treatment protocol for
HCC has been based on the BCLC algorithm, underlying disease
severity, and expected benets of the major intervention [5, 8,
11, 2, 4]. In principle, asymptomatic patients with low tumor
burden and well-preserved liver function (BCLC stage 0/A) are
assigned to be treated with local curative treatments (resection,
ablation, or transplantation, depending upon the presence of portal
hypertension, number of nodules, and liver function). Similarly,
asymptomatic patients with multiple nodules and adequate
liver function (BCLC stage B) are recommended to receive
chemoembolization. In contrast, patients with portal thrombosis or
extrahepatic metastasis (BCLC stage C) are allocated to treatment
with rst and second-line systemic chemotherapies [5, 8, 11, 2,
4]. HCC patients at their terminal stage (stage D) received the
best supportive care and an estimated survival time of only three
months [2].
Surgical resection
The ideal candidates for hepatic resection are patients
without cirrhosis and an early-stage HCC (BCLC stage 0 or A),
irrespective of tumor size and well-preserved liver function. In HCC
patients with cirrhosis, hepatic resection is restricted with a single
nodule (regardless of size), Child-Pugh A with total bilirubin <1
mg/dl, absence of clinically relevant portal hypertension (without
ascites and varices), and ECOG score 0. For these patients, hepatic
resection is associated with a 5-year survival of 70% with low
postoperative mortality (<3%) [5, 10, 11, 2, 3]. However, many
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
5Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
of those 70% of patients have tumor recurrence at ve years
because the underlying chronic liver disease puts the patient at
risk of developing new HCC [41]. Surprisingly, there is no data
on adjuvant therapies to reduce recurrence in HCC patients [42].
In Asian countries and in USA, <5% of patients are candidates
for surgical resection while in Asia, a greater number of young
people with HBV-related HCC with minimal or no cirrhosis [7]. In
the setting of HCV cirrhosis, about 75-80% of patients experience
tumor recurrence following 5 years of resection [43].
Liver transplantation
Liver transplantation (LT) is recommended in HCC patients
with a limited tumor burden (the Milan Criteria – single nodule
≤ 5 cm or 2-3 nodules ≤ 3cm without vascular invasion) and not
on the hepatic resection list. LT has shown excellent survival
outcomes with a 5-year survival of 70% and 10-year survival of
50% with only a 10-15% recurrence rate at 5 years [5, 10, 11].
Indeed, the long-term outcome of LT has shown to be superior
to hepatic resection, which has a recurrence rate of 70% and a
10-year survival between 7-15% [5, 11, 3, 4]. Living donor LT is
an alternative elective procedure that can eliminate dropout and
enable LT in patients with HCC beyond the Milan criteria [43]. The
calcineurin inhibitors (CNIs) such as cyclosporine and tacrolimus
continue to remain potent immunosuppressants used in post LT
[44] and have been shown to promote HCC progression through
nonimmunologic mechanisms [43]. Moreover, the uncontrolled
clinical study revealed that sirolimus, an alternative to tacrolimus,
delay the appearance and retard the progression of recurrent HCC
[45].
Non-surgical treatment
HCC patients with different tumor characteristics or with
cirrhosis are not an ideal candidate for resection. Consequently,
non-surgical procedures such as radiofrequency ablation (RFA),
microwave ablations (MWA), percutaneous ethanol injection (PEI)
and transarterial chemo embolization (TACE) have well proven
anti-tumor effects. Image-guided ablation is restricted for small
nodules < 2 cm as rst-line treatment or as an alternative to hepatic
resection for early-stage single nodules ≤ 4 cm or 2-3 nodules
3 cm [46, 43, 5, 11, 2-4]. For PEI, nodule diameter < 2 cm is still
recommended. RFA is used as a rst-line treatment for nodules
< 2 cm or as an alternate surgery for early-stage single nodules
≤ 4 cm or 2-3 nodules ≤ 3 cm [46, 43, 5, 11, 2-4]. Moreover, the
length of the hospital stay was shorter in the RFA group than
in surgical resection. RFA has shown superior to percutaneous
ethanol injection in improving OS. Indeed, RFA treatment has
demonstrated median overall survival of 60 months and a 5-year
recurrence of 50-70%. MWA has shown similar efcacy to RFA,
however, MWA showed a higher complication rate in tumors > 3
cm [46, 43, 5, 11, 2-4]. Eventually, the success rate of both hepatic
resection and PEI depends on careful follow-up and treatment of
new tumors.
Globally, TACE has been recommended as rst-line therapy
for intermediate stage HCC patients (BCLC-B), particularly those
with Child-Pugh A class cirrhosis who do not have extrahepatic
metastasis or vascular invasion [7]. A systematic review of
randomized trials for unresectable HCC showed survival benets
with TACE when compared to conservative treatment [47].
Furthermore, a systematic review of TACE showed an objective
response of 52.5% and the mortality associated was below 1%
[48]. Selective internal radiation therapy (SIRT) with yttrium-90
microspheres has recently been used as palliative treatment for
BCLC stage B HCC patients. Indeed, no phase 3 trials compare
yttrium-90 radiation therapy with TACE or other types of
treatment with respect to survival [49]. Moreover, Vilgrain et al.
showed in a phase 3 trial that SIRT to BCLC stage C HCC patients
did not improve OS as compared with sorafenib and there was
no improvement with a combination of SIRT with sorafenib when
compared to sorafenib alone [50].
Systemic therapy
More than 70% of HCC patients are not amenable to
treatment with LT or locoregional therapies and thus there is a
great need for effective systemic therapies. Systemic therapy is
the preferred treatment modality for advanced HCC patients
(BCLC-C) and intermediate-stage HCC who do not qualify for
local therapies. Over the past three decay, sorafenib was the rst-
line targeted therapy to show efcacy in advanced HCC patients.
Sorafenib hepatocellular carcinoma assessment randomized
protocol (SHARP) investigators study group showed median
overall survival (OS) in the sorafenib arm was10.7-months
compared to 7.9 months in the placebo group, representing a 31%
decrease in the relative risk of death [51]. Of note, recently, Kelley
et al. showed in a COSMIC-312 phase 3 trial, despite the lack of
improvement in OS, cabozantinib plus atezolizumab signicantly
improved progression-free survival and showed increased disease
control and lower primary progression compared with sorafenib
[52]. In addition, Kudo et al. showed non-inferiority of lenvatinib
(13.6 months) versus sorafenib (12.3 months) in terms of OS and
improvement in progression-free survival, time to progression,
and objective response rate thus, lenvatinib was superior to
sorafenib [53]. A recent phase III trial (REFLECT study) conrms
that FDA has approved Lenvatinib as rst-line systemic therapy
to advanced HCC [46, 43, 5, 11, 2-4]. Sunitinib is an oral multi-
tyrosine kinase inhibitor (TKI) approved for treating other cancers
but not recommended for HCC treatment due to safety issues and
futility reasons [54]. Brivanib alaninate, an oral broblast growth
factor (FGF)‐FGF receptor (FGFR) and vascular endothelial
growth factors (VEGFs) TKI used for advanced stage HCC with
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
6Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
the median OS was 10 months and 9.8 months in the rst- and
second-line treated groups, respectively with manageable adverse
events [55].
In addition, three phase III trials showed negative results
for primary endpoints when testing brivanib in the rst line
blinded to sorafenib, [56] in second line blinded to placebo [57]
and in combination with chemoembolization [58]. The other
TKI, linifanib, which targets VEGF and platelet-derived growth
factor (PDGF), and ramucirumab, a monoclonal antibody against
VEGFR2 [59], failed in phase III studies in rst-line and second-
line indications, respectively [60, 61]. Vatalanib, axitinib and
cediranib are new anti-angiogenic agents involved in treating
HCC, but the results are yet to come. Transforming growth factor-
beta (TGFb), c-MET inhibitors, MEK (MAP2K1) inhibitors, and
Janus kinase 2 (JAK2) inhibitors are also at the very early stage of
investigation [62].
Regorafenib, an oral multi-kinase inhibitor used as second-
line therapy, showed OS benets over placebo (10.6 months vs
7.8 months) in HCC patients who tolerated and progressed on
sorafenib in the phase III RESOURCE trial [63]. The FDA and the
European medicines agency (EMA) have approved regorafenib
to HCC patients who have already been treated with sorafenib.
Similarly, cabozantinib (CELESTIAL study) showed superior to
placebo in terms of OS (10.2 months vs 8.0 months) as second-
line therapy [5, 11, 2-4]. Very recently, an updated result of the
combination of atezolizumab + bevacizumab (IMbrave150 study)
showed survival improvement over sorafenib as rst-line therapy
(19.2 months vs 13.4 months) [5, 11, 2-4]. However, these therapies
are associated with weight loss, diarrhea, anorexia, asthenia, hand-
foot reaction, hypertension, and proteinuria. Unfortunately, in the
phase III trial, investigating a new agent or in combination with
sorafenib as rst-line or second-line therapy merely improved
overall survival and failed to demonstrate a 5-year survival benet
[5, 11, 2-4]. Therefore, the discovery of new therapeutic agents
is warranted, considering the survival benet and adverse effects.
Immunotherapy
Immunotherapy has been proven effective and safe and
improves survival rate and tolerable toxicity in HCC patients [64,
65]. The liver is a unique anatomical and immunological organ
capable of producing antigen-specic tolerance and accepting LT.
Therefore, the development of anti-tumor immunity against HCC
is synergistically hindered by the tolerogenic properties of the liver
and the immunosuppressive tumor microenvironment of HCC
[66]. The US food and drug administration (FDA) has approved
several immune checkpoint inhibitors (ICIs) for HCC and other
cancers. These ICIs and inhibitory receptors include programmed
cell death protein-1 (PD-1), or its ligand programmed cell death-
ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-
4), lymphocyte-activation gene 3 (LAG3), B and T lymphocyte
attenuator (BTLA), T cell immunoglobulin and mucin domain
containing-3 (TIM3) and T cell immunoreceptor with Ig and ITIM
domains (TIGIT) [67]. The PD1 inhibitor nivolumab was used as
the second-line therapy following sorafenib treatment to advance
HCC patients. Moreover, many countries have recommended the
PD-1 and PD-L1 inhibitors pembrolizumab and atezolizumab,
respectively, as clinical treatment options for HCC. A clinical
trial of the cytotoxic T-lymphocyte– associated protein 4 (CTLA-
4) blockade tremelimumab showed a partial response rate of
17.6% in HCC patients with HCV [68]. Antiviral treatment shows
improved liver function and histology and reduced HBV-DNA
levels in HCC patients [69]. The primary nucleoside/nucleotide
analogues (NAs), such as lamivudine treatment for HCC patients,
reduced the incidence but did not eliminate early and mid-level
HCC risk [70], however, entecavir therapy showed decreased HCC
risk [71]. Of note, Papatheodoridis etal. showed in Caucasians
with chronic HBV, following 5 years of entecavir or tenofovir
treatment reduced the risk of HCC among persons with cirrhosis,
but the overall risk was higher among cases without cirrhosis [70].
Moreover, numerous ongoing phase III trials exploring immune-
based therapies may begin their role in the management of HCC.
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
7Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
Name of biomarkers Potential Clinical application Validation Method References
Serum biomarker
Glypican 3 Diagnostic and prognostic Prospective, multi-centre phase II study [72]
AFP-L3 Diagnostic marker – AFP negative cases retrospective study [73,74]
Osteopontin Diagnostic and prognostic Prospective, multi-centre study [75]
PIVKAII Diagnostic marker – AFP negative cases large-scale, multicentre study [76, 77]
Golgi protein-73 Diagnostic and prognostic Prospective, single centre [78,74, 79]
Annexin A4 Early diagnosis Cross sectional – single centre [40]
Heat shock protein 90alpha Diagnostic marker Cross sectional multicentre study [80]
MicroRNA-4651 Diagnostic and prognostic - AFB1-positive cases case-control study [81]
miRNA classier (Cmi) a multicentre, retrospective, longitudinal case-
control study [82]
Metabolite biomarker panel Diagnostic potential at-risk populations
A Large-scale, multicentre case-control
study with
AFP false-negative patients
[83]
Dermcidin Diagnostic marker case-control study [84]
Tumor microenvironment
a) Cellular components
Tumor associated
macrophages Prognostic Retrospective, single centre [85]
Tumor-inltrating
lymphocytes Prognostic Meta-analysis [86]
b) Non-cellular components
Vascular endothelial
growth factor
Prognosis -HBV
Therapeutic target
Cross-sectional, single canter [87]
Transforming growth
factor-beta Diagnosis and Prognosis – HBV Cross-sectional, multi-centre [88]
Cancer stem cell marker
EpCAM Therapeutic potential target Retrospective, multi-centre [89]
CD90 Prognostic. Associated to drug resistance Retrospective, single centre [89]
Note: Aatoxin B1 (AFB1); miRNA classier (Cmi) containing seven differentially expressed miRNAs (miR-29a, miR-29c, miR-133a, miR-
143, miR-145, miR-192, and miR-505); Protein Induced by Vitamin K deciency or antagonist-II (PIVKA-II), also known as Des-γ -carboxy-
prothrombin (DCP); A serum metabolite biomarker - phenylalanyl-tryptophan and glycocholate.
Table 1: Biomarkers and their clinical application in HCC.
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
8Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
Name of Drugs Study outcome Clinical trial design References
Systemic therapyFirst-line therapy
Sorafenib Superior to overall survival SHARP NCT00105443
[51,90]
Lenvatinib Lenvatinib was non-inferior to sorafenib in overall
survival in untreated advanced HCC Open-Label, Phase 3 REFLECT Trial NCT01761266 [53,
91, 92]
Sunitinib
Negative outcome for HCC patients
Pronounced toxicities
Randomized PRODIGE 16 trial,
Intervention Model
An Open Label Multi-Centre Phase
2 study
NCT01164202 [93]
NCT00247676 [94]
Brivanib Negative outcome for HCC patients Multi-centre Phase III Study NCT00858871 [58]
Erlotinib Negative outcome for HCC patients Open Label, Non-Randomized NCT00287222 [95]
Linifanib
Negative outcome for HCC patients
Ongoing
An Open-Label, Phase 2 Study
An Open-label, Randomized Phase
3 Study
An Open label Randomized Clinical
Control Trial
NCT00517920
NCT01009593 [60]
NCT05391867
1. Second-line therapy
Cabozantinib Improves overall survival and progression-free
survival Phase III CELESTIAL trial NCT01908426 [96,
97]
Ramucirumab Improves survival benet phase III REACH and REACH-2
randomized trials
NCT01140347 [98,
99]
NCT02435433 [100]
Regorafenib
Improves
overall survival in patients with HCC who had
disease
progression during rst-line treatment with sorafenib
Multicentre Phase III RESORCE trial NCT01774344 [63,
101, 102]
Nivolumab ongoing Phase Ib, Open label NCT01658878
Pembrolizumab ongoing Phase II, Open label NCT02702414
Everolimus did not improve overall survival the EVOLVE-1 randomized clinical
trial NCT01035229 [103]
Surgical treatments
Liver transplantation Increases survival
Non-population based, consecutive
case
series
[8]
Surgical resection Increases survival
Non-population based, consecutive
case
series
[8]
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
9Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
Locoregional treatments
Percutaneous treatment Increases survival
Non-population based, consecutive
case
Series
[8]
Radiofrequency Increases survival Non-blinded, randomised controlled
trial,meta-analysis [8]
Chemoembolization Increases survival
Non-blinded, randomised controlled
trial,
meta-analysis
[8]
Table 2: Molecular targeted therapies for advanced HCC.
Conclusion and Future Perspectives
HCC is a growing health problem, and globally we are
expected to see over one million new cases each year by 2025.
HCC is a complex disease predominantly seen on a background
of advanced liver cirrhosis, a condition already associated with
signicant morbidity and mortality, from associated complications,
with a yearly incidence of HCC development evident in around
1-5% of patients with cirrhosis compounding the problem. In at
risk groups, early detection via a dedicated screening programme
is pivotal and has a profound impact on outcomes. Moreover, in
those later diagnosed with HCC, a multidisciplinary approach with
the necessary full complement of best treatment options, whether
surgical, radiological and/or oncological, ultimately provide best
treatment outcomes. Over the past decade, with the introduction
of global guidelines, HCC cancer networks and the introduction of
systemic therapies like Sorafenib, the clinical management of HCC
has evolved considerably, though ultimately any improvements in
outcomes remained modest. The big challenge regarding advanced
non-surgical approaches to HCC management is identifying novel
combination regimens for greater and continued improvement
in outcome in the front-line setting. Any new therapy has to be
compared to Sorafenib, which represents the gold standard for
systemic therapy in clinical trials and clinical care. However,
there is still the possibility of seeing further improvements with
Sorafenib as part of combination therapy and thus further phase III
trials are urgently needed to evaluate sorafenib as adjuvant therapy
after curative or locoregional therapies. Moreover, additional
second-line therapies are required if sorafenib is unsuccessful in
advanced stage HCC. Future trials involving effective systemic
therapies, especially immunotherapies based on (i.e., checkpoint
inhibitors) should continue to rise along with the pursuit of new
biomarkers that enable personalized and cost-effective therapeutic
stratication and advancement in managing all stages of HCC.
Declaration
We declare that we have no conicts of interest. All the
authors contributed equally to this review.
Consent for publication
All the authors provide consent for publishing the manuscript.
Acknowledgement
The author is grateful for the funding provided by the
Indian Council of Medical Research (ICMR) extramural grant
(No.5/13/83/2020/NCD/III).
References
1. Bosetti C, Turati F, La Vecchia C. Hepatocellular carcinoma
epidemiology. Best Pract Res Clin Gastroenterol. 2014 Oct;28(5):753-
70.
2. Villanueva A. Hepatocellular Carcinoma. N Engl J Med. 2019 Apr
11;380(15):1450-62.
3. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie
S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021 Jan
21;7(1):6.
4. Vogel A MT, Sapisochin G, Salem R, Saborowski, A. Hepatocellular
carcinoma. The Lancet. 2022:1-18.
5. O’mata M CA, Kokudo N, Kudo M et al. Asia–Pacic clinical practice
guidelines on the management of hepatocellular carcinoma: a 2017
update. Hepatol Int. 2017;11:317-70.
6. Organaization. WH. Cancer. 2017.
7. El-Serag HB. Hepatocellular carcinoma. N Engl J Med. 2011 Sep
22;365(12):1118-27.
8. Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet. 2018
Mar 31;391(10127):1301-14.
9. Vitale A, Trevisani F, Farinati F, Cillo U. Treatment of Hepatocellular
Carcinoma in the Precision Medicine Era: From Treatment Stage
Migration to Therapeutic Hierarchy. Hepatology. 2020 Dec;72(6):2206-
18.
10. Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts
LR, et al. AASLD guidelines for the treatment of hepatocellular
carcinoma. Hepatology. 2018 Jan;67(1):358-80.
11. Liver EAftSot. EASL Clinical Practice Guidelines: Management of
hepatocellular carcinoma. Journal of Hepatology. 2018;69:182-236.
12. Turati F, Galeone C, Rota M, Pelucchi C, Negri E, Bagnardi V, et al.
Alcohol and liver cancer: a systematic review and meta-analysis of
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
10 Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
prospective studies. Ann Oncol. 2014 Aug;25(8):1526-35.
13. Dyson J, Jaques B, Chattopadyhay D, Lochan R, Graham J, Das D, et
al. Hepatocellular cancer: the impact of obesity, type 2 diabetes and a
multidisciplinary team. J Hepatol. 2014 Jan;60(1):110-7.
14. Kanwal F, Kramer JR, Duan Z, Yu X, White D, El-Serag HB. Trends
in the Burden of Nonalcoholic Fatty Liver Disease in a United States
Cohort of Veterans. Clin Gastroenterol Hepatol. 2016 Feb;14(2):301-8
e1-2.
15. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS,
Rubinstein L, et al. New guidelines to evaluate the response to
treatment in solid tumors. European Organization for Research and
Treatment of Cancer, National Cancer Institute of the United States,
National Cancer Institute of Canada. J Natl Cancer Inst. 2000 Feb
2;92(3):205-16.
16. Sun B, Karin M. Obesity, inammation, and liver cancer. J Hepatol.
2012 Mar;56(3):704-13.
17. Chang MH, You SL, Chen CJ, Liu CJ, Lai MW, Wu TC, et al. Long-
term Effects of Hepatitis B Immunization of Infants in Preventing Liver
Cancer. Gastroenterology. 2016 Sep;151(3):472-80 e1.
18. Liao SH, Chen CL, Hsu CY, Chien KL, Kao JH, Chen PJ, et al. Long-
term effectiveness of population-wide multifaceted interventions for
hepatocellular carcinoma in Taiwan. J Hepatol. 2021 Jul;75(1):132-41.
19. Liaw YF, Sung JJ, Chow WC, Farrell G, Lee CZ, Yuen H, et al.
Lamivudine for patients with chronic hepatitis B and advanced liver
disease. N Engl J Med. 2004 Oct 7;351(15):1521-31.
20. Di Bisceglie AM, Shiffman ML, Everson GT, Lindsay KL, Everhart JE,
Wright EC, et al. Prolonged therapy of advanced chronic hepatitis C
with low-dose peginterferon. N Engl J Med. 2008 Dec 4;359(23):2429-
41.
21. Singal AG, Volk ML, Jensen D, Di Bisceglie AM, Schoenfeld PS. A
sustained viral response is associated with reduced liver-related
morbidity and mortality in patients with hepatitis C virus. Clin
Gastroenterol Hepatol. 2010 Mar;8(3):280-8, 88 e1.
22. Larsson SC, Wolk A. Coffee consumption and risk of liver cancer: a
meta-analysis. Gastroenterology. 2007 May;132(5):1740-5.
23. Sherman M. Surveillance for hepatocellular carcinoma. Best Pract
Res Clin Gastroenterol. 2014 Oct;28(5):783-93.
24. Bolondi L. Screening for hepatocellular carcinoma in cirrhosis. J
Hepatol. 2003 Dec;39(6):1076-84.
25. 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 Jul;30(1):37-47.
26. Singal AG, Nehra M, Adams-Huet B, Yopp AC, Tiro JA, Marrero JA,
et al. Detection of hepatocellular carcinoma at advanced stages
among patients in the HALT-C trial: where did surveillance fail? Am J
Gastroenterol. 2013 Mar;108(3):425-32.
27. Luo P, Wu S, Yu Y, Ming X, Li S, Zuo X, et al. Current Status and
Perspective Biomarkers in AFP Negative HCC: Towards Screening for
and Diagnosing Hepatocellular Carcinoma at an Earlier Stage. Pathol
Oncol Res. 2020 Apr;26(2):599-603.
28. Han LL, Lv Y, Guo H, Ruan ZP, Nan KJ. Implications of biomarkers in
human hepatocellular carcinoma pathogenesis and therapy. World J
Gastroenterol. 2014 Aug 14;20(30):10249-61.
29. Chen S, Chen H, Gao S, Qiu S, Zhou H, Yu M, et al. Differential
expression of plasma microRNA-125b in hepatitis B virus-related
liver diseases and diagnostic potential for hepatitis B virus-induced
hepatocellular carcinoma. Hepatol Res. 2017 Mar;47(4):312-20.
30. Shen Q, Fan J, Yang XR, Tan Y, Zhao W, Xu Y, et al. Serum DKK1 as
a protein biomarker for the diagnosis of hepatocellular carcinoma: a
large-scale, multicentre study. Lancet Oncol. 2012 Aug;13(8):817-26.
31. 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 Nov;125(10):1350-55.
32. Forner A, Vilana R, Ayuso C, Bianchi L, Sole M, Ayuso JR, et al.
Diagnosis of hepatic nodules 20 mm or smaller in cirrhosis: Prospective
validation of the noninvasive diagnostic criteria for hepatocellular
carcinoma. Hepatology. 2008 Jan;47(1):97-104.
33. Scheiner B, Pomej K, Kirstein MM, Hucke F, Finkelmeier F, Waidmann
O, et al. Prognosis of patients with hepatocellular carcinoma treated
with immunotherapy - development and validation of the CRAFITY
score. J Hepatol. 2022 Feb;76(2):353-63.
34. Wang X, Song X, Zhuo W, Fu Y, Shi H, Liang Y, et al. The regulatory
mechanism of Hsp90alpha secretion and its function in tumor
malignancy. Proc Natl Acad Sci U S A. 2009 Dec 15;106(50):21288-
93.
35. Lyra-Gonzalez I, Flores-Fong LE, Gonzalez-Garcia I, Medina-Preciado
D, Armendariz-Borunda J. MicroRNAs dysregulation in hepatocellular
carcinoma: Insights in genomic medicine. World J Hepatol. 2015 Jun
18;7(11):1530-40.
36. Bertino G, Ardiri A, Malaguarnera M, Malaguarnera G, Bertino N,
Calvagno GS. Hepatocellualar carcinoma serum markers. Semin
Oncol. 2012 Aug;39(4):410-33.
37. Debes JD RP, Prieto J, Arrese M, Mattos AZ, Boonstra A. Serum
biomarkers for the prediction of hepatocellular carcinoma. Cancers.
2021;13:1681.
38. Ram AK, Pottakat B, Vairappan B. Increased systemic zonula
occludens 1 associated with inammation and independent biomarker
in patients with hepatocellular carcinoma. BMC Cancer. 2018 May
18;18(1):572.
39. Zhou JM, Wang T, Zhang KH. AFP-L3 for the diagnosis of early
hepatocellular carcinoma: A meta-analysis. Medicine (Baltimore).
2021 Oct 29;100(43):e27673.
40. Saad ZM, Fouad Y, Ali LH, Hassanin TM. Clinical Signicance of
Annexin A4 as a Biomarker in the Early Diagnosis of Hepatocellular
Carcinoma. Asian Pac J Cancer Prev. 2020 Sep 1;21(9):2661-65.
41. Ishizawa T, Hasegawa K, Aoki T, Takahashi M, Inoue Y, Sano K,
et al. Neither multiple tumors nor portal hypertension are surgical
contraindications for hepatocellular carcinoma. Gastroenterology.
2008 Jun;134(7):1908-16.
42. Bruix J, Takayama T, Mazzaferro V, Chau GY, Yang J, Kudo M, et
al. Adjuvant sorafenib for hepatocellular carcinoma after resection or
ablation (STORM): a phase 3, randomised, double-blind, placebo-
controlled trial. Lancet Oncol. 2015 Oct;16(13):1344-54.
43. Schwartz M, Roayaie S, Konstadoulakis M. Strategies for the
management of hepatocellular carcinoma. Nat Clin Pract Oncol. 2007
Jul;4(7):424-32.
44. Saliba F, Duvoux C, Dharancy S, Dumortier J, Calmus Y, Gugenheim
J, et al. Five-year outcomes in liver transplant patients receiving
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
11 Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
everolimus with or without a calcineurin inhibitor: Results from the
CERTITUDE study. Liver Int. 2022 Aug 13.
45. Kneteman NM, Oberholzer J, Al Saghier M, Meeberg GA, Blitz M, Ma
MM, et al. Sirolimus-based immunosuppression for liver transplantation
in the presence of extended criteria for hepatocellular carcinoma. Liver
Transpl. 2004 Oct;10(10):1301-11.
46. Burrel M, Llovet JM, Ayuso C, Iglesias C, Sala M, Miquel R, et al.
MRI angiography is superior to helical CT for detection of HCC prior
to liver transplantation: an explant correlation. Hepatology. 2003
Oct;38(4):1034-42.
47. Llovet JM, Bruix J. Systematic review of randomized trials for
unresectable hepatocellular carcinoma: Chemoembolization improves
survival. Hepatology. 2003 Feb;37(2):429-42.
48. Lencioni R, de Baere T, Soulen MC, Rilling WS, Geschwind JF.
Lipiodol transarterial chemoembolization for hepatocellular carcinoma:
A systematic review of efcacy and safety data. Hepatology. 2016
Jul;64(1):106-16.
49. Salem R, Gordon AC, Mouli S, Hickey R, Kallini J, Gabr A, et al.
Y90 Radioembolization Signicantly Prolongs Time to Progression
Compared With Chemoembolization in Patients With Hepatocellular
Carcinoma. Gastroenterology. 2016 Dec;151(6):1155-63 e2.
50. Vilgrain V, Pereira H, Assenat E, Guiu B, Ilonca AD, Pageaux GP,
et al. Efcacy and safety of selective internal radiotherapy with
yttrium-90 resin microspheres compared with sorafenib in locally
advanced and inoperable hepatocellular carcinoma (SARAH): an
open-label randomised controlled phase 3 trial. Lancet Oncol. 2017
Dec;18(12):1624-36.
51. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al.
Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008
Jul 24;359(4):378-90.
52. Kelley RK, Rimassa L, Cheng AL, Kaseb A, Qin S, Zhu AX, et al.
Cabozantinib plus atezolizumab versus sorafenib for advanced
hepatocellular carcinoma (COSMIC-312): a multicentre, open-label,
randomised, phase 3 trial. Lancet Oncol. 2022 Aug;23(8):995-1008.
53. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib
versus sorafenib in rst-line treatment of patients with unresectable
hepatocellular carcinoma: a randomised phase 3 non-inferiority trial.
Lancet. 2018 Mar 24;391(10126):1163-73.
54. Cheng AL, Kang YK, Lin DY, Park JW, Kudo M, Qin S, et al. Sunitinib
versus sorafenib in advanced hepatocellular cancer: results of a
randomized phase III trial. J Clin Oncol. 2013 Nov 10;31(32):4067-75.
55. Park JH, McMillan DC, Powell AG, Richards CH, Horgan PG, Edwards
J, et al. Evaluation of a tumor microenvironment-based prognostic
score in primary operable colorectal cancer. Clin Cancer Res. 2015
Feb 15;21(4):882-8.
56. Johnson PJ, Qin S, Park JW, Poon RT, Raoul JL, Philip PA, et al.
Brivanib versus sorafenib as rst-line therapy in patients with
unresectable, advanced hepatocellular carcinoma: results from
the randomized phase III BRISK-FL study. J Clin Oncol. 2013 Oct
1;31(28):3517-24.
57. Llovet JM, Decaens T, Raoul JL, Boucher E, Kudo M, Chang C, et
al. Brivanib in patients with advanced hepatocellular carcinoma who
were intolerant to sorafenib or for whom sorafenib failed: results from
the randomized phase III BRISK-PS study. J Clin Oncol. 2013 Oct
1;31(28):3509-16.
58. Kudo M, Han G, Finn RS, Poon RT, Blanc JF, Yan L, et al. Brivanib as
adjuvant therapy to transarterial chemoembolization in patients with
hepatocellular carcinoma: A randomized phase III trial. Hepatology.
2014 Nov;60(5):1697-707.
59. Spratlin JL, Cohen RB, Eadens M, Gore L, Camidge DR, Diab S, et
al. Phase I pharmacologic and biologic study of ramucirumab (IMC-
1121B), a fully human immunoglobulin G1 monoclonal antibody
targeting the vascular endothelial growth factor receptor-2. J Clin
Oncol. 2010 Feb 10;28(5):780-7.
60. Cainap C, Qin S, Huang WT, Chung IJ, Pan H, Cheng Y, et al.
Linifanib versus Sorafenib in patients with advanced hepatocellular
carcinoma: results of a randomized phase III trial. J Clin Oncol. 2015
Jan 10;33(2):172-9.
61. Zhu AX, Park JO, Ryoo BY, Yen CJ, Poon R, Pastorelli D, et al.
Ramucirumab versus placebo as second-line treatment in patients
with advanced hepatocellular carcinoma following rst-line therapy
with sorafenib (REACH): a randomised, double-blind, multicentre,
phase 3 trial. Lancet Oncol. 2015 Jul;16(7):859-70.
62. Villanueva A, Llovet JM. Targeted therapies for hepatocellular
carcinoma. Gastroenterology. 2011 May;140(5):1410-26.
63. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al.
Regorafenib for patients with hepatocellular carcinoma who progressed
on sorafenib treatment (RESORCE): a randomised, double-blind,
placebo-controlled, phase 3 trial. Lancet. 2017 Jan 7;389(10064):56-
66.
64. Schizas D, Charalampakis N, Kole C, Economopoulou P, Koustas E,
Gkotsis E, et al. Immunotherapy for pancreatic cancer: A 2020 update.
Cancer Treat Rev. 2020 Jun;86:102016.
65. Keilson JM, Knochelmann HM, Paulos CM, Kudchadkar RR, Lowe
MC. The evolving landscape of immunotherapy in solid tumors. J Surg
Oncol. 2021 Mar;123(3):798-806.
66. Liu Z, Liu X, Liang J, Liu Y, Hou X, Zhang M, et al. Immunotherapy
for Hepatocellular Carcinoma: Current Status and Future Prospects.
Front Immunol. 2021;12:765101.
67. He X, Xu C. Immune checkpoint signaling and cancer immunotherapy.
Cell Res. 2020 Aug;30(8):660-69.
68. Sangro B, Gomez-Martin C, de la Mata M, Inarrairaegui M, Garralda E,
Barrera P, et al. A clinical trial of CTLA-4 blockade with tremelimumab
in patients with hepatocellular carcinoma and chronic hepatitis C. J
Hepatol. 2013 Jul;59(1):81-8.
69. Singal AG, El-Serag HB. Hepatocellular Carcinoma From Epidemiology
to Prevention: Translating Knowledge into Practice. Clin Gastroenterol
Hepatol. 2015 Nov;13(12):2140-51.
70. Papatheodoridis GV, Lampertico P, Manolakopoulos S, Lok A.
Incidence of hepatocellular carcinoma in chronic hepatitis B patients
receiving nucleos(t)ide therapy: a systematic review. J Hepatol. 2010
Aug;53(2):348-56.
71. Hosaka T, Suzuki F, Kobayashi M, Seko Y, Kawamura Y, Sezaki H, et
al. Long-term entecavir treatment reduces hepatocellular carcinoma
incidence in patients with hepatitis B virus infection. Hepatology. 2013
Jul;58(1):98-107.
72. Sawada Y, Yoshikawa T, Ofuji K, Yoshimura M, Tsuchiya N, Takahashi
M, et al. Phase II study of the GPC3-derived peptide vaccine
as an adjuvant therapy for hepatocellular carcinoma patients.
Oncoimmunology. 2016 May;5(5):e1129483.
73. Toyoda H, Kumada T, Tada T, Kaneoka Y, Maeda A, Kanke F, et al.
Citation: Vairappan B, Wright G, Ravikumar TS (2023) Incidence, Diagnosis, and Management of Hepatocellular Carcinoma: Current Perspectives and Future Direction.
J Dig Dis Hepatol 8: 188. DOI: 10.29011/2574-3511.100088
12 Volume 8; Issue 01
J Dig Dis Hepatol, an open access journal
ISSN: 2574-3511
Clinical utility of highly sensitive Lens culinaris agglutinin-reactive
alpha-fetoprotein in hepatocellular carcinoma patients with alpha-
fetoprotein <20 ng/mL. Cancer Sci. 2011 May;102(5):1025-31.
74. Zhang Z, Zhang Y, Wang Y, Xu L, Xu W. Alpha-fetoprotein-L3 and
Golgi protein 73 may serve as candidate biomarkers for diagnosing
alpha-fetoprotein-negative hepatocellular carcinoma. Onco Targets
Ther. 2016;9:123-9.
75. Wan HG, Xu H, Gu YM, Wang H, Xu W, Zu MH. Comparison
osteopontin vs AFP for the diagnosis of HCC: a meta-analysis. Clin
Res Hepatol Gastroenterol. 2014 Dec;38(6):706-14.
76. Ji J, Wang H, Li Y, Zheng L, Yin Y, Zou Z, et al. Diagnostic Evaluation
of Des-Gamma-Carboxy Prothrombin versus alpha-Fetoprotein for
Hepatitis B Virus-Related Hepatocellular Carcinoma in China: A Large-
Scale, Multicentre Study. PLoS One. 2016;11(4):e0153227.
77. Feng H, Li B, Li Z, Wei Q, Ren L. PIVKA-II serves as a potential
biomarker that complements AFP for the diagnosis of hepatocellular
carcinoma. BMC Cancer. 2021 Apr 13;21(1):401.
78. Dai M, Chen X, Liu X, Peng Z, Meng J, Dai S. Diagnostic Value of the
Combination of Golgi Protein 73 and Alpha-Fetoprotein in Hepatocellular
Carcinoma: A Meta-Analysis. PLoS One. 2015;10(10):e0140067.
79. Dong M, Chen ZH, Li X, Li XY, Wen JY, Lin Q, et al. Serum Golgi protein
73 is a prognostic rather than diagnostic marker in hepatocellular
carcinoma. Oncol Lett. 2017 Nov;14(5):6277-84.
80. Fu Y, Xu X, Huang D, Cui D, Liu L, Liu J, et al. Plasma Heat Shock
Protein 90alpha as a Biomarker for the Diagnosis of Liver Cancer:
An Ofcial, Large-scale, and Multicenter Clinical Trial. EBioMedicine.
2017 Oct;24:56-63.
81. Wu XM, Xi ZF, Liao P, Huang HD, Huang XY, Wang C, et al. Diagnostic
and prognostic potential of serum microRNA-4651 for patients with
hepatocellular carcinoma related to aatoxin B1. Oncotarget. 2017 Oct
6;8(46):81235-49.
82. Lin XJ, Chong Y, Guo ZW, Xie C, Yang XJ, Zhang Q, et al. A serum
microRNA classier for early detection of hepatocellular carcinoma: a
multicentre, retrospective, longitudinal biomarker identication study
with a nested case-control study. Lancet Oncol. 2015 Jul;16(7):804-
15.
83. Luo P, Yin P, Hua R, Tan Y, Li Z, Qiu G, et al. A Large-scale, multicenter
serum metabolite biomarker identication study for the early detection
of hepatocellular carcinoma. Hepatology. 2018 Feb;67(2):662-75.
84. Qiu F, Qiu F, Liu L, Liu J, Xu J, Huang X. The Role of Dermcidin in the
Diagnosis and Staging of Hepatocellular Carcinoma. Genet Test Mol
Biomarkers. 2018 Apr;22(4):218-23.
85. Shu QH, Ge YS, Ma HX, Gao XQ, Pan JJ, Liu D, et al. Prognostic value
of polarized macrophages in patients with hepatocellular carcinoma
after curative resection. J Cell Mol Med. 2016 Jun;20(6):1024-35.
86. Yao W, He JC, Yang Y, Wang JM, Qian YW, Yang T, et al. The
Prognostic Value of Tumor-inltrating Lymphocytes in Hepatocellular
Carcinoma: a Systematic Review and Meta-analysis. Sci Rep. 2017
Aug 8;7(1):7525.
87. Mao CS, Yin H, Ning HB, Peng Z, Li K, Ding GQ. Levels of HBx, VEGF,
and CEACAM1 in HBV-related hepatocellular carcinoma and their
correlation with cancer prognosis. Eur Rev Med Pharmacol Sci. 2017
Oct;21(17):3827-33.
88. Dong ZZ, Yao DF, Yao M, Qiu LW, Zong L, Wu W, et al. Clinical impact
of plasma TGF-beta1 and circulating TGF-beta1 mRNA in diagnosis
of hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int. 2008
Jun;7(3):288-95.
89. Yamashita T, Honda M, Nakamoto Y, Baba M, Nio K, Hara Y, et al.
Discrete nature of EpCAM+ and CD90+ cancer stem cells in human
hepatocellular carcinoma. Hepatology. 2013 Apr;57(4):1484-97.
90. Bruix J, Cheng AL, Meinhardt G, Nakajima K, De Sanctis Y, Llovet J.
Prognostic factors and predictors of sorafenib benet in patients with
hepatocellular carcinoma: Analysis of two phase III studies. J Hepatol.
2017 Nov;67(5):999-1008.
91. Briggs A, Daniele B, Dick K, Evans TRJ, Galle PR, Hubner RA, et
al. Covariate-adjusted analysis of the Phase 3 REFLECT study
of lenvatinib versus sorafenib in the treatment of unresectable
hepatocellular carcinoma. Br J Cancer. 2020 Jun;122(12):1754-59.
92. Vogel A, Qin S, Kudo M, Su Y, Hudgens S, Yamashita T, et al. Lenvatinib
versus sorafenib for rst-line treatment of unresectable hepatocellular
carcinoma: patient-reported outcomes from a randomised, open-label,
non-inferiority, phase 3 trial. Lancet Gastroenterol Hepatol. 2021
Aug;6(8):649-58.
93. Turpin A, de Baere T, Heurgue A, Le Malicot K, Ollivier-Hourmand I,
Lecomte T, et al. Liver transarterial chemoembolization and sunitinib
for unresectable hepatocellular carcinoma: Results of the PRODIGE
16 study. Clin Res Hepatol Gastroenterol. 2021 Mar;45(2):101464.
94. Faivre S, Raymond E, Boucher E, Douillard J, Lim HY, Kim JS, et al.
Safety and efcacy of sunitinib in patients with advanced hepatocellular
carcinoma: an open-label, multicentre, phase II study. Lancet Oncol.
2009 Aug;10(8):794-800.
95. Govindarajan R, Siegel E, Makhoul I, Williamson S. Bevacizumab
and erlotinib in previously untreated inoperable and metastatic
hepatocellular carcinoma. Am J Clin Oncol. 2013 Jun;36(3):254-7.
96. Abou-Alfa GK, Borgman-Hagey AE, Kelley RK. Cabozantinib in
Hepatocellular Carcinoma. N Engl J Med. 2018 Oct 4;379(14):1384-
85.
97. Freemantle N, Mollon P, Meyer T, Cheng AL, El-Khoueiry AB, Kelley
RK, et al. Quality of life assessment of cabozantinib in patients with
advanced hepatocellular carcinoma in the CELESTIAL trial. Eur J
Cancer. 2022 Jun;168:91-98.
98. Zhu AX, Baron AD, Malfertheiner P, Kudo M, Kawazoe S, Pezet D, et al.
Ramucirumab as Second-Line Treatment in Patients With Advanced
Hepatocellular Carcinoma: Analysis of REACH Trial Results by Child-
Pugh Score. JAMA Oncol. 2017 Feb 1;3(2):235-43.
99. Llovet JM, Singal AG, Villanueva A, Finn RS, Kudo M, Galle PR, et
al. Prognostic and Predictive Factors in Patients with Advanced
HCC and Elevated Alpha-Fetoprotein Treated with Ramucirumab
in Two Randomized Phase III Trials. Clin Cancer Res. 2022 Jun
1;28(11):2297-305.
100. Zhu AX, Finn RS, Kang YK, Yen CJ, Galle PR, Llovet JM, et al. Serum
alpha-fetoprotein and clinical outcomes in patients with advanced
hepatocellular carcinoma treated with ramucirumab. Br J Cancer.
2021 Apr;124(8):1388-97.
101. Finn RS, Merle P, Granito A, Huang YH, Bodoky G, Pracht M, et
al. Outcomes of sequential treatment with sorafenib followed by
regorafenib for HCC: Additional analyses from the phase III RESORCE
trial. J Hepatol. 2018 Aug;69(2):353-58.
102. Teufel M, Seidel H, Kochert K, Meinhardt G, Finn RS, Llovet JM, et al.
Biomarkers Associated With Response to Regorafenib in Patients With
Hepatocellular Carcinoma. Gastroenterology. 2019 May;156(6):1731-
41.
103. Zhu AX, Kudo M, Assenat E, Cattan S, Kang YK, Lim HY, et al. Effect
of everolimus on survival in advanced hepatocellular carcinoma after
failure of sorafenib: the EVOLVE-1 randomized clinical trial. JAMA.
2014 Jul 2;312(1):57-67.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background The CELESTIAL trial (NCT01908426) demonstrated overall survival benefit for cabozantinib versus placebo in patients with advanced hepatocellular carcinoma (aHCC) who had received prior sorafenib treatment. This analysis of CELESTIAL compared the impact of cabozantinib versus placebo on health-related quality of life (HRQoL). Materials and methods Health status was assessed using the EuroQol five-dimension five-level (EQ-5D-5L) questionnaire over the 800-day follow-up period. EQ-5D-5L health states were mapped to health utility scores using reference values for the UK population. Quality-adjusted life years (QALYs) were calculated for each treatment group as the area under the curve for the plot of health utility score over time. The between-treatment group difference in restricted mean QALYs was calculated by generalized linear models and adjusted for baseline differences. A difference of 0.08 in health utility score (or in QALY) was deemed a minimally important difference and to be clinically significant. Results At week 5, the difference in mean health utility score between cabozantinib and placebo was −0.097 (95% confidence interval [95% CI]: −0.126, −0.067; p ≤ 0.001). Between-group differences in health utility scores diminished over time and were generally non-significant. The cabozantinib group accrued more QALYs than the placebo group over follow-up. Differences in mean QALYs (cabozantinib minus placebo) were statistically and clinically significant, ranging from +0.092 (95% CI: 0.016, 0.169) to +0.185 (95% CI: 0.126, 0.243) in favour of cabozantinib, depending on the reference value set used. Conclusions These HRQoL findings support a positive benefit–risk profile for cabozantinib in previously treated patients with aHCC.
Article
Full-text available
Purpose: Ramucirumab is an effective treatment for patients with advanced HCC (aHCC) and baseline AFP {greater than or equal to}400 ng/mL. We aimed to identify prognostic and predictive factors of response to ramucirumab in patients with aHCC with AFP {greater than or equal to}400 ng/mL from the Phase III REACH and REACH-2 randomized trials. Experimental design: Patients with aHCC, Child-Pugh class A with prior sorafenib treatment were randomized in REACH and REACH-2 (ramucirumab 8 mg/kg or placebo, biweekly). Meta-analysis of individual patient-level data (pooled population) from REACH (AFP {greater than or equal to}400 ng/mL) and REACH-2 was performed. A drug exposure analysis was conducted for those with evaluable pharmacokinetics data. To identify potential prognostic factors for overall survival (OS), multivariate analyzes were performed using a Cox proportional hazard regression model. To define predictors of ramucirumab benefit, subgroup-by-treatment interactions terms were evaluated. Results: Of 542 patients (316 ramucirumab, 226 placebo) analyzed, 8 variables had independent prognostic value associated with poor outcome (geographical region, ECOG PS {greater than or equal to}1, AFP >1000 ng/mL, Child-Pugh >A5, extrahepatic spread, high neutrophil-to-lymphocyte, high alkaline phosphatase and aspartate aminotransferase). Ramucirumab benefit was present across all subgroups, including patients with very aggressive HCC (above median AFP; HR: 0.64; 95%CI:0.49-0.84) and non-viral aHCC (HR: 0.56; 95%CI:0.40-0.79). While no baseline factor was predictive of a differential OS benefit with ramucirumab, analyzes demonstrated an association between high drug exposure, treatment-emergent hypertension (Grade {greater than or equal to}3) and increased ramucirumab benefit. Conclusions: Ramucirumab provided a survival benefit irrespective of baseline prognostic covariates, and this benefit was greatest in patients with high ramucirumab drug exposure and/or those with treatment-related hypertension.
Article
Full-text available
Background: Immunotherapy with atezolizumab plus bevacizumab represents the new standard of care in systemic front-line treatment of hepatocellular carcinoma (HCC). Prognostic biomarkers are an unmet need. Methods: Patients with HCC put on PD-(L)1-based immunotherapy in 6 European centers (training set; n=190) and in 8 European centers (validation set; n=102) were included. We investigated the prognostic value of baseline variables on overall survival by using a Cox model in the training set and developed the easily applicable CRAFITY (CRP and AFP in ImmunoTherapY) score. The score was validated in the independent, external cohort, and evaluated in a cohort of patients treated with sorafenib (n=204). Results: Baseline serum alpha-fetoprotein ≥100 ng/ml (HR, 1.7; p=0.007) and C-reactive protein ≥1 mg/dl (HR, 1.7; p=0.007) were identified as independent prognostic factors in multivariable analysis and were used to develop the CRAFITY score. Patients who fulfilled no criterion (0 points; CRAFITY-low) had the longest median overall survival (27.6 (95%CI, 19.5-35.8) months), followed by those fulfilling one criterion (1 point; CRAFITY-intermediate; 11.3 (95%CI, 8.0-14.6) months), and patients meeting both criteria (2 points; CRAFITY-high; 6.4 (95%CI, 4.8-8.1) months; p<0.001). Additionally, best radiological response (complete response/partial response/stable disease/progressive disease) was significantly better in patients with lower CRAFITY score (CRAFITY-low:9%/20%/52%/20% vs. CRAFITY-intermediate:3%/25%/36%/36% vs. CRAFITY-high:2%/15%/22%/61%; p=0.003). These results were confirmed in the independent validation set as well as in different subgroups including Child-Pugh A and B, performance status 0 and ≥1, and first-line and later lines. In the sorafenib cohort, CRAFITY was associated with survival, but not radiological response. Conclusions: The CRAFITY score is associated with survival and radiological response. The score may help with patient counseling, but requires prospective validation. Lay summary: The immunotherapy based regimen of atezolizumab plus bevacizumab represents the new standard of care in systemic first-line therapy of hepatocellular carcinoma (HCC). Biomarkers to predict treatment outcome are an unmet need in patients undergoing immunotherapy for HCC. We developed and externally validated a score that predicts outcome in patients with HCC undergoing immunotherapy with immune checkpoint blockers.
Article
Full-text available
Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer with poor prognosis. Surgery, chemotherapy, and radiofrequency ablation are three conventional therapeutic options that will help only a limited percentage of HCC patients. Cancer immunotherapy has achieved dramatic advances in recent years and provides new opportunities to treat HCC. However, HCC has various etiologies and can evade the immune system through multiple mechanisms. With the rapid development of genetic engineering and synthetic biology, a variety of novel immunotherapies have been employed to treat advanced HCC, including immune checkpoint inhibitors, adoptive cell therapy, engineered cytokines, and therapeutic cancer vaccines. In this review, we summarize the current landscape and research progress of different immunotherapy strategies in the treatment of HCC. The challenges and opportunities of this research field are also discussed.
Article
Full-text available
Background Advanced hepatocellular carcinoma (HCC) is commonly diagnosed using non-invasive radiological criteria (NIRC) defined by the European Association for the Study of the Liver or the American Association for the Study of Liver Diseases. In 2017, The National Institute for Clinical Excellence mandated histological confirmation of disease to authorise the use of sorafenib in the UK. Methods This was a prospective multicentre audit in which patients suitable for sorafenib were identified at multidisciplinary meetings. The primary analysis cohort (PAC) was defined by the presence of Child-Pugh class A liver disease and performance status 0–2. Clinical, radiological and histological data were reported locally and collected on a standardised case report form. Results Eleven centres reported 418 cases, of which 361 comprised the PAC. Overall, 76% had chronic liver disease and 66% were cirrhotic. The diagnostic imaging was computed tomography in 71%, magnetic resonance imaging in 27% and 2% had both. Pre-existing histology was available in 45 patients and 270 underwent a new biopsy, which confirmed HCC in 93.4%. Alternative histological diagnoses included cholangiocarcinoma (CC) and combined HCC-CC. In cirrhotic patients, NIRC criteria had a sensitivity of 65.4% and a positive predictive value of 91.4% to detect HCC. Two patients (0.7%) experienced mild post-biopsy bleeding. Conclusion The diagnostic biopsy is safe and feasible for most patients eligible for systemic therapy
Article
Full-text available
Background Hepatocellular carcinoma (HCC) is one of the most common malignant tumors of the digestive system and has high morbidity and mortality rates. It is essential to search new biomarkers to improve the accuracy of early HCC diagnosis. Therefore, we evaluated the diagnostic value of prothrombin induced by vitamin K deficiency or antagonist- II (PIVKA-II) as a potential biomarker that complements α-fetoprotein (AFP) in HCC by detecting the serum PIVKA-II levels. Methods Serum PIVKA-II levels were compared in 168 HCC patients, 150 benign liver disease patients and 153 healthy controls to investigate the PIVKA-II potential to be a HCC biomarker. Receiver operating characteristic curve (ROC) analysis was used to evaluate the value of PIVKA-II in the diagnosis of HCC and its complementary role of AFP. The correlation between serum PIVKA-II levels and clinicopathological characteristics was analyzed to study the value of PIVKA-II in assessing HCC progression and prognosis. Finally, the ability of PIVKA-II in assessing the surgical treatment effects of HCC was studied by comparing the pre- and post-operative serum PIVKA-II levels in 89 HCC patients. Results Serum PIVKA-II levels in HCC patients were significantly higher than that in patients with benign liver disease and healthy controls. The PIVKA-II performance in the diagnosing HCC as an individual biomarker was remarkable. The combined detection of PIVKA-II and AFP improved the diagnostic efficiency of HCC. PIVKA-II retained significant diagnosis capabilities for AFP-negative HCC patients. Significant correlations were found between PIVKA-II expression levels and some clinicopathological characteristics, including tumor size, tumor stage, tumor metastasis, differentiation degree and complications. PIVKA-II expression obviously decreased after surgical resection. Conclusions PIVKA-II is a promising serum biomarker for the HCC diagnosis that can be used as a supplement for AFP. The combined diagnosis of the two markers greatly improved the diagnostic efficiency of HCC. The PIVKA-II levels in HCC patients were widely associated with clinicopathological characteristics representing tumor cell dissemination and/or poor prognosis. PIVKA-II can be used to evaluate the curative effects of HCC resection.
Article
Full-text available
Hepatocellular carcinoma (HCC) is a leading cause of global cancer death. Major etiologies of HCC relate to chronic viral infections as well as metabolic conditions. The survival rate of people with HCC is very low and has been attributed to late diagnosis with limited treatment options. Combining ultrasound and the biomarker alpha-fetoprotein (AFP) is currently one of the most widely used screening combinations for HCC. However, the clinical utility of AFP is controversial, and the frequency and operator-dependence of ultrasound lead to a variable degree of sensitivity and specificity across the globe. In this review, we summarize recent developments in the search for non-invasive serum biomarkers for early detection of HCC to improve prognosis and outcome for patients. We focus on tumor-associated protein markers, immune mediators (cytokines and chemokines), and micro-RNAs in serum or circulating extracellular vesicles and examine their potential for clinical application.
Article
Full-text available
Background & Aims Taiwan has launched a series of population-wide interventions on hepatocellular carcinoma (HCC) related to hepatitis B and C virus infection since 1984. It therefore provides a natural opportunity to evaluate whether the reduction of HCC incidence, to a greater extent, and the improvement of case-fatality, to a lesser extent, or vice versa make contribution to the resultant mortality reduction given each intervention program. Methods Population-based registry data on HCC mortality and incidence of individuals aged 0 to 84 years between 1979 and 2016 were collected before (period 1) and after intervention with three periods, universal hepatitis B vaccination from 1984 (period 2), universal health care from 1995 (period 3), and viral hepatitis therapy from 2003 (period 4). A Bayesian Poisson regression model for decomposing mortality rate was developed to estimate respective contributions to the reduction of incidence and case-fatality rate by various age groups. Results The mortality trends of children, young- and middle-aged groups substantially declined but there was only a slight decrease in the elderly group. The declining trends of mortality were in part explained by incidence reduction and in part by a remarkable decline in case-fatality rate attributed to universal health care. Hepatitis B vaccination led to 35.9% (26.8% to 44.4%) reduction of incidence for aged 30 years or below, whereas antiviral therapy made contribution to the reduction of 14.9% (11.8% to 17.9%) and 15.4% (14.1% to 16.6%) for aged 30-49 years and 50-69 years, respectively. Conclusions Vaccination and anti-viral therapy were effective in reducing HCC incidence and mortality for the young and middle-aged groups when the case-fatality was improved due to universal health care for all age groups.
Article
Background Cabozantinib has shown clinical activity in combination with checkpoint inhibitors in solid tumours. The COSMIC-312 trial assessed cabozantinib plus atezolizumab versus sorafenib as first-line systemic treatment for advanced hepatocellular carcinoma. Methods COSMIC-312 is an open-label, randomised, phase 3 trial that enrolled patients aged 18 years or older with advanced hepatocellular carcinoma not amenable to curative or locoregional therapy and previously untreated with systemic anticancer therapy at 178 centres in 32 countries. Patients with fibrolamellar carcinoma, sarcomatoid hepatocellular carcinoma, or combined hepatocellular cholangiocarcinoma were not eligible. Tumours involving major blood vessels, including the main portal vein, were permitted. Patients were required to have measurable disease per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), Barcelona Clinic Liver Cancer stage B or C disease, an Eastern Cooperative Oncology Group performance status of 0 or 1, adequate organ and marrow function, and Child-Pugh class A. Previous resection, tumour ablation, radiotherapy, or arterial chemotherapy was allowed if more than 28 days before randomisation. Patients were randomly assigned (2:1:1) via a web-based interactive response system to cabozantinib 40 mg orally once daily plus atezolizumab 1200 mg intravenously every 3 weeks, sorafenib 400 mg orally twice daily, or single-agent cabozantinib 60 mg orally once daily. Randomisation was stratified by disease aetiology, geographical region, and presence of extrahepatic disease or macrovascular invasion. Dual primary endpoints were progression-free survival per RECIST 1.1 as assessed by a blinded independent radiology committee in the first 372 patients randomly assigned to the combination treatment of cabozantinib plus atezolizumab or sorafenib (progression-free survival intention-to-treat [ITT] population), and overall survival in all patients randomly assigned to cabozantinib plus atezolizumab or sorafenib (ITT population). Final progression-free survival and concurrent interim overall survival analyses are presented. This trial is registered with ClinicalTrials.gov, NCT03755791. Findings Analyses at data cut-off (March 8, 2021) included the first 837 patients randomly assigned between Dec 7, 2018, and Aug 27, 2020, to combination treatment of cabozantinib plus atezolizumab (n=432), sorafenib (n=217), or single-agent cabozantinib (n=188). Median follow-up was 15·8 months (IQR 14·5–17·2) in the progression-free survival ITT population and 13·3 months (10·5–16·0) in the ITT population. Median progression-free survival was 6·8 months (99% CI 5·6–8·3) in the combination treatment group versus 4·2 months (2·8–7·0) in the sorafenib group (hazard ratio [HR] 0·63, 99% CI 0·44–0·91, p=0·0012). Median overall survival (interim analysis) was 15·4 months (96% CI 13·7–17·7) in the combination treatment group versus 15·5 months (12·1–not estimable) in the sorafenib group (HR 0·90, 96% CI 0·69–1·18; p=0·44). The most common grade 3 or 4 adverse events were alanine aminotransferase increase (38 [9%] of 429 patients in the combination treatment group vs six [3%] of 207 in the sorafenib group vs 12 [6%] of 188 in the single-agent cabozantinib group), hypertension (37 [9%] vs 17 [8%] vs 23 [12%]), aspartate aminotransferase increase (37 [9%] vs eight [4%] vs 18 [10%]), and palmar-plantar erythrodysaesthesia (35 [8%] vs 17 [8%] vs 16 [9%]); serious treatment-related adverse events occurred in 78 (18%) patients in the combination treatment group, 16 (8%) patients in the sorafenib group, and 24 (13%) in the single-agent cabozantinib group. Treatment-related grade 5 events occurred in six (1%) patients in the combination treatment group (encephalopathy, hepatic failure, drug-induced liver injury, oesophageal varices haemorrhage, multiple organ dysfunction syndrome, and tumour lysis syndrome), one (<1%) patient in the sorafenib group (general physical health deterioration), and one (<1%) patient in the single-agent cabozantinib group (gastrointestinal haemorrhage). Interpretation Cabozantinib plus atezolizumab might be a treatment option for select patients with advanced hepatocellular carcinoma, but additional studies are needed. Funding Exelixis and Ipsen.
Article
Background Hepatocellular carcinoma is the third-leading cause of cancer-related death worldwide. Preservation of health-related quality of life (HRQOL) during treatment is an important therapeutic goal. The aim of this study was to evaluate the effect of treatment with lenvatinib versus sorafenib on HRQOL. Methods REFLECT was a previously published multicentre, randomised, open-label, non-inferiority phase 3 study comparing the efficacy and safety of lenvatinib versus sorafenib as a first-line systemic treatment for unresectable hepatocellular carcinoma. Eligible patients were aged 18 years or older with unresectable hepatocellular carcinoma and one or more measurable target lesion per modified Response Evaluation Criteria in Solid Tumors criteria, Barcelona Clinic Liver Cancer stage B or C categorisation, Child-Pugh class A, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or lower, and adequate organ function. Patients were randomly assigned (1:1) via an interactive voice–web response system; stratification factors for treatment allocation included region; macroscopic portal vein invasion, extrahepatic spread, or both; ECOG performance status; and bodyweight. Patient-reported outcomes (PROs), collected at baseline, on day 1 of each subsequent cycle, and at the end of treatment, were evaluated in post-hoc analyses of secondary and exploratory endpoints in the analysis population, which was the subpopulation of patients with a PRO assessment at baseline. A linear mixed-effects model evaluated change from baseline in PROs, including European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Core 30 (QLQ-C30) and hepatocellular carcinoma-specific QLQ-HCC18 scales (both secondary endpoints of the REFLECT trial). Time-to-definitive-deterioration analyses were done based on established thresholds for minimum differences for worsening in PROs. Responder analyses explored associations between HRQOL and clinical response. This study is registered with ClinicalTrials.gov, NCT01761266. Findings Of 954 eligible patients randomly assigned to lenvatinib (n=478) or sorafenib (n=476) between March 14, 2013, and July 30, 2015, 931 patients (n=468 for lenvatinib; n=463 for sorafenib) were included in this analysis. Baseline PRO scores reflected impaired HRQOL and functioning and considerable symptom burden relative to full HRQOL. Differences in overall mean change from baseline estimates in most PRO scales generally favoured the lenvatinib over the sorafenib group, although the differences were not nominally statistically or clinically significant. Patients treated with lenvatinib experienced nominally statistically significant delays in definitive, meaningful deterioration on the QLQ-C30 fatigue (hazard ratio [HR] 0·83, 95% CI 0·69–0·99), pain (0·80, 0·66–0·96), and diarrhoea (0·52, 0·42–0·65) domains versus patients treated with sorafenib. Significant differences in time to definitive deterioration were not observed for other QLQ-C30 domains, and there was no difference in time to definitive deterioration on the global health status/QOL score (0·89, 0·73–1·09). For most PRO scales, differences in overall mean change from baseline estimates favoured responders versus non-responders. Across all scales, HRs for time to definitive deterioration were in favour of responders; median time to definitive deterioration for responders exceeded those for non-responders by a range of 4·8 to 14·6 months. Interpretation HRQOL for patients undergoing treatment for unresectable hepatocellular carcinoma is an important therapeutic consideration. The evidence of HRQOL benefits in clinically relevant domains support the use of lenvatinib compared with sorafenib to delay functional deterioration in advanced hepatocellular carcinoma. Funding Eisai and Merck Sharp & Dohme.