Skip to main content
Biologics : Targets & Therapy logoLink to Biologics : Targets & Therapy
. 2012 Jul 17;6:207–219. doi: 10.2147/BTT.S23907

Hepatocellular carcinoma: natural history, current management, and emerging tools

Christopher L Tinkle 1, Daphne Haas-Kogan 1,
PMCID: PMC3421475  PMID: 22904613

Abstract

Hepatocellular carcinoma (HCC) is the most common primary liver tumor and represents the third-leading cause of cancer-related death in the world. The incidence of HCC continues to increase worldwide, with a unique geographic, age, and sex distribution. The most important risk factor associated with HCC is liver cirrhosis, with the majority of cases caused by chronic infection with hepatitis B (HBV) and C (HCV) viruses and alcohol abuse, although nonalcoholic fatty liver disease is emerging as an increasingly important cause. Primary prevention in the form of HBV vaccination has led to a significant decrease in HBV-related HCC, and initiation of antiviral therapy appears to reduce the incidence of HCC in patients with chronic HBV or HCV infection. Additionally, the use of ultrasonography enables the early detection of small liver tumors and forms the backbone of recommended surveillance programs for patients at high risk for the development of HCC. Cross-sectional imaging studies, including computed tomography and magnetic resonance imaging, represent further noninvasive techniques that are increasingly employed to diagnose HCC in patients with cirrhosis. The mainstay of potentially curative therapy includes surgery – either resection or liver transplantation. However, most patients are ineligible for surgery, because of either advanced disease or underlying liver dysfunction, and are managed with locoregional and/or systemic therapies. Randomized controlled trials have demonstrated a survival benefit with both local therapies, either ablation or embolization, and systemic therapy in the form of the multikinase inhibitor sorafenib. Despite this, median survival remains poor and recurrence rates significant. Further advances in our understanding of the molecular pathogenesis of HCC hold promise in improving the diagnosis and treatment of this highly lethal cancer.

Keywords: hepatocellular carcinoma, viral hepatitis, liver transplantation, ablation, embolization, sorafenib

Natural history

Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death worldwide and the leading cause of death in patients with cirrhosis.1,2 Symptoms attributable to HCC are usually absent. Rather, patients typically manifest symptoms related to underlying cirrhosis, a condition present in 80%–90% of patients with HCC.3 Consequently, the majority of patients are diagnosed with advanced disease, often precluding potentially curative therapies. This has resulted, in part, in a 5-year overall survival rate of 12% and a median survival following diagnosis ranging from 6 to 20 months.3,4

When symptomatic, HCC is often associated with nonspecific complaints, including right upper abdominal or epigastric pain, early satiety, weight loss, and malaise. The onset of ascites, encephalopathy, jaundice, or variceal bleeding in patients with previously compensated cirrhosis should raise clinical suspicion for HCC.5 Rarely, patients may present with acute onset of severe abdominal pain and distension, hypotension, and a sudden drop in hematocrit resulting from tumor rupture and intraperitoneal bleeding. Emergent embolization of the bleeding blood vessel and/or surgery may be required for this complication.6

HCC is associated with a number of paraneoplastic syndromes resulting in hypoglycemia, erythrocytosis, hypercholesterolemia, hypercalcemia, severe watery diarrhea, and cutaneous manifestations.7 Extrahepatic spread at presentation is relatively uncommon, ranging between 10% and 30%.810 The most common sites of metastasis include lung, adrenal gland, regional lymph node regions (including the portahepatic and celiac lymph node chains), and bone.11 Importantly, extrahepatic nodal involvement is often difficult to assess based on cross-sectional imaging, as perihepatic lymphadenopathy is frequently present in patients with cirrhosis.12

Risk factors and epidemiology

The vast majority of cases of HCC occur in the setting of liver cirrhosis, and thus the etiology of HCC mirrors that of chronic liver disease and cirrhosis. Broadly, risk factors for HCC include viral infections, environmental toxins, comorbid conditions, inherited errors of metabolism, and autoimmune disorders.13 The unique geographic, sex, and age distributions of HCC are largely a result of the specific patterns of these risk factors, with the majority of cases the sequelae of hepatitis B and C viral infection and alcoholic liver disease.14

Chronic HBV infection (ie, hepatitis B surface antigen seropositivity) is the leading cause of HCC in Asia and Africa, where the virus is endemic and vertical transmission common.1,3 While HBV-induced HCC typically develops in cirrhotic livers, select populations of hepatitis B carriers remain at risk for HCC even in the absence of liver cirrhosis (Table 1).15 While the incidence of HCC in hepatitis B carriers depends on several factors, results from a prospective controlled study have revealed the annual incidence of HCC in hepatitis B carriers to be 0.5% in those without known cirrhosis and 2.5% in those with cirrhosis.16

Table 1.

At-risk groups for which surveillance for HCC is recommended

Patients at increased risk for HCC

Cirrhosis Without cirrhosis
• Hepatitis B Hepatitis B carrier
• Hepatitis C • Family history of HCC
• Alcoholic cirrhosis • Africans >20 years
• Nonalcoholic steatosis • Asian males ≥40 years
• Genetic hemachromatosis • Asian females ≥50 years
• Primary biliary cirrhosis
• α-1 antitrypsin deficiency

© John Wiley and Sons 2012. Adapted with permission from Bruix and Sherman.15

Abbreviation: HCC, hepatocellular carcinoma.

HCV infection represents a major risk factor for HCC, accounting for around one-third of cases in the United States and is thought, at least in part, to be responsible for the increasing incidence of HCC in the US.17,18 HCV infection confers the highest risk of HCC in patients with cirrhosis, with an annual incidence ranging between 2% and 8%.15 A prospective population-based study has revealed a 20-fold increased risk of HCC in those with HCV infection compared to those without infection.19 While the incidence of HBV-related HCC has decreased after the initiation of a universal vaccination program, the incidence of HCV-induced HCC, where an effective vaccine is currently unavailable, is expected to continue to increase over the next several decades.3,20

Excess alcohol consumption is a well-recognized risk factor for HCC, although the threshold dose and duration are less clear. Furthermore, viral hepatitis is a frequent comorbid condition in many patients suffering from alcoholism, and infection with either HBV or HCV further increases the risk of alcohol-induced HCC.21,22 Cigarette smoking has also been shown to be a risk factor for HCC, and a recent European-population-based case-control study revealed that smoking contributed to nearly 50% of all HCCs.23 However, given the high prevalence of smoking in this population and the frequent comorbid conditions of alcohol and viral hepatitis, smoking likely represents a cofactor in the development of HCC, rather than an independent etiologic agent.24

Epidemiologic studies have suggested a close correlation with obesity and diabetes and an increased risk of development of HCC.2527 Both of these conditions are also strongly associated with the development of nonalcoholic steatohepatitis (NASH), an increasingly recognized cause of cirrhosis.28 A recent observational study of patients with cirrhosis due to either NASH or HCV infection revealed annual incidence rates of HCC of 2.6% and 4%, respectively, suggesting that HCC surveillance may be indicated in NASH-induced cirrhotic patients.29 The most commonly associated environmental factor associated with HCC development is aflatoxin, a product of the Aspergillus fungus. High rates of dietary aflatoxin exposure, which commonly contaminates peanuts, soybeans, and corn, are frequent in developing countries and are associated with HCC.30,31

Several inherited metabolic disorders of the liver have been implicated in the development of HCC, including alpha-1 antitrypsin deficiency, certain porphyrias, Wilson’s disease, and hereditary hemachromatosis, each typically in the setting of cirrhosis.3234 Additionally, a number of automimmune disorders have been implicated in HCC pathogenesis, including autoimmune hepatitis, primary biliary cirrhosis, and primary sclerosing cholangitis.13,33 Among these disease entities, available data suggesting that the incidence rate of HCC in patients with cirrhosis resulting from both hereditary hemochromatosis and advanced primary biliary cirrhosis appear significant enough to justify active surveillance.3538

Surveillance

Surveillance is generally recommended in patients considered high risk for the development of HCC (Table 1).3,37,38 Evidence for the ability of surveillance to impact overall survival comes from a large randomized controlled trial conducted in China comparing no surveillance to semiannual evaluation of serum α-fetoprotein (AFP) and abdominal ultrasonography (US) in HBV-infected patients or those with chronic hepatitis.39 Despite the fact that less than 60% of the patients in the surveillance arm were screened appropriately, a 37% reduction in HCC-related mortality was found. Additionally, several nonrandomized trials and observational studies have reported a survival benefit in patients ultimately diagnosed with early stage disease, the target population in which early treatment interventions would most likely provide meaningful improvement in survival duration.40

Thus, serum AFP and US are the most commonly employed methods for screening for HCC, and while controversial,41 they are frequently performed in combination. The use of US in the detection of HCC generally results in >60% sensitivity and >90% specificity.42 However, US is highly operator-dependent, and the detection of tumors within a nodular cirrhotic liver is compromised and the sensitivity inferior.43 The sensitivity of AFP, using the commonly employed 20 ng/mL cutoff point, ranges between 25% and 60%,3 and is compromised by the fact that AFP is frequently not elevated in early stage disease. As a result, the sole use of AFP as a screening tool is not recommended. The time interval of surveillance is dependent on tumor doubling time, and based on estimates for HCC, is between 6 and 12 months.15 However, shorter interval follow-up has been recommended for cirrhotic patients with documented small liver nodules.44

Diagnosis

The modalities employed in the diagnosis of HCC depend on both the size of the lesion and underlying liver function, and include cross-sectional imaging, biopsy, and serum AFP. The incidental finding of a liver nodule, the detection of a liver nodule during surveillance US, or a rising AFP in the absence of a liver nodule on US, is typically followed by cross-sectional imaging. HCC lesions possess a distinct blood supply from the surrounding normal liver parenchyma, relying primarily on arterial blood from the hepatic artery while the surrounding liver is supplied mainly via the portal vein.45 Based on this, the use of four-phase (unenhanced, arterial, venous, delayed venous) helical computed tomography (CT) and dynamic contrast-enhanced magnetic resonance imaging (MRI) have established a classic imaging profile of HCC lesions characterized by arterial enhancement followed by hypointensity, or “washout,” in the delayed venous phase.46,47

Lesions less than 1 cm are generally followed by serial imaging every 3–6 months to assess changes in growth or vasculature that are suggestive of malignancy. Stability over a period of 1–2 years suggests the lesion is unlikely to be HCC.15 In cirrhotic patients or those with chronic liver disease, lesions greater than 1 cm in size that display both arterial enhancement and venous washout on either CT or MRI are considered diagnostic, and biopsy is typically not necessary.37,38 If, however, either of these classic imaging findings is not observed, complementary imaging is recommended, where again both imaging findings would be considered diagnostic of HCC. Discordant imaging on subsequent evaluation is frequently followed by biopsy. It should be pointed out that characterization of these-sized lesions is often difficult, and some experts recommend confirmation of enhancement and washout with both CT and MRI.3,48

In patients without underlying liver dysfunction, biopsy is generally recommended for lesions larger than 1 cm to confirm the diagnosis of HCC. Biopsy, either via fine-needle aspiration or core biopsy, unfortunately has several limitations, including sampling error, the inherent difficulty in discerning between high-grade dysplastic nodules and HCC, and the remote possibility of seeding tumor cells along the needle track.4951 Thus, in patients with a nondiagnostic biopsy, repeat biopsy and/or imaging are recommended.

The role of AFP in the diagnosis of HCC is controversial. While elevated levels of AFP in combination with a known hepatic mass have been shown to have a high positive predictive value in retrospective studies, AFP can be elevated in other primary liver malignancies as well as metastasis and is within normal limits in a large proportion of patients with known HCC.37,5254 As a result, both the National Comprehensive Cancer Network (NCCN) and the American Association for the Study of Liver Diseases (AASLD) no longer recommend its use in the diagnosis of HCC. Additionally, while other serum biomarkers, including des-gamma-carboxyprothrombin and lectin-bound α-fetoprotein, have been investigated in both surveillance and diagnosis of HCC, they are not currently recommended.55,56

Introduction to the management of HCC

Surgical resection

Surgical resection (ie, partial hepatectomy) is the optimal treatment for HCC. Preoperative assessment aims to address both resectability and operability. Resectability focuses on two issues: anatomical constraints of the primary tumor(s) and the probability of extrahepatic spread, and is typically ascertained via four-phase dynamic CT or MRI of the liver.46,57 Given the most common metastatic sites of HCC, a chest CT and bone scan are most commonly employed to evaluate distant spread.

Assessment of operability relies on a variety of factors, including a patient’s performance status, presence of comorbidities, severity of underlying liver disease, and estimates of the volume and function of the future liver remnant. The severity of underlying liver disease is most commonly determined through a combination of clinical and laboratory evaluation within the context of a scoring system, namely the Child–Pugh classification and the Model for End-Stage Liver Disease. However, additional information required to ensure adequate patient selection includes evaluation of portal hypertension, including clinical signs and symptoms, findings on CT or MRI, and/or measurement of the hepatic vein pressure gradient.58,59 Estimates of predicted postoperative liver volume and function are obtained through CT-based volumetric imaging, and these estimates have been shown to predict hepatic dysfunction in patients undergoing major liver resections.60,61 Finally, determination of the future liver remnant volume and function may be used to help determine if portal vein embolization, a technique employed to initiate hypertrophy in the anticipated liver remnant, is of value to extend the possibility of resection in otherwise suitable candidates.62,63

While thorough preoperative evaluation of the above parameters represents the standard of care, there are no fixed rules regarding tumor size, tumor number, presence of major vascular invasion, or extent of underlying liver disease. However, optimal patient and tumor characteristics include normal hepatic function or compensated (Child–Pugh A classification) cirrhosis, no evidence of portal hypertension, sufficient future liver remnant, and solitary, intrahepatic tumor without evidence of major vascular invasion.37,38,64 While perioperative mortality ranges widely in modern retrospective series (1%–24%), with more rigorous patient selection partial hepatectomy mortality rates are generally less than 5%.6568 Overall 5-year survival rates following resection of early stage HCC are often greater than 50%, and in highly selected patients with solitary lesions without vascular invasion, survival rates greater than 70% have been reported.59,66,69,70

Despite impressive survival rates, surgical resection is limited by high rates of local recurrence, often exceeding numbers achieved with 5-year survival outcomes.15,71 In the majority of cases, this is thought to be secondary to occult micrometastasis within the remaining liver parenchyma rather than inadequate surgical resection, and thus both neoadjuvant and adjuvant therapies have been explored. However, at least one controlled trial examining the role of neoadjuvant transarterial chemoembolization and a meta-analysis of three randomized trials of adjuvant systemic chemotherapy have demonstrated a worse outcome compared with resection alone.72,73 Yet given the recent success of systemic targeted therapy in advanced HCC (see below), the role of the multikinase inhibitor sorafenib in the postresection setting is being explored.74

Orthotopic liver transplantation

Orthotopic liver transplantation (OLT) represents an additional potentially curative therapy for early stage HCC. In a landmark paper, Mazzaferro et al reported an 85% 5-year survival and less than 10% recurrence rate of a highly selected group of patients with unresectable HCC who underwent liver transplantation.75 The shared tumor characteristics: solitary lesion <5 cm or < three lesions <3 cm each, became known as the Milan criteria and have since been adopted by the United Network for Organ Sharing as the requisite tumor characteristics for potential transplantation for the treatment of HCC. Additional United Network for Organ Sharing criteria include lack of radiographic evidence of macroscopic invasion or extrahepatic spread.76 Importantly, subsequent studies in which the Milan criteria were employed have demonstrated similar outcomes.77,78

An area of active debate focuses on the possibility of expanding the Milan criteria in order to provide OLT to patients with higher-stage disease. Several centers using expanded criteria have demonstrated 5-year survival rates within the range of those achieved using the Milan criteria.7981 Chief among these is the University of California, San Francisco, which has expanded criteria to include solitary tumors <6.5 cm and < three tumors <4.5 cm and cumulative tumor size <8 cm. While wide variation in survival exists in studies using these criteria, a recent study demonstrated a 5-year survival rate of 75%.82 Despite this, given the correlation between tumor size and number and risk of dissemination and the critical shortage of liver donors, Milan criteria are considered the standard of care at most transplant centers.83

Given the importance of maintaining sufficient functional liver capacity following surgical resection for HCC, it is generally accepted that first-line treatment for early stage HCC in patients with moderate to severe cirrhosis (Child–Pugh classes B and C) is OLT. However, with recurrence rates generally ∼10% with OTL versus up to ∼70% with surgical resection, optimal treatment for patients with compensated cirrhosis (Child–Pugh class A) with early HCC who meet the Milan criteria is controversial.84,85 There are currently no randomized controlled trials comparing these two modalities in this patient population, and current NCCN guidelines indicate initial treatment with either is acceptable.38

Ablation

The use of ablation for the treatment of HCC runs the gamut from agent used – toxic chemical agents (ethanol, acetic acid) or selective temperature gradients (radiofrequency, cryotherapy) – to method used – percutaneous, laproscopic, or open. The anticipated end result of each method is the same, that of selective tumor necrosis, commonly assessed by a lack of contrast uptake on dynamic CT or MR imaging following treatment and assessment of periprocedure serum AFP.86,87

The two most frequently used methods include percutaneous radiofrequency ablation (RFA) and percutaneous ethanol injection (PEI). Percutaneous ablation is a well-tolerated procedure, with major complications generally less than 10% for RFA and less than 3% for PEI, and morbidity rates less than 1% for RFA and extremely rare for PEI.8890 With regards to the optimal type of ablation, multiple prospective randomized controlled trials have demonstrated superior local tumor control with RFA, and three meta-analyses have demonstrated superior overall survival.9196 That said, the efficacy of complete control rates between RFA and PEI appears to be fairly similar for tumors less than 2 cm.91,92,97 While both of these methods have distinct advantages and disadvantages, it is important to note that they are limited by tumor size and location.98100

There is wide variation in reported overall survival rates, yet long-term studies of both RFA and PEI have achieved 5-year overall survival of over 50%.101,102 Like survival outcomes, local recurrence rates range widely among studies. Results from a recent randomized controlled trial demonstrate local recurrence rates with both RFA and PEI to be between 13% and 34%,103 while estimated 4-year recurrence rates of 70% and 85% with RFA and PEI, respectively, were obtained in a separate randomized controlled trial.93 On the other hand, a recent cohort study found a less-than-3% recurrence rate at 31 months in select patients with tumors less than 2 cm treated with RFA.104

Given the generally comparable survival and recurrence rates of ablation to surgical resection for early HCC, three randomized controlled trials have compared ablation with surgical resection. Two studies failed to show a significant benefit to surgery, with similar 3-year overall and disease-free survival.105,106 However, a recent larger study of patients with HCC meeting the Milan criteria demonstrated superior 5-year overall survival (76% vs 55%) and recurrence-free survival (51% vs 29%), as well as significantly lower overall recurrence rates (42% vs 63%) for those who underwent surgical resection compared to RFA.23 Taken together, most physicians favor surgical resection for patients with early HCC who are deemed both resectable and operable, even with small tumors. For nonsurgical patients with tumors less than 2 cm, either ablative technique can be used and case-specific characteristics often guide ultimate treatment decisions, while RFA is generally preferred for larger tumors.

Transarterial embolization

The principle of embolization relies on the same biologic feature of HCC exploited by hepatic imaging used for diagnosis: selective hepatic artery blood supply to tumors.45 In the case of embolization, selective tumor hypoxia and resultant necrosis is achieved via significant reduction in arterial blood flow through the use of image-guided catheter-based infusion of particles.107 Much like ablation, there is wide variation in the types of particles used, types of antitumor adjuvants employed, types of emulsifying agents used with adjuvant treatments, and number of treatment sessions given, all of which complicate the interpretation of the many studies examining its clinical utility.

Broadly speaking, embolization can be categorized as transarterial embolization (TAE) (ie, bland embolization), transarterial chemoembolization (TACE), and transarterial radioembolization (TARE). In the case of TACE, transarterial infusion of a chemotherapeutic agent (frequently doxorubicin or cisplatin) with or without an emulsifying agent (commonly Lipiodol, an oily substance with selective tumor retention) precedes infusion of embolic particles.108 Newer methods of TACE have employed drug-eluting beads as a way to concomitantly reduce blood flow and deliver sustained and controlled adjuvant therapy and reduce systemic side effects.109

Conversely, radionuclide conjugation of Lipiodol (iodine-131 Lipiodol) is generally done in the absence of specific embolization and is thought to exert sufficient antitumor effect through selective high-dose radiation.110 Additionally, while radionuclide-conjugated microspheres (commonly yttrium-90) do exhibit microvascular occlusive properties based on sizing of microspheres, embolization is less dramatic and resulting tumor necrosis is thought to be mediated predominantly via radiation rather than ischemia.107,111

Two landmark randomized controlled trials involving patients with unresectable HCC with preserved liver function have demonstrated a significant survival advantage with TACE compared to best supportive care. In the study by Llovet et al, 112 patients were randomized to either TAE, TACE, or best supportive care, and survival probabilities at 1 year and 2 years were 75% and 50% for TAE, 82% and 63% for TACE, and 63% and 27%, respectively, for supportive care. The study was stopped early after interim analysis revealed statistically significant survival advantage for TACE versus supportive care (P = 0.009); unfortunately, the study was underpowered to detect a difference between TACE and TAE.112 In the study by Lo et al, 80 patients were randomized to TACE vs best supportive care, and actuarial survival was significantly better in the TACE arm than in the supportive-care arm (1 year, 57% vs 32%; 2 years, 31% vs 11%; 3 years, 26% vs 3%; P = 0.002).113

Based in part on these results, TACE has become the most commonly employed embolization technique. Several recent studies have subsequently compared “conventional” TACE with TACE using drug-eluting beads (DEB-TACE). While a retrospective study has suggested a survival advantage with drug-eluting beads,114 a recent phase II prospective randomized trial revealed comparable effectiveness with the two modalities, albeit with significantly less systemic toxicity with DEB-TACE.115 Additionally, results from this randomized trial as well as an additional randomized trial that compared DEB-TACE to TAE revealed nonsignificantly higher rates of complete tumor response and disease control with DEB-TACE.115,116 There are currently no published randomized controlled trials comparing TACE (conventional or with drug-eluting beads) with TARE employing yttrium-90 microspheres. However, a recent comparative effective analysis of patients with unresectable HCC treated with either TACE or TARE with yttrium-90 revealed similar median survival outcomes (20.5 months vs 17.4 months, respectively, P = 0.232), while radioembolization resulted in significantly longer time to progression and reduced toxicity.117

While morbidity with embolization is relatively low, (generally < 5%), complications are common and include a transient postembolization syndrome consisting of abdominal pain, nausea, ileus, and fever.112,118,119 Overall survival rates vary widely, yet studies with TACE and TAE reported 2-year overall survival ranging from 20% to 60%.112,113,120 However, durable long-term survival is rare with embolization, as initial complete response rates are low and revascularization within the tumor bed frequently occurs.15 Thus, it is generally not employed with curative intent.

Systemic therapy

Despite the fact that most patients with HCC present with advanced disease, prior to 2008 no systemic therapy had demonstrated sustained improved survival, including single-agent or combined cytotoxic chemotherapy,121,122 immunotherapy alone or in combination with cytotoxic chemotherapy,123,124 tamoxifen or antiandrogens,125127 or somatostatin analogs.128,129 However, with the publication of two randomized placebo-controlled phase III trials, the oral multikinase inhibitor sorafenib has become the new reference standard for treatment of advanced HCC.130,131 Sorafenib inhibits both the rapidly accelerated fibrosarcoma (Raf) kinase and the vascular endothelial growth factor receptor (VEGFR) intracellular kinase pathways, resulting in suppression of tumor cell proliferation and angiogenesis.132,133

In the multicenter European Sorafenib Hepatocellular Carcinoma Assessment Randomized Protocol (SHARP) trial, 602 patients with advanced HCC who were systemic therapy–naive received either sorafenib or placebo until radiological or symptomatic progression.130 The study was stopped at the second planned interim analysis because of advantages in median overall survival (10.7 vs 7.9 months; hazard ratio, 0.69; 95% CI, 0.55–0.87; P < 0.001) and time to radiological progression (5.5 vs 2.8 months; P < 0.001) in the sorafenib arm. However, objective response rates to sorafenib were low according to radiographic criteria, and sorafenib did not increase the time to symptomatic progression. The underlying etiologies of HCC were primarily HCV (28%), alcohol (26%), and HBV (18%). The vast majority of patients had compensated liver disease and good performance status.

In the Asia–Pacific trial, the efficacy of sorafenib in the Asian population was evaluated in 226 patients in a similar design and dosing to the SHARP trial.131 Patients who received sorafenib again demonstrated significant benefits in median overall survival (6.5 vs 4.2 months; hazard ratio, 0.68; 95% CI, 0.50–0.93; P = 0.014) and time to radiological progression (2.8 vs 1.4 months) compared to placebo. While most patients again had preserved liver function, patients in the study tended to be younger, have more advanced-stage disease, and generally worse performance status. Additionally, the underlying etiology was different, with more than 70% of patients infected with HBV. These differences in patient and disease characteristics may account for the markedly reduced magnitude of benefit between the two trials (10.7 vs 6.5 months median overall survival).134 That said, the hazard ratio for survival was nearly identical between the two trials, suggesting comparable efficacy of sorafenib. Sorafenib was well tolerated with manageable side effects in both trials. Despite the fact that the original Food and Drug Administration approval of sorafenib in the US did not specify its use in underlying liver disease, current NCCN guidelines recommend sorafenib with Child–Pugh classes A and B only with unresectable, inoperable, or metastatic disease.38

Emerging tools for early diagnosis and management

Significant advances in diagnostic and therapeutic interventions are required to help transform HCC from a disease often diagnosed late and restricted largely to palliative therapies to one diagnosed early where curative intent is the goal. Core to this is the elucidation of the molecular pathways operative in HCC pathogenesis. Histopathologic analysis and epidemiologic studies of HCC arising in the setting of cirrhosis have suggested multistep hepatocarcinogenesis through the identification of morphologically distinct precancerous lesions, including dysplastic foci and dysplastic nodules, and early small HCC (<2 cm).135,136 Yet the pathologic distinction between these types of lesions is often difficult and biopsies of small tumors carry a false-negative rate of 30%–50%.137

Recent expression profiles of human samples have suggested particular mRNA expression subsets, or signatures, associated with both improved diagnostic accuracy and lesion discrimination.138141 For example, a 13-gene signature identified through high-throughput quantitative real-time polymerase chain reaction in 128 patient samples provided high diagnostic accuracy for HCC.138 Additionally, the expression of three genes (glypican 3, lymphatic vessel endothelial hyaluronan receptor 1 and survivin), has been reported to provide ∼90% accuracy in distinguishing dysplastic nodules from early HCC.139 Results of many of these genomic studies have been applied to more conventional immuno-histochemical analysis, and a recent prospective analysis of the protein expression of glypican 3, heat shock protein 70, and glutamine synthetase revealed 72% sensitivity and 100% specificity in detecting well-differentiated HCC in biopsy samples.142 These markers have been validated in a larger number of patients and have recently been suggested as part of the diagnostic algorithm for the American Association for the Study of Liver Diseases.37,143

Serum levels of several of these biomarkers have also been evaluated in diagnosis and surveillance, including glypican 3, des-gamma-carboxyl prothrombin (DCP), and Lens culinaris agglutinin-reactive AFP, an isoform of AFP.144147 While a recent study in a large population of patients with HCV-associated HCC suggested an improved sensitivity with a combination of AFP and DCP compared with either alone, to date additional serum biomarkers beyond AFP have not been incorporated in major HCC management guidelines.55

Radiographic imaging also plays a key role in the early diagnosis of HCC, and is critical for accurate staging and treatment-response assessment. The major difficulty in diagnostic imaging of the cirrhotic patient remains the accurate characterization of lesions less than 2 cm, and the incorporation of functional hepatic imaging and development of new contrast agents will likely improve the detection and characterization of these small tumors. Diffusion-weighted MRI has been shown to improve detection of HCC smaller than 2 cm, with sensitivities higher than multiphasic MRI alone, and represents a promising technique given its ease of implementation into current clinical practice.148,149

The use of two new hepatocyte-specific gadolinium chelate contrast agents, gadobenate dimeglumine (Multihance; Bracco, Milan, Italy) and gadoxetic acid (Primovist; Schering, Berlin, Germany), with multiphasic contrast-enhanced MRI has been shown to improve in the diagnosis of HCC over MRI or CT alone.150,151 These agents have also been shown to enhance the differentiation of small HCCs (≤2 cm) from arterial-enhancing pseudolesions, common benign hepatic vascular lesions that mimic early HCC.152,153

[18F]fluorodeoxyglucose positron emission tomography (FDG-PET) is limited by the varying degrees of FDG accumulation in primary HCC, and is not currently recommended in the diagnosis of HCC.37,38 While several studies have demonstrated a greater sensitivity for detection of distant metastasis with FDG-PET compared to CT, MRI, or radionuclide bone scan, its use is limited by high false-positive rates and it is currently not recommended by the AASLD or NCCN.8,37,38,154,155 The use of new radiotracers, including [11C]acetate may, however, improve the sensitivity and specificity of PET scanning in the diagnosis and evaluation of HCC.156

Assessment of tumor response to nonsurgical treatments has traditionally been based on changes in tumor size and number, delineated by the World Health Organization and the Response Evaluation Criteria in Solid Tumors criteria.157,158 However, these criteria are often inadequate for the evaluation of HCC, particularly as it relates to novel molecular therapies, which often result in tumor necrosis.159 Preliminary studies of both diffusion-weighted MRI and perfusion CT show promise in the evaluation of tumor response to both local ablative and systemic therapies.160162

Elucidation of signal transduction pathways implicated in tumorigenesis and development of targeted molecular therapies have fueled a revolution in oncology, and this has impacted the management of HCC directly with the demonstration of a significant survival advantage with the use of sorafenib. Given this success, the role of sorafenib alone and in combination with traditional locoregional therapies, systemic chemotherapy and novel molecularly targeted therapies is currently being investigated in various stages of disease (Table 2).163 Additionally, alterations in several signal transduction pathways have been detected in HCC, including Wnt signaling, p53, insulin-like growth factor, Ras, epidermal growth factor receptor (EGFR), Akt/target of rapamycin (TOR), c-MET, VEGFR, and fibroblast growth factor (FGF).164 This has resulted in the application of a myriad of new targeted therapies in both the preclinical and clinical setting, including the monoclonal antibodies bevacizumab (anti-VEGF) and cetuximab (anti-EGFR), and the small molecule inhibitors erlotinib (EGFR inhibitor), brivanib (FGF and VEGFR inhibitor), everolimus (mammalian TOR inhibitor) and linifanib (VEGFR and PDGFR inhibitor).133

Table 2.

Select phase III trials evaluating sorafenib

ID Acronym Active arm Control arm Primary outcome Indications
NCT01009593 BRISK-FL Brivanib Sorafenib Overall survival First line tx for advanced HCC
NCT01135056 SIRveNIB Yttrium-90 microspheres Sorafenib Overall survival First line tx for advanced HCC
NCT00901901 SEARCH Sorafenib + erlotinib Sorafenib Overall survival First line tx for advanced HCC
NCT01015833 Sorafenib + doxirubicin Sorafenib Overall survival First line tx for advanced HCC
NCT01004978 TACE + sorafenib TACE + placebo Progression-free survival First line tx for intermediate HCC
ISRCTN24081794 HeiLivCa TACE + sorafenib TACE + placebo Time to progression Bridge therapy for liver transplantation
NCT00494299 Sorafenib Placebo Time to progression Prevention of recurrence after embolization
NCT00692770 STORM Sorafenib Placebo Recurrence-free survival Prevention of recurrence after resection or ablation

Adapted from Gastroenterology, 140:5, Villanueva A and Llovet JM, Targeted therapies for hepatocellular carcinoma, p. 17, © 2011, with permission from Elsevier Ltd.163

Abbreviations: tx, treatment; HCC, hepatocellular carcinoma; TACE, transarterial chemoembolization.

Many of these therapies target well-established proteins that regulate critical aspects of tumorigenesis, including angiogenesis, proliferation, and survival. However, recent work has implicated microRNA dysregulation in many of these processes during hepatocarcinogenesis, and early studies of human HCC microRNA expression profiling have suggested expression signatures predictive of metastasis and survival.165,166 Epigenetic alterations, including DNA methylation and histone modification, have also been implicated in the pathogenesis of HCC.164 Importantly, therapeutic interventions targeting some of these aberrant processes in the form of histone deacetylase inhibitors are currently being investigated in clinical trials of HCC.163

Conclusion

Hepatocellular carcinoma is an aggressive cancer that frequently occurs in the setting of chronic liver disease and cirrhosis. It is typically diagnosed late in the disease course, often precluding curative surgical therapies. Furthermore, concomitant liver dysfunction often further hampers both curative and palliative therapies. However, HCC is unique among many neoplasms in that effective preventative measures, including HBV vaccination, universal screening of blood products, use of safe injection practices, education and treatment of alcoholism and intravenous drug abuse, reduction in aflatoxin through control of fungal contamination, and initiation of antiviral therapy are in place and have been shown to be effective.13,167

Advances in locoregional therapies, including the use of drug-eluting beads and radionuclide-conjugated microspheres, represent strategies that may further improve survival among patients with intermediate-stage HCC and preserved liver function. Moreover, the demonstration of the safety and efficacy of sorafenib as a frontline treatment for advanced HCC in patients with preserved liver function represents a significant advancement in what was an otherwise bleak set of treatment options for this group of patients. Effective molecularly targeted therapies may also hold promise as adjuvants to primary surgical therapies, currently limited by high rates of disease recurrence. Finally, it is hoped that, much as has been shown for breast and colorectal carcinoma, active research aimed at the elucidation of the molecular pathogenesis of HCC and the identification of new biomarkers will result in further advances in the prevention, diagnosis, and treatment of HCC.

Footnotes

Disclosure

The authors disclose no conflict of interest.

References

  • 1.Bosch FX, Ribes J, Cleries R, Diaz M. Epidemiology of hepatocellular carcinoma. Clin Liver Dis. 2005;9(2):191–211. v. doi: 10.1016/j.cld.2004.12.009. [DOI] [PubMed] [Google Scholar]
  • 2.Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
  • 3.El-Serag HB. Hepatocellular carcinoma. N Engl J Med. 2011;365(12):1118–1127. doi: 10.1056/NEJMra1001683. [DOI] [PubMed] [Google Scholar]
  • 4.A new prognostic system for hepatocellular carcinoma: a retrospective study of 435 patients: the Cancer of the Liver Italian Program (CLIP) investigators. Hepatology. 1998;28(3):751–755. doi: 10.1002/hep.510280322. [DOI] [PubMed] [Google Scholar]
  • 5.Bartlett DL, Di Bisceglie AM, Dawson LA. Cancer of the liver. In: DeVita VT, Lawrence TS, Rosenberg SA, editors. Cancer: Principles and Practice of Oncology. 8th ed. Philadelphia: Lippincott Williams and Wilkins; 2008. pp. 1129–1156. [Google Scholar]
  • 6.Rossetto A, Adani GL, Risaliti A, et al. Combined approach for spontaneous rupture of hepatocellular carcinoma. World J Hepatol. 2010;2(1):49–51. doi: 10.4254/wjh.v2.i1.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kassianides C, Kew MC. The clinical manifestations and natural history of hepatocellular carcinoma. Gastroenterol Clin North Am. 1987;16(4):553–562. [PubMed] [Google Scholar]
  • 8.Yoon KT, Kim JK, Kim do Y, et al. Role of 18F-fluorodeoxyglucose positron emission tomography in detecting extrahepatic metastasis in pretreatment staging of hepatocellular carcinoma. Oncology (Williston Park) 2007;72(Suppl 1):104–110. doi: 10.1159/000111715. [DOI] [PubMed] [Google Scholar]
  • 9.Wudel LJ, Jr, Delbeke D, Morris D, et al. The role of [18F]fluorodeoxyglucose positron emission tomography imaging in the evaluation of hepatocellular carcinoma. Am Surg. 2003;69(2):117–124. discussion 124–126. [PubMed] [Google Scholar]
  • 10.Carriaga MT, Henson DE. Liver, gallbladder, extrahepatic bile ducts, and pancreas. Cancer. 1995;75(Suppl 1):171–190. doi: 10.1002/1097-0142(19950101)75:1+<171::aid-cncr2820751306>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 11.Katyal S, Oliver JH, 3rd, Peterson MS, Ferris JV, Carr BS, Baron RL. Extrahepatic metastases of hepatocellular carcinoma. Radiology. 2000;216(3):698–703. doi: 10.1148/radiology.216.3.r00se24698. [DOI] [PubMed] [Google Scholar]
  • 12.Dodd GD, 3rd, Baron RL, Oliver JH, 3rd, Federle MP, Baumgartel PB. Enlarged abdominal lymph nodes in end-stage cirrhosis: CT-histopathologic correlation in 507 patients. Radiology. 1997;203(1):127–130. doi: 10.1148/radiology.203.1.9122379. [DOI] [PubMed] [Google Scholar]
  • 13.Sohal DP, Sun W. Hepatocellular carcinoma: prevention and therapy. Curr Oncol Rep. 2011;13(3):186–194. doi: 10.1007/s11912-011-0165-0. [DOI] [PubMed] [Google Scholar]
  • 14.McGlynn KA, London WT. The global epidemiology of hepatocellular carcinoma: present and future. Clin Liver Dis. 2011;15(2):223–243. vii–x. doi: 10.1016/j.cld.2011.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bruix J, Sherman M. Management of hepatocellular carcinoma. Hepatology. 2005;42(5):1208–1236. doi: 10.1002/hep.20933. [DOI] [PubMed] [Google Scholar]
  • 16.Beasley RP, Hwang LY, Lin CC, Chien CS. Hepatocellular carcinoma and hepatitis B virus. A prospective study of 22 707 men in Taiwan. Lancet. 1981;2(8256):1129–1133. doi: 10.1016/s0140-6736(81)90585-7. [DOI] [PubMed] [Google Scholar]
  • 17.Davila JA, Morgan RO, Shaib Y, McGlynn KA, El-Serag HB. Hepatitis C infection and the increasing incidence of hepatocellular carcinoma: a population-based study. Gastroenterology. 2004;127(5):1372–1380. doi: 10.1053/j.gastro.2004.07.020. [DOI] [PubMed] [Google Scholar]
  • 18.El-Serag HB, Mason AC. Risk factors for the rising rates of primary liver cancer in the United States. Arch Intern Med. 2000;160(21):3227–3230. doi: 10.1001/archinte.160.21.3227. [DOI] [PubMed] [Google Scholar]
  • 19.Sun CA, Wu DM, Lin CC, et al. Incidence and cofactors of hepatitis C virus-related hepatocellular carcinoma: a prospective study of 12,008 men in Taiwan. Am J Epidemiol. 2003;157(8):674–682. doi: 10.1093/aje/kwg041. [DOI] [PubMed] [Google Scholar]
  • 20.Zanetti AR, Van Damme P, Shouval D. The global impact of vaccination against hepatitis B: a historical overview. Vaccine. 2008;26(49):6266–6273. doi: 10.1016/j.vaccine.2008.09.056. [DOI] [PubMed] [Google Scholar]
  • 21.Befrits R, Hedman M, Blomquist L, et al. Chronic hepatitis C in alcoholic patients: prevalence, genotypes, and correlation to liver disease. Scand J Gastroenterol. 1995;30(11):1113–1118. doi: 10.3109/00365529509101616. [DOI] [PubMed] [Google Scholar]
  • 22.Donato F, Tagger A, Gelatti U, et al. Alcohol and hepatocellular carcinoma: the effect of lifetime intake and hepatitis virus infections in men and women. Am J Epidemiol. 2002;155(4):323–331. doi: 10.1093/aje/155.4.323. [DOI] [PubMed] [Google Scholar]
  • 23.Trichopoulos D, Bamia C, Lagiou P, et al. Hepatocellular carcinoma risk factors and disease burden in a European cohort: a nested case-control study. J Natl Cancer Inst. 2011;103(22):1686–1695. doi: 10.1093/jnci/djr395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sherman M, Llovet JM. Smoking, hepatitis B virus infection, and development of hepatocellular carcinoma. J Natl Cancer Inst. 2011;103(22):1642–1643. doi: 10.1093/jnci/djr430. [DOI] [PubMed] [Google Scholar]
  • 25.Wolk A, Gridley G, Svensson M, et al. A prospective study of obesity and cancer risk (Sweden) Cancer Causes Control. 2001;12(1):13–21. doi: 10.1023/a:1008995217664. [DOI] [PubMed] [Google Scholar]
  • 26.Polesel J, Zucchetto A, Montella M, et al. The impact of obesity and diabetes mellitus on the risk of hepatocellular carcinoma. Ann Oncol. 2009;20(2):353–357. doi: 10.1093/annonc/mdn565. [DOI] [PubMed] [Google Scholar]
  • 27.Regimbeau JM, Colombat M, Mognol P, et al. Obesity and diabetes as a risk factor for hepatocellular carcinoma. Liver Transpl. 2004;10(2 Suppl 1):S69–S73. doi: 10.1002/lt.20033. [DOI] [PubMed] [Google Scholar]
  • 28.Takamatsu S, Noguchi N, Kudoh A, et al. Influence of risk factors for metabolic syndrome and non-alcoholic fatty liver disease on the progression and prognosis of hepatocellular carcinoma. Hepatogastroenterology. 2008;55(82–83):609–614. [PubMed] [Google Scholar]
  • 29.Ascha MS, Hanouneh IA, Lopez R, Tamimi TA, Feldstein AF, Zein NN. The incidence and risk factors of hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. Hepatology. 2010;51(6):1972–1978. doi: 10.1002/hep.23527. [DOI] [PubMed] [Google Scholar]
  • 30.Chen CJ, Wang LY, Lu SN, et al. Elevated aflatoxin exposure and increased risk of hepatocellular carcinoma. Hepatology. 1996;24(1):38–42. doi: 10.1002/hep.510240108. [DOI] [PubMed] [Google Scholar]
  • 31.Sanyal AJ, Yoon SK, Lencioni R. The etiology of hepatocellular carcinoma and consequences for treatment. Oncologist. 2010;15(Suppl 4):14–22. doi: 10.1634/theoncologist.2010-S4-14. [DOI] [PubMed] [Google Scholar]
  • 32.Villanueva A, Newell P, Hoshida Y. Inherited hepatocellular carcinoma. Best Pract Res Clin Gastroenterol. 2010;24(5):725–734. doi: 10.1016/j.bpg.2010.07.008. [DOI] [PubMed] [Google Scholar]
  • 33.Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology. 2004;127(5 Suppl 1):S35–S50. doi: 10.1053/j.gastro.2004.09.014. [DOI] [PubMed] [Google Scholar]
  • 34.Dragani TA. Risk of HCC: genetic heterogeneity and complex genetics. J Hepatol. 2010;52(2):252–257. doi: 10.1016/j.jhep.2009.11.015. [DOI] [PubMed] [Google Scholar]
  • 35.Fracanzani AL, Conte D, Fraquelli M, et al. Increased cancer risk in a cohort of 230 patients with hereditary hemochromatosis in comparison to matched control patients with non-iron-related chronic liver disease. Hepatology. 2001;33(3):647–651. doi: 10.1053/jhep.2001.22506. [DOI] [PubMed] [Google Scholar]
  • 36.Caballeria L, Pares A, Castells A, Gines A, Bru C, Rodes J. Hepatocellular carcinoma in primary biliary cirrhosis: similar incidence to that in hepatitis C virus-related cirrhosis. Am J Gastroenterol. 2001;96(4):1160–1163. doi: 10.1111/j.1572-0241.2001.03695.x. [DOI] [PubMed] [Google Scholar]
  • 37.Bruix J, Sherman M. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53(3):1020–1022. doi: 10.1002/hep.24199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.National Comprehensive Cancer Network National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology: Hepatobiliary Cancers. 2012. Version 2. Available from: http://www.nccn.org/professionals/physician_gls/pdf/hepatobiliary.pdf. Accessed January 27, 2012. [DOI] [PMC free article] [PubMed]
  • 39.Zhang BH, Yang BH, Tang ZY. Randomized controlled trial of screening for hepatocellular carcinoma. J Cancer Res Clin Oncol. 2004;130(7):417–422. doi: 10.1007/s00432-004-0552-0. [DOI] [PubMed] [Google Scholar]
  • 40.Wong LL, Limm WM, Severino R, Wong LM. Improved survival with screening for hepatocellular carcinoma. Liver Transpl. 2000;6(3):320–325. doi: 10.1053/lv.2000.4875. [DOI] [PubMed] [Google Scholar]
  • 41.Sherman M. Serological surveillance for hepatocellular carcinoma: time to quit. J Hepatol. 2010;52(4):614–615. doi: 10.1016/j.jhep.2009.11.026. [DOI] [PubMed] [Google Scholar]
  • 42.Bolondi L, Sofia S, Siringo S, et al. Surveillance programme of cirrhotic patients for early diagnosis and treatment of hepatocellular carcinoma: a cost effectiveness analysis. Gut. 2001;48(2):251–259. doi: 10.1136/gut.48.2.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chen TH, Chen CJ, Yen MF, et al. Ultrasound screening and risk factors for death from hepatocellular carcinoma in a high risk group in Taiwan. Int J Cancer. 2002;98(2):257–261. doi: 10.1002/ijc.10122. [DOI] [PubMed] [Google Scholar]
  • 44.Yao FY. Should surveillance for hepatocellular carcinoma be increased in patients with cirrhosis and small liver nodules? Nat Clin Pract Gastroenterol Hepatol. 2006;3(10):544–545. doi: 10.1038/ncpgasthep0608. [DOI] [PubMed] [Google Scholar]
  • 45.Breedis C, Young G. The blood supply of neoplasms in the liver. Am J Pathol. 1954;30(5):969–977. [PMC free article] [PubMed] [Google Scholar]
  • 46.Miller G, Schwartz LH, D’Angelica M. The use of imaging in the diagnosis and staging of hepatobiliary malignancies. Surg Oncol Clin N Am. 2007;16(2):343–368. doi: 10.1016/j.soc.2007.04.001. [DOI] [PubMed] [Google Scholar]
  • 47.Forner A, Vilana R, Ayuso C, et al. Diagnosis of hepatic nodules 20 mm or smaller in cirrhosis: prospective validation of the noninvasive diagnostic criteria for hepatocellular carcinoma. Hepatology. 2008;47(1):97–104. doi: 10.1002/hep.21966. [DOI] [PubMed] [Google Scholar]
  • 48.Yeh JJ, Uemura M. Hepatocellular carcinoma. N Engl J Med. 2012;366(1):92. doi: 10.1056/NEJMc1112501. author reply 92–93. [DOI] [PubMed] [Google Scholar]
  • 49.Wee A. Fine needle aspiration biopsy of hepatocellular carcinoma and hepatocellular nodular lesions: role, controversies and approach to diagnosis. Cytopathology. 2011;22(5):287–305. doi: 10.1111/j.1365-2303.2011.00882.x. [DOI] [PubMed] [Google Scholar]
  • 50.Pawlik TM, Gleisner AL, Anders RA, Assumpcao L, Maley W, Choti MA. Preoperative assessment of hepatocellular carcinoma tumor grade using needle biopsy: implications for transplant eligibility. Ann Surg. 2007;245(3):435–442. doi: 10.1097/01.sla.0000250420.73854.ad. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Silva MA, Hegab B, Hyde C, Guo B, Buckels JA, Mirza DF. Needle track seeding following biopsy of liver lesions in the diagnosis of hepatocellular cancer: a systematic review and meta-analysis. Gut. 2008;57(11):1592–1596. doi: 10.1136/gut.2008.149062. [DOI] [PubMed] [Google Scholar]
  • 52.Torzilli G, Minagawa M, Takayama T, et al. Accurate preoperative evaluation of liver mass lesions without fine-needle biopsy. Hepatology. 1999;30(4):889–893. doi: 10.1002/hep.510300411. [DOI] [PubMed] [Google Scholar]
  • 53.Levy I, Greig PD, Gallinger S, Langer B, Sherman M. Resection of hepatocellular carcinoma without preoperative tumor biopsy. Ann Surg. 2001;234(2):206–209. doi: 10.1097/00000658-200108000-00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lok AS, Lai CL. alpha-Fetoprotein monitoring in Chinese patients with chronic hepatitis B virus infection: role in the early detection of hepatocellular carcinoma. Hepatology. 1989;9(1):110–115. doi: 10.1002/hep.1840090119. [DOI] [PubMed] [Google Scholar]
  • 55.Lok AS, Sterling RK, Everhart JE, et al. Des-gamma-carboxy prothrombin and alpha-fetoprotein as biomarkers for the early detection of hepatocellular carcinoma. Gastroenterology. 2010;138(2):493–502. doi: 10.1053/j.gastro.2009.10.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Marrero JA, Feng Z, Wang Y, et al. Alpha-fetoprotein, des-gamma carboxyprothrombin, and lectin-bound alpha-fetoprotein in early hepatocellular carcinoma. Gastroenterology. 2009;137(1):110–118. doi: 10.1053/j.gastro.2009.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sangiovanni A, Manini MA, Iavarone M, et al. The diagnostic and economic impact of contrast imaging techniques in the diagnosis of small hepatocellular carcinoma in cirrhosis. Gut. 2010;59(5):638–644. doi: 10.1136/gut.2009.187286. [DOI] [PubMed] [Google Scholar]
  • 58.Bruix J, Castells A, Bosch J, et al. Surgical resection of hepatocellular carcinoma in cirrhotic patients: prognostic value of preoperative portal pressure. Gastroenterology. 1996;111(4):1018–1022. doi: 10.1016/s0016-5085(96)70070-7. [DOI] [PubMed] [Google Scholar]
  • 59.Llovet JM, Fuster J, Bruix J. Intention-to-treat analysis of surgical treatment for early hepatocellular carcinoma: resection versus transplantation. Hepatology. 1999;30(6):1434–1440. doi: 10.1002/hep.510300629. [DOI] [PubMed] [Google Scholar]
  • 60.Kubota K, Makuuchi M, Kusaka K, et al. Measurement of liver volume and hepatic functional reserve as a guide to decision-making in resectional surgery for hepatic tumors. Hepatology. 1997;26(5):1176–1181. doi: 10.1053/jhep.1997.v26.pm0009362359. [DOI] [PubMed] [Google Scholar]
  • 61.Shoup M, Gonen M, D’Angelica M, et al. Volumetric analysis predicts hepatic dysfunction in patients undergoing major liver resection. J Gastrointest Surg. 2003;7(3):325–330. doi: 10.1016/s1091-255x(02)00370-0. [DOI] [PubMed] [Google Scholar]
  • 62.Abdalla EK, Hicks ME, Vauthey JN. Portal vein embolization: rationale, technique and future prospects. Br J Surg. 2001;88(2):165–175. doi: 10.1046/j.1365-2168.2001.01658.x. [DOI] [PubMed] [Google Scholar]
  • 63.Farges O, Belghiti J, Kianmanesh R, et al. Portal vein embolization before right hepatectomy: prospective clinical trial. Ann Surg. 2003;237(2):208–217. doi: 10.1097/01.SLA.0000048447.16651.7B. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Vauthey JN, Dixon E, Abdalla EK, et al. Pretreatment assessment of hepatocellular carcinoma: expert consensus statement. HPB (Oxford) 2010;12(5):289–299. doi: 10.1111/j.1477-2574.2010.00181.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pawlik TM, Poon RT, Abdalla EK, et al. Critical appraisal of the clinical and pathologic predictors of survival after resection of large hepatocellular carcinoma. Arch Surg. 2005;140(5):450–457. doi: 10.1001/archsurg.140.5.450. discussion 457–458. [DOI] [PubMed] [Google Scholar]
  • 66.Chok KS, Ng KK, Poon RT, Lo CM, Fan ST. Impact of postoperative complications on long-term outcome of curative resection for hepatocellular carcinoma. Br J Surg. 2009;96(1):81–87. doi: 10.1002/bjs.6358. [DOI] [PubMed] [Google Scholar]
  • 67.Yang LY, Fang F, Ou DP, Wu W, Zeng ZJ, Wu F. Solitary large hepatocellular carcinoma: a specific subtype of hepatocellular carcinoma with good outcome after hepatic resection. Ann Surg. 2009;249(1):118–123. doi: 10.1097/SLA.0b013e3181904988. [DOI] [PubMed] [Google Scholar]
  • 68.Tjandra JJ, Fan ST, Wong J. Peri-operative mortality in hepatic resection. Aust N Z J Surg. 1991;61(3):201–206. doi: 10.1111/j.1445-2197.1991.tb07592.x. [DOI] [PubMed] [Google Scholar]
  • 69.Arii S, Yamaoka Y, Futagawa S, et al. Results of surgical and nonsurgical treatment for small-sized hepatocellular carcinomas: a retrospective and nationwide survey in Japan. The Liver Cancer Study Group of Japan. Hepatology. 2000;32(6):1224–1229. doi: 10.1053/jhep.2000.20456. [DOI] [PubMed] [Google Scholar]
  • 70.Shi M, Guo RP, Lin XJ, et al. Partial hepatectomy with wide versus narrow resection margin for solitary hepatocellular carcinoma: a prospective randomized trial. Ann Surg. 2007;245(1):36–43. doi: 10.1097/01.sla.0000231758.07868.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kianmanesh R, Regimbeau JM, Belghiti J. Selective approach to major hepatic resection for hepatocellular carcinoma in chronic liver disease. Surg Oncol Clin N Am. 2003;12(1):51–63. doi: 10.1016/s1055-3207(02)00090-x. [DOI] [PubMed] [Google Scholar]
  • 72.Sasaki A, Iwashita Y, Shibata K, Ohta M, Kitano S, Mori M. Preoperative transcatheter arterial chemoembolization reduces long-term survival rate after hepatic resection for resectable hepatocellular carcinoma. Eur J Surg Oncol. 2006;32(7):773–779. doi: 10.1016/j.ejso.2006.04.002. [DOI] [PubMed] [Google Scholar]
  • 73.Ono T, Yamanoi A, Nazmy El Assal O, Kohno H, Nagasue N. Adjuvant chemotherapy after resection of hepatocellular carcinoma causes deterioration of long-term prognosis in cirrhotic patients: metaanalysis of three randomized controlled trials. Cancer. 2001;91(12):2378–2385. [PubMed] [Google Scholar]
  • 74.Printz C. Clinical trials of note. Sorafenib as adjuvant treatment in the prevention of disease recurrence in patients with hepatocellular carcinoma (HCC) (STORM) Cancer. 2009;115(20):4646. doi: 10.1002/cncr.24673. [DOI] [PubMed] [Google Scholar]
  • 75.Mazzaferro V, Regalia E, Doci R, et al. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996;334(11):693–699. doi: 10.1056/NEJM199603143341104. [DOI] [PubMed] [Google Scholar]
  • 76.Martin AP, Bartels M, Hauss J, Fangmann J. Overview of the MELD score and the UNOS adult liver allocation system. Transplant Proc. 2007;39(10):3169–3174. doi: 10.1016/j.transproceed.2007.04.025. [DOI] [PubMed] [Google Scholar]
  • 77.Jonas S, Bechstein WO, Steinmuller T, et al. Vascular invasion and histopathologic grading determine outcome after liver transplantation for hepatocellular carcinoma in cirrhosis. Hepatology. 2001;33(5):1080–1086. doi: 10.1053/jhep.2001.23561. [DOI] [PubMed] [Google Scholar]
  • 78.Figueras J, Ibanez L, Ramos E, et al. Selection criteria for liver transplantation in early-stage hepatocellular carcinoma with cirrhosis: results of a multicenter study. Liver Transpl. 2001;7(10):877–883. doi: 10.1053/jlts.2001.27856. [DOI] [PubMed] [Google Scholar]
  • 79.Duffy JP, Vardanian A, Benjamin E, et al. Liver transplantation criteria for hepatocellular carcinoma should be expanded: a 22-year experience with 467 patients at UCLA. Ann Surg. 2007;246(3):502–509. doi: 10.1097/SLA.0b013e318148c704. discussion 509–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yao FY, Ferrell L, Bass NM, Bacchetti P, Ascher NL, Roberts JP. Liver transplantation for hepatocellular carcinoma: comparison of the proposed UCSF criteria with the Milan criteria and the Pittsburgh modified TNM criteria. Liver Transpl. 2002;8(9):765–774. doi: 10.1053/jlts.2002.34892. [DOI] [PubMed] [Google Scholar]
  • 81.Lee SG, Hwang S, Moon DB, et al. Expanded indication criteria of living donor liver transplantation for hepatocellular carcinoma at one large-volume center. Liver Transpl. 2008;14(7):935–945. doi: 10.1002/lt.21445. [DOI] [PubMed] [Google Scholar]
  • 82.Yao FY, Ferrell L, Bass NM, et al. Liver transplantation for hepatocellular carcinoma: expansion of the tumor size limits does not adversely impact survival. Hepatology. 2001;33(6):1394–1403. doi: 10.1053/jhep.2001.24563. [DOI] [PubMed] [Google Scholar]
  • 83.Hsu HC, Wu TT, Wu MZ, Sheu JC, Lee CS, Chen DS. Tumor invasiveness and prognosis in resected hepatocellular carcinoma. Clinical and pathogenetic implications. Cancer. 1988;61(10):2095–2099. doi: 10.1002/1097-0142(19880515)61:10<2095::aid-cncr2820611027>3.0.co;2-h. [DOI] [PubMed] [Google Scholar]
  • 84.Llovet JM, Bruix J, Gores GJ. Surgical resection versus transplantation for early hepatocellular carcinoma: clues for the best strategy. Hepatology. 2000;31(4):1019–1021. doi: 10.1053/he.2000.6959. [DOI] [PubMed] [Google Scholar]
  • 85.Poon RT. Optimal initial treatment for early hepatocellular carcinoma in patients with preserved liver function: transplantation or resection? Ann Surg Oncol. 2007;14(2):541–547. doi: 10.1245/s10434-006-9156-z. [DOI] [PubMed] [Google Scholar]
  • 86.Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30(1):52–60. doi: 10.1055/s-0030-1247132. [DOI] [PubMed] [Google Scholar]
  • 87.Riaz A, Ryu RK, Kulik LM, et al. Alpha-fetoprotein response after locoregional therapy for hepatocellular carcinoma: oncologic marker of radiologic response, progression, and survival. J Clin Oncol. 2009;27(34):5734–5742. doi: 10.1200/JCO.2009.23.1282. [DOI] [PubMed] [Google Scholar]
  • 88.Livraghi T, Bolondi L, Lazzaroni S, et al. Percutaneous ethanol injection in the treatment of hepatocellular carcinoma in cirrhosis. A study on 207 patients. Cancer. 1992;69(4):925–929. doi: 10.1002/1097-0142(19920215)69:4<925::aid-cncr2820690415>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
  • 89.Kasugai H, Osaki Y, Oka H, Kudo M, Seki T. Severe complications of radiofrequency ablation therapy for hepatocellular carcinoma: an analysis of 3,891 ablations in 2,614 patients. Oncology (Williston Park) 2007;72(Suppl 1):72–75. doi: 10.1159/000111710. [DOI] [PubMed] [Google Scholar]
  • 90.Ebara M, Okabe S, Kita K, et al. Percutaneous ethanol injection for small hepatocellular carcinoma: therapeutic efficacy based on 20-year observation. J Hepatol. 2005;43(3):458–464. doi: 10.1016/j.jhep.2005.03.033. [DOI] [PubMed] [Google Scholar]
  • 91.Brunello F, Veltri A, Carucci P, et al. Radiofrequency ablation versus ethanol injection for early hepatocellular carcinoma: A randomized controlled trial. Scand J Gastroenterol. 2008;43(6):727–735. doi: 10.1080/00365520701885481. [DOI] [PubMed] [Google Scholar]
  • 92.Lencioni RA, Allgaier HP, Cioni D, et al. Small hepatocellular carcinoma in cirrhosis: randomized comparison of radio-frequency thermal ablation versus percutaneous ethanol injection. Radiology. 2003;228(1):235–240. doi: 10.1148/radiol.2281020718. [DOI] [PubMed] [Google Scholar]
  • 93.Shiina S, Teratani T, Obi S, et al. A randomized controlled trial of radiofrequency ablation with ethanol injection for small hepatocellular carcinoma. Gastroenterology. 2005;129(1):122–130. doi: 10.1053/j.gastro.2005.04.009. [DOI] [PubMed] [Google Scholar]
  • 94.Cho YK, Kim JK, Kim MY, Rhim H, Han JK. Systematic review of randomized trials for hepatocellular carcinoma treated with percutaneous ablation therapies. Hepatology. 2009;49(2):453–459. doi: 10.1002/hep.22648. [DOI] [PubMed] [Google Scholar]
  • 95.Orlando A, Leandro G, Olivo M, Andriulli A, Cottone M. Radiofrequency thermal ablation vs percutaneous ethanol injection for small hepatocellular carcinoma in cirrhosis: meta-analysis of randomized controlled trials. Am J Gastroenterol. 2009;104(2):514–524. doi: 10.1038/ajg.2008.80. [DOI] [PubMed] [Google Scholar]
  • 96.Bouza C, Lopez-Cuadrado T, Alcazar R, Saz-Parkinson Z, Amate JM. Meta-analysis of percutaneous radiofrequency ablation versus ethanol injection in hepatocellular carcinoma. BMC Gastroenterol. 2009;9:31. doi: 10.1186/1471-230X-9-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Livraghi T, Goldberg SN, Lazzaroni S, Meloni F, Solbiati L, Gazelle GS. Small hepatocellular carcinoma: treatment with radio-frequency ablation versus ethanol injection. Radiology. 1999;210(3):655–661. doi: 10.1148/radiology.210.3.r99fe40655. [DOI] [PubMed] [Google Scholar]
  • 98.Mazzaferro V, Battiston C, Perrone S, et al. Radiofrequency ablation of small hepatocellular carcinoma in cirrhotic patients awaiting liver transplantation: a prospective study. Ann Surg. 2004;240(5):900–909. doi: 10.1097/01.sla.0000143301.56154.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Llovet JM, Vilana R, Bru C, et al. Increased risk of tumor seeding after percutaneous radiofrequency ablation for single hepatocellular carcinoma. Hepatology. 2001;33(5):1124–1129. doi: 10.1053/jhep.2001.24233. [DOI] [PubMed] [Google Scholar]
  • 100.Lu DS, Yu NC, Raman SS, et al. Radiofrequency ablation of hepatocellular carcinoma: treatment success as defined by histologic examination of the explanted liver. Radiology. 2005;234(3):954–960. doi: 10.1148/radiol.2343040153. [DOI] [PubMed] [Google Scholar]
  • 101.Sala M, Llovet JM, Vilana R, et al. Initial response to percutaneous ablation predicts survival in patients with hepatocellular carcinoma. Hepatology. 2004;40(6):1352–1360. doi: 10.1002/hep.20465. [DOI] [PubMed] [Google Scholar]
  • 102.Livraghi T, Giorgio A, Marin G, et al. Hepatocellular carcinoma and cirrhosis in 746 patients: long-term results of percutaneous ethanol injection. Radiology. 1995;197(1):101–108. doi: 10.1148/radiology.197.1.7568806. [DOI] [PubMed] [Google Scholar]
  • 103.Lin SM, Lin CJ, Lin CC, Hsu CW, Chen YC. Randomised controlled trial comparing percutaneous radiofrequency thermal ablation, percutaneous ethanol injection, and percutaneous acetic acid injection to treat hepatocellular carcinoma of 3 cm or less. Gut. 2005;54(8):1151–1156. doi: 10.1136/gut.2004.045203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Livraghi T, Meloni F, Di Stasi M, 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(1):82–89. doi: 10.1002/hep.21933. [DOI] [PubMed] [Google Scholar]
  • 105.Chen MS, Li JQ, Zheng Y, et al. A prospective randomized trial comparing percutaneous local ablative therapy and partial hepatectomy for small hepatocellular carcinoma. Ann Surg. 2006;243(3):321–328. doi: 10.1097/01.sla.0000201480.65519.b8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lü MD, Kuang M, Liang LJ, et al. Surgical resection versus percutaneous thermal ablation for early–stage hepatocellular carcinoma: a randomized clinical trial [Chinese] Zhonghua Yi Xue Za Zhi. 2006;86(12):801–805. [PubMed] [Google Scholar]
  • 107.Lewandowski RJ, Geschwind JF, Liapi E, Salem R. Transcatheter intraarterial therapies: rationale and overview. Radiology. 2011;259(3):641–657. doi: 10.1148/radiol.11081489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Biolato M, Marrone G, Racco S, et al. Transarterial chemoembolization (TACE) for unresectable HCC: a new life begins? Eur Rev Med Pharmacol Sci. 2010;14(4):356–362. [PubMed] [Google Scholar]
  • 109.Varela M, Real MI, Burrel M, et al. Chemoembolization of hepatocellular carcinoma with drug eluting beads: efficacy and doxorubicin pharmacokinetics. J Hepatol. 2007;46(3):474–481. doi: 10.1016/j.jhep.2006.10.020. [DOI] [PubMed] [Google Scholar]
  • 110.Raoul JL, Guyader D, Bretagne JF, et al. Randomized controlled trial for hepatocellular carcinoma with portal vein thrombosis: intra-arterial iodine-131-iodized oil versus medical support. J Nucl Med. 1994;35(11):1782–1787. [PubMed] [Google Scholar]
  • 111.Kulik LM, Carr BI, Mulcahy MF, et al. Safety and efficacy of 90Y radiotherapy for hepatocellular carcinoma with and without portal vein thrombosis. Hepatology. 2008;47(1):71–81. doi: 10.1002/hep.21980. [DOI] [PubMed] [Google Scholar]
  • 112.Llovet JM, Real MI, Montana X, et al. Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet. 2002;359(9319):1734–1739. doi: 10.1016/S0140-6736(02)08649-X. [DOI] [PubMed] [Google Scholar]
  • 113.Lo CM, Ngan H, Tso WK, et al. Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology. 2002;35(5):1164–1171. doi: 10.1053/jhep.2002.33156. [DOI] [PubMed] [Google Scholar]
  • 114.Dhanasekaran R, Kooby DA, Staley CA, Kauh JS, Khanna V, Kim HS. Comparison of conventional transarterial chemoembolization (TACE) and chemoembolization with doxorubicin drug eluting beads (DEB) for unresectable hepatocelluar carcinoma (HCC) J Surg Oncol. 2010;101(6):476–480. doi: 10.1002/jso.21522. [DOI] [PubMed] [Google Scholar]
  • 115.Lammer J, Malagari K, Vogl T, et al. Prospective randomized study of doxorubicin-eluting-bead embolization in the treatment of hepatocellular carcinoma: results of the PRECISION V study. Cardiovasc Intervent Radiol. 2010;33(1):41–52. doi: 10.1007/s00270-009-9711-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Malagari K, Pomoni M, Kelekis A, et al. Prospective randomized comparison of chemoembolization with doxorubicin-eluting beads and bland embolization with BeadBlock for hepatocellular carcinoma. Cardiovasc Intervent Radiol. 2010;33(3):541–551. doi: 10.1007/s00270-009-9750-0. [DOI] [PubMed] [Google Scholar]
  • 117.Salem R, Lewandowski RJ, Kulik L, et al. Radioembolization results in longer time-to-progression and reduced toxicity compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology. 2011;140(2):497–507. doi: 10.1053/j.gastro.2010.10.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Molinari M, Kachura JR, Dixon E, et al. Transarterial chemoembolisation for advanced hepatocellular carcinoma: results from a North American cancer centre. Clin Oncol (R Coll Radiol) 2006;18(9):684–692. doi: 10.1016/j.clon.2006.07.012. [DOI] [PubMed] [Google Scholar]
  • 119.El-Serag HB, Marrero JA, Rudolph L, Reddy KR. Diagnosis and treatment of hepatocellular carcinoma. Gastroenterology. 2008;134(6):1752–1763. doi: 10.1053/j.gastro.2008.02.090. [DOI] [PubMed] [Google Scholar]
  • 120.Maluccio MA, Covey AM, Porat LB, et al. Transcatheter arterial embolization with only particles for the treatment of unresectable hepatocellular carcinoma. J Vasc Interv Radiol. 2008;19(6):862–869. doi: 10.1016/j.jvir.2008.02.013. [DOI] [PubMed] [Google Scholar]
  • 121.Gish RG, Porta C, Lazar L, et al. Phase III randomized controlled trial comparing the survival of patients with unresectable hepatocellular carcinoma treated with nolatrexed or doxorubicin. J Clin Oncol. 2007;25(21):3069–3075. doi: 10.1200/JCO.2006.08.4046. [DOI] [PubMed] [Google Scholar]
  • 122.Parikh PM, Fuloria J, Babu G, et al. A phase II study of gemcitabine and cisplatin in patients with advanced hepatocellular carcinoma. Trop Gastroenterol. 2005;26(3):115–118. [PubMed] [Google Scholar]
  • 123.Llovet JM, Sala M, Castells L, et al. Randomized controlled trial of interferon treatment for advanced hepatocellular carcinoma. Hepatology. 2000;31(1):54–58. doi: 10.1002/hep.510310111. [DOI] [PubMed] [Google Scholar]
  • 124.Yeo W, Mok TS, Zee B, et al. A randomized phase III study of doxorubicin versus cisplatin/interferon alpha-2b/doxorubicin/fluorouracil (PIAF) combination chemotherapy for unresectable hepatocellular carcinoma. J Natl Cancer Inst. 2005;97(20):1532–1538. doi: 10.1093/jnci/dji315. [DOI] [PubMed] [Google Scholar]
  • 125.Barbare JC, Bouche O, Bonnetain F, et al. Randomized controlled trial of tamoxifen in advanced hepatocellular carcinoma. J Clin Oncol. 2005;23(19):4338–4346. doi: 10.1200/JCO.2005.05.470. [DOI] [PubMed] [Google Scholar]
  • 126.Melia WM, Johnson PJ, Williams R. Controlled clinical trial of doxorubicin and tamoxifen versus doxorubicin alone in hepatocellular carcinoma. Cancer Treat Rep. 1987;71(12):1213–1216. [PubMed] [Google Scholar]
  • 127.Randomized trial of leuprorelin and flutamide in male patients with hepatocellular carcinoma treated with tamoxifen. Hepatology. 2004;40(6):1361–1369. doi: 10.1002/hep.20474. [DOI] [PubMed] [Google Scholar]
  • 128.Becker G, Allgaier HP, Olschewski M, Zahringer A, Blum HE. Long-acting octreotide versus placebo for treatment of advanced HCC: a randomized controlled double-blind study. Hepatology. 2007;45(1):9–15. doi: 10.1002/hep.21468. [DOI] [PubMed] [Google Scholar]
  • 129.Barbare JC, Bouche O, Bonnetain F, et al. Treatment of advanced hepatocellular carcinoma with long-acting octreotide: a phase III multicentre, randomised, double blind placebo-controlled study. Eur J Cancer. 2009;45(10):1788–1797. doi: 10.1016/j.ejca.2009.02.018. [DOI] [PubMed] [Google Scholar]
  • 130.Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
  • 131.Cheng AL, Kang YK, Chen Z, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10(1):25–34. doi: 10.1016/S1470-2045(08)70285-7. [DOI] [PubMed] [Google Scholar]
  • 132.Liu L, Cao Y, Chen C, et al. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 2006;66(24):11851–11858. doi: 10.1158/0008-5472.CAN-06-1377. [DOI] [PubMed] [Google Scholar]
  • 133.Huynh H. Molecularly targeted therapy in hepatocellular carcinoma. Biochem Pharmacol. 2010;80(5):550–560. doi: 10.1016/j.bcp.2010.03.034. [DOI] [PubMed] [Google Scholar]
  • 134.Raoul JL, Santoro A, Beaugrand A, et al. Efficacy and safety of sorafenib in patients with advanced hepatocellular carcinoma according to ECOG performance status: a subanalysis from the SHARP trial. J Clin Oncol. 2008 May 26;(Suppl 20) Abstr 4587. [Google Scholar]
  • 135.Park YN. Update on precursor and early lesions of hepatocellular carcinomas. Arch Pathol Lab Med. 2011;135(6):704–715. doi: 10.5858/2010-0524-RA.1. [DOI] [PubMed] [Google Scholar]
  • 136.Borzio M, Fargion S, Borzio F, et al. Impact of large regenerative, low grade and high grade dysplastic nodules in hepatocellular carcinoma development. J Hepatol. 2003;39(2):208–214. doi: 10.1016/s0168-8278(03)00190-9. [DOI] [PubMed] [Google Scholar]
  • 137.Villanueva A, Minguez B, Forner A, Reig M, Llovet JM. Hepatocellular carcinoma: novel molecular approaches for diagnosis, prognosis, and therapy. Annu Rev Med. 2010;61:317–328. doi: 10.1146/annurev.med.080608.100623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Paradis V, Bieche I, Dargere D, et al. Molecular profiling of hepatocellular carcinomas (HCC) using a large-scale real-time RT-PCR approach: determination of a molecular diagnostic index. Am J Pathol. 2003;163(2):733–741. doi: 10.1016/S0002-9440(10)63700-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Llovet JM, Chen Y, Wurmbach E, et al. A molecular signature to discriminate dysplastic nodules from early hepatocellular carcinoma in HCV cirrhosis. Gastroenterology. 2006;131(6):1758–1767. doi: 10.1053/j.gastro.2006.09.014. [DOI] [PubMed] [Google Scholar]
  • 140.Nam SW, Park JY, Ramasamy A, et al. Molecular changes from dysplastic nodule to hepatocellular carcinoma through gene expression profiling. Hepatology. 2005;42(4):809–818. doi: 10.1002/hep.20878. [DOI] [PubMed] [Google Scholar]
  • 141.Wurmbach E, Chen YB, Khitrov G, et al. Genome-wide molecular profiles of HCV-induced dysplasia and hepatocellular carcinoma. Hepatology. 2007;45(4):938–947. doi: 10.1002/hep.21622. [DOI] [PubMed] [Google Scholar]
  • 142.Di Tommaso L, Franchi G, Park YN, et al. Diagnostic value of HSP70, glypican 3, and glutamine synthetase in hepatocellular nodules in cirrhosis. Hepatology. 2007;45(3):725–734. doi: 10.1002/hep.21531. [DOI] [PubMed] [Google Scholar]
  • 143.Di Tommaso L, Destro A, Seok JY, et al. The application of markers (HSP70 GPC3 and GS) in liver biopsies is useful for detection of hepatocellular carcinoma. J Hepatol. 2009;50(4):746–754. doi: 10.1016/j.jhep.2008.11.014. [DOI] [PubMed] [Google Scholar]
  • 144.Liu H, Li P, Zhai Y, et al. Diagnostic value of glypican-3 in serum and liver for primary hepatocellular carcinoma. World J Gastroenterol. 2010;16(35):4410–4415. doi: 10.3748/wjg.v16.i35.4410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bertino G, Ardiri AM, Calvagno GS, Bertino N, Boemi PM. Prognostic and diagnostic value of des-gamma-carboxy prothrombin in liver cancer. Drug News Perspect. 2010;23(8):498–508. doi: 10.1358/dnp.2010.23.8.1444236. [DOI] [PubMed] [Google Scholar]
  • 146.Toyoda H, Kumada T, Tada T, et al. Clinical utility of highly sensitive Lens culinaris agglutinin-reactive alpha-fetoprotein in hepatocellular carcinoma patients with alpha-fetoprotein < 20 ng/mL. Cancer Sci. 2011;102(5):1025–1031. doi: 10.1111/j.1349-7006.2011.01875.x. [DOI] [PubMed] [Google Scholar]
  • 147.Durazo FA, Blatt LM, Corey WG, et al. Des-gamma-carboxyprothrombin, alpha-fetoprotein and AFP-L3 in patients with chronic hepatitis, cirrhosis and hepatocellular carcinoma. J Gastroenterol Hepatol. 2008;23(10):1541–1548. doi: 10.1111/j.1440-1746.2008.05395.x. [DOI] [PubMed] [Google Scholar]
  • 148.Vandecaveye V, De Keyzer F, Verslype C, et al. Diffusion-weighted MRI provides additional value to conventional dynamic contrast-enhanced MRI for detection of hepatocellular carcinoma. Eur Radiol. 2009;19(10):2456–2466. doi: 10.1007/s00330-009-1431-5. [DOI] [PubMed] [Google Scholar]
  • 149.Tan CH, Low SC, Thng CH. APASL and AASLD Consensus Guidelines on Imaging Diagnosis of Hepatocellular Carcinoma: a review. Int J Hepatol. 2011;2011:1–11. doi: 10.4061/2011/519783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Marin D, Di Martino M, Guerrisi A, et al. Hepatocellular carcinoma in patients with cirrhosis: qualitative comparison of gadobenate dimeglumine-enhanced MR imaging and multiphasic 64-section CT. Radiology. 2009;251(1):85–95. doi: 10.1148/radiol.2511080400. [DOI] [PubMed] [Google Scholar]
  • 151.Di Martino M, Marin D, Guerrisi A, et al. Intraindividual comparison of gadoxetate disodium-enhanced MR imaging and 64-section multidetector CT in the detection of hepatocellular carcinoma in patients with cirrhosis. Radiology. 2010;256(3):806–816. doi: 10.1148/radiol.10091334. [DOI] [PubMed] [Google Scholar]
  • 152.Sun HY, Lee JM, Shin CI, et al. Gadoxetic acid-enhanced magnetic resonance imaging for differentiating small hepatocellular carcinomas (< or = 2 cm in diameter) from arterial enhancing pseudolesions: special emphasis on hepatobiliary phase imaging. Invest Radiol. 2010;45(2):96–103. doi: 10.1097/RLI.0b013e3181c5faf7. [DOI] [PubMed] [Google Scholar]
  • 153.Ahn JH, Yu JS, Hwang SH, Chung JJ, Kim JH, Kim KW. Nontumorous arterioportal shunts in the liver: CT and MRI findings considering mechanisms and fate. Eur Radiol. 2010;20(2):385–394. doi: 10.1007/s00330-009-1542-z. [DOI] [PubMed] [Google Scholar]
  • 154.Khan MA, Combs CS, Brunt EM, et al. Positron emission tomography scanning in the evaluation of hepatocellular carcinoma. J Hepatol. 2000;32(5):792–797. doi: 10.1016/s0168-8278(00)80248-2. [DOI] [PubMed] [Google Scholar]
  • 155.Kim YK, Lee KW, Cho SY, et al. Usefulness 18F-FDG positron emission tomography/computed tomography for detecting recurrence of hepatocellular carcinoma in posttransplant patients. Liver Transpl. 2010;16(6):767–772. doi: 10.1002/lt.22069. [DOI] [PubMed] [Google Scholar]
  • 156.Ho CL, Chen S, Yeung DW, Cheng TK. Dual-tracer PET/CT imaging in evaluation of metastatic hepatocellular carcinoma. J Nucl Med. 2007;48(6):902–909. doi: 10.2967/jnumed.106.036673. [DOI] [PubMed] [Google Scholar]
  • 157.Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45(2):228–247. doi: 10.1016/j.ejca.2008.10.026. [DOI] [PubMed] [Google Scholar]
  • 158.World Health Organization . WHO Handbook for Reporting Results of Cancer Treatment. Geneva: WHO; 1979. [Google Scholar]
  • 159.Forner A, Ayuso C, Varela M, et al. Evaluation of tumor response after locoregional therapies in hepatocellular carcinoma: are response evaluation criteria in solid tumors reliable? Cancer. 2009;115(3):616–623. doi: 10.1002/cncr.24050. [DOI] [PubMed] [Google Scholar]
  • 160.Mannelli L, Kim S, Hajdu CH, Babb JS, Clark TW, Taouli B. Assessment of tumor necrosis of hepatocellular carcinoma after chemoembolization: diffusion-weighted and contrast-enhanced MRI with histopathologic correlation of the explanted liver. AJR Am J Roentgenol. 2009;193(4):1044–1052. doi: 10.2214/AJR.08.1461. [DOI] [PubMed] [Google Scholar]
  • 161.Chen G, Ma DQ, He W, Zhang BF, Zhao LQ. Computed tomography perfusion in evaluating the therapeutic effect of transarterial chemoembolization for hepatocellular carcinoma. World J Gastroenterol. 2008;14(37):5738–5743. doi: 10.3748/wjg.14.5738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Maksimovic O, Schraml C, Hartmann JT, et al. Evaluation of response in malignant tumors treated with the multitargeted tyrosine kinase inhibitor sorafenib: a multitechnique imaging assessment. AJR Am J Roentgenol. 2010;194(1):5–14. doi: 10.2214/AJR.09.2744. [DOI] [PubMed] [Google Scholar]
  • 163.Villanueva A, Llovet JM. Targeted therapies for hepatocellular carcinoma. Gastroenterology. 2011;140(5):1410–1426. doi: 10.1053/j.gastro.2011.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hoshida Y, Nijman SM, Kobayashi M, et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res. 2009;69(18):7385–7392. doi: 10.1158/0008-5472.CAN-09-1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Ji J, Shi J, Budhu A, et al. MicroRNA expression, survival, and response to interferon in liver cancer. N Engl J Med. 2009;361(15):1437–1447. doi: 10.1056/NEJMoa0901282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Roessler S, Jia HL, Budhu A, et al. A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res. 2010;70(24):10202–10212. doi: 10.1158/0008-5472.CAN-10-2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Kew MC. Prevention of hepatocellular carcinoma. Ann Hepatol. 2010;9(2):120–132. [PubMed] [Google Scholar]

Articles from Biologics : Targets & Therapy are provided here courtesy of Dove Press

RESOURCES