Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Apr 21, 2014; 20(15): 4115-4127
Published online Apr 21, 2014. doi: 10.3748/wjg.v20.i15.4115
Hepatocellular carcinoma review: Current treatment, and evidence-based medicine
Ali Raza, Department of Internal Medicine, St. Luke’s Episcopal Hospital, Baylor College of Medicine, Houston, TX 77030, United States
Gagan K Sood, Division of Gastroenterology and Hepatology, Baylor College of Medicine, St. Luke’s Liver Center, Houston, TX 77030, United States
Author contributions: Raza A performed the data research and helped writing the manuscript; Sood GK designed the manuscript; performed the research; helped writing the manuscript and critically analyzed the manuscript.
Correspondence to: Gagan K Sood, MD, Division of Gastroenterology and Hepatology, Baylor College of Medicine, St. Luke’s Liver Center, 6620 Main Street, Suite 1425, Baylor Clinic, Houston, TX 77030, United States. gksood@bcm.edu
Telephone: +1-832-3551400 Fax: +1-713-6102479
Received: September 28, 2013
Revised: December 6, 2013
Accepted: March 6, 2014
Published online: April 21, 2014

Abstract

Hepatocellular carcinoma (HCC) is the fifth most common tumor worldwide. Multiple treatment options are available for HCC including curative resection, liver transplantation, radiofrequency ablation, trans-arterial chemoembolization, radioembolization and systemic targeted agent like sorafenib. The treatment of HCC depends on the tumor stage, patient performance status and liver function reserve and requires a multidisciplinary approach. In the past few years with significant advances in surgical treatments and locoregional therapies, the short-term survival of HCC has improved but the recurrent disease remains a big problem. The pathogenesis of HCC is a multistep and complex process, wherein angiogenesis plays an important role. For patients with advanced disease, sorafenib is the only approved therapy, but novel systemic molecular targeted agents and their combinations are emerging. This article provides an overview of treatment of early and advanced stage HCC based on our extensive review of relevant literature.

Key Words: Hepatocellular carcinoma, Trans-arterial chemoembolization, Drug-eluting beads, Radiofrequency ablation, Liver transplantation, Chemotherapy, Sorafenib, Radioembolization

Core tip: The article discusses the current evidence based treatment of hepatocellular carcinoma. Specific focus is placed on emerging systemic molecular targeted therapies.



INTRODUCTION

Hepatocellular carcinoma (HCC) is the fifth most common form of cancer worldwide and the third most common cause of cancer-related deaths. HCC often occurs in the background of a cirrhotic liver[1]. Orthotopic liver transplantation (OLT) is an effective treatment for both HCC and underlying cirrhosis, and is considered the best therapeutic option. Unfortunately, most cases of HCC present in an advanced stage and are not suitable candidates for OLT[2]. In recent years surveillance strategies in patients at a higher risk of HCC have led to the diagnosis of the disease at much earlier stages. Patients in early stages have a much higher chance of curative response with different treatment options[2,3]. Tumor staging plays an essential role in guiding the treatment decisions, but prognosis is affected by the severity of underlying liver dysfunction. A number of staging systems are available for use in HCC, and there is no worldwide consensus on a preferred system. The Child- Pugh classification system and the model for end-stage liver disease (MELD) score only assess the severity of liver disease and do not include the patient’s performance status (PS) or cancer-related symptoms. The only staging system currently in use that addresses each of these concerns is the Barcelona Clinic Liver Cancer (BCLC) classification. This classification links HCC staging with patient’s PS and co-morbidities. This allows for an appropriate treatment strategy and defines the standard of care for each tumor stage. The major advantage of the BCLC system is that it can be used to identify the patients with early-stage HCC, who may benefit from curative therapies. This differentiates them from the patients with advanced-stage disease who would benefit more from palliative treatment. American Association for the Study of Liver Diseases (AASLD) and European Association for the Study of the Liver (EASL) have endorsed the BCLC system[4,5]. Several therapies have been proposed for these patients with proven survival benefits in the early-stage of HCC. These therapies comprise the surgical resection, various locoregional treatments including percutaneous ethanol injection (PEI), radiofrequency ablation (RFA), trans-arterial chemoembolization (TACE) and radioembolization[4-6].

LIVER TRANSPLANTATION

Liver transplantation (LT) is a potentially curative treatment and the best treatment option for the patients with decompensated cirrhosis. Currently LT is recommended for the patients with HCC, whose tumor is within the Milan criteria for HCC (one lesion not larger than 5 cm, or up to 3 lesions with each 3 cm or smaller). This selection criterion results in a 5-year overall survival rate of 75% and a tumor recurrence rate of less than 15%[7-9]. This tumor burden is compatible with early-stage HCC in the BCLC staging system. Priority for assignment to the LT waiting list is based upon the MELD score, which is a good predictor of early mortality in patients with cirrhosis. However, MELD score is not able to predict mortality in the patient with HCC, therefore, a “MELD exception” has been developed to assign extra points to the HCC patients on the basis of the tumor burden. The exception criteria have resulted in an increased number of LTs being performed in the HCC patients; currently 30%-40% of the LTs are performed for HCC[10]. Some centers also consider the patients for LT who exceed the Milan criteria. Transplanting the patients with HCC beyond the established criteria falls into two categories; those whose tumors exceed the Milan criteria at presentation without any prior treatment (expanded criteria), and those who fulfill the Milan criteria after locoregional treatments (downstaging). Currently, however, there is no international consensus regarding these approaches in clinical practice[11,12]. Evidence for listing the patients for LT with tumor burden beyond Milan criteria is poor. Yet, it is clear that some patients with tumor burden beyond Milan criteria may benefit from transplantation. Similarly, studies looking at the LT outcomes in the patients with HCC after downstaging are very heterogeneous and no evidence-based recommendations can be made at this point. Few studies have shown that successful downstaging of HCC can be achieved in carefully selected patients and is associated with excellent post-transplantation outcomes[13]. Success in downstaging has been reported in many studies, although most of these are uncontrolled observational studies[9,14,15]. Multiple modalities including resection, RFA and TACE have been used for downsizing. The largest experience is with TACE and RFA. The two prospective studies showed that survival after liver transplantation in patients with large tumor burden successfully treated by downstaging was similar to survival in patients who initially met the criteria for LT[16,17]. It is essential to consider how expansion of criteria beyond the Milan criteria might affect the survival of candidates for liver transplantation who do not have HCC. In the European Liver Transplant Registry, Organ Procurement and Transplantation Network, and Australia and New Zealand Liver Transplant Registry, 5-year survival for non-HCC was 65%-87%[18]. According to studies based on Markov models using data from the United States, patients outside the Milan criteria would need to achieve 5-year survival of 60% or higher to prevent a substantial decrement to the life-years available to the entire population of candidates for liver transplantation[19,20]. International consensus conference on recommendations for liver transplantation in 2010 recommended that modest expansion of Milan criteria should be considered[18]. Among many proposals, only the University of California San Francisco criteria (one tumor ≤ 6.5 cm, three nodules at most with the largest ≤ 4.5 cm, and total tumor diameter ≤ 8 cm) have been prospectively validated by the proponent group, with outcome data comparable to those from other retrospective studies[17,21].

A minimum observational period of 3-6 mo after downstaging was required before the LT[17]. It has been recognized that tumor size and number are crude measures of prognosis. In future, studies with molecular markers or gene signatures will define tumor biology and these will be incorporated in the eligibility criteria for the transplant listing[22].

SURGICAL RESECTION

Surgical resection is the treatment option for a small number of patients with single nodules, good liver function and no underlying cirrhosis. Surgical resection has an increased risk of hepatic decompensation in the patients with cirrhosis[23,24]. Thus, only patients with well-compensated cirrhosis, Child-Pugh class A, are considered the ideal candidates for surgical resection. Portal hypertension in cirrhotic patients is considered a relative contraindication for surgical resection according to EASL/AASLD guidelines. In earlier studies Bruix et al[4,25] reported that in Child-Pugh A cirrhotic patients undergoing hepatic resection, the presence of portal hypertension based on hepatic venous pressure gradient (HVPG) ≥ 10 mmHg, to be the best predictor of post-operative liver decompensation and poor long-term outcomes. However, measurement of HVPG is an invasive procedure and requires technical expertise. Some studies have used other surrogate markers of portal hypertension like the presence of esophageal varices or splenomegaly (major diameter > 12 cm) with a platelet count of < 100000/mm3. Few recent studies have reported comparable postoperative and long-term outcome in patients with and without portal hypertension using these surrogate markers of portal hypertension. These studies demonstrated that cirrhotic patients with both clinically significant portal hypertension and well-preserved liver function have similar short- and long-term outcomes compared with patients without portal hypertension. Overall surgical results depend not only on the presence of portal hypertension but also on the residual liver function, size of segmental resection and the remnant liver volume[26,27]. Moreover with improvement in anesthesia and surgical techniques, specifically laparoscopic resection, results of surgery are much superior[28]. Therefore, the prognostic relevance of clinically significant portal hypertension after hepatic resection in patients with HCC is still a matter for debate. The recent study by Santambrogio et al[29] reported that the presence of clinical portal hypertension alone does not influence the post-operative course of cirrhotic patients submitted to hepatic resection. If stringent pre-operative selection criteria are met (i.e., Child-Pugh class A patients undergoing resection with a laparoscopic approach and limited segmental hepatic resection) the post-operative mortality rate is very low.

Patients without portal hypertension or with clinically significant portal hypertension and preserved liver function (Child-Pugh A5 class) can undergo hepatic resection without hepatic decompensation and good long-term survival, if limited hepatic resection with enough remnant liver volume is done with laparoscopic approach.

The patients who undergo surgical resection have nearly 70% five-year survival but have a high risk of recurrence. Recurrence rate correlates with the presence of microscopic vascular invasion, which is present in more than 30% of HCC patients without any evidence of macroscopic vascular invasion[30,31]. Early tumor recurrence within two years of surgery is mainly related to local invasion and intrahepatic metastasis. Late recurrence, occurring after two years of surgery, is mainly related to de novo tumor formation. Some studies have shown benefit of adjuvant therapies in decreasing the postoperative recurrence rate[32-34].

Though the hepatic resection is not often considered as an option in patients with multiple tumors, some centers have reported optimal results with hepatic resections even in patients with multiple tumors.

Some of the biomarkers (gene signatures or molecular biomarkers) are promising in predicting the late recurrence[35]. These biomarkers are likely to improve selection of candidates for surgical resection with lower risk of recurrence.

At present, surgical resection is recommended in the patients with early-stage disease and preserved liver function. But the surgical option should be weighed against the availability and the response rate of other local ablative therapies like radiofrequency ablation.

RFA

Surgical resection is currently considered the most curative strategy, but in the last decade highly satisfactory results have been obtained with local ablative therapies[36]. RFA is currently considered the most effective local ablative therapy. There has been considerable improvement in the RFA technique like the use of expandable-tipped or cool-tip electrodes. A single electrode insertion can produce a necrotic area of up to 3.0 cm in diameter, thus allowing complete ablation of a 2 cm with necrosis of adjacent 0.5 cm to 1.0 cm margins, achieving tumor free margins like surgical resection.

Local ablation with RFA is considered a standard of care for the patients with very early and early stage (BCLC 0-A) tumors not suitable for surgery. The best results were reported for RFA-treated patients with tumors < 2 cm in diameter who had 5-year survival rates ranging from 40% to 70%. A cohort study of RFA demonstrated that complete ablation of lesions smaller than 2 cm is possible in more than 90% of cases, with a local recurrence rate of less than 1%[37].

RFA has replaced percutaneous ethanol ablation as the locoregional therapy of choice. Three independent meta-analyses, including five randomized controlled trials, have provided evidence for better local control and increased survival benefits in patients treated with RFA compared to ablation with PEI. RFA has also been shown to provide a survival benefit in patients with tumors > 2 cm but < 5 cm, as compared to PEI[38-40]. Consequently, RFA has progressively replaced PEI for patients with small HCC who are not candidates for surgery.

There is no consensus so far whether percutaneous RFA can replace surgical resection as first-line treatment for small tumors. Two RCTs provided conflicting evidence regarding the benefits of RFA vs surgical resection. The results from one RCT suggest a benefit for surgery over RFA in patients who met the Milan criteria followed for up to 5 years[41]. Another RCT did not identify a significant difference in survival between RFA and surgery in patients with solitary HCC and a diameter up to 4 cm[42]. At present strong evidence of the superiority or equality of RFA in comparison with surgical resection is lacking, but it is also true that there is no solid evidence that surgical resection is better than RFA for the treatment of small HCC. Studies addressing this issue have been shown in Table 1[41-48].

Table 1 Surgical resection vs radiofrequency ablation.
Ref.5-yr recurrence free survival5-yr overall survival1-yr recurrence-free survival1-yr overall survivalTumor numbern (SR, RFA)Mean tumor size (cm)Patients (n)Study designYear
Hasegawa et al[43]N/ASR: 71.1%RFA: 61.1%N/AN/A1 (83, 73)2 (13%, 20%)3 (3%, 7%)SR: 2.3RFA: 2.0SR: 5361RFA: 5548Cohort study2013
Tohme et al[44]SR: 34%RFA: 28%SR: 47%RFA: 35%SR: 66%RFA: 68%SR: 88%RFA: 86%1 (78, 78)2 (20%, 18%)3 (2%, 3%)SR: 3.07RFA: 2.36SR: 50RFA: 60Cohort study2013
Feng et al[42]N/AN/ASR: 90.6%RFA: 86.2%SR: 96%RFA: 93.1%1 (62, 57)2 (38%, 43%)SR:_SR: 84RFA: 84RCT2012
Huang et al[41]SR: 51.3%RFA: 28.6%SR: 75.6%RFA: 54.7%SR: 85.2%RFA: 81.7%SR: 98.2%RFA: 86.9%1 (77, 73)2 (20%, 26%)3 (3%, 1%) ≤ 5_SR: 115RFA: 115RCT2010
Hasegawa et al[45]N/AN/AN/ASR: 98.3%RFA: 98.5%1 (84, 72)2 (12%, 21%)3 (3%, 7%)SR: 2.2RFA: 2.0SR: 2857RFA: 3022Cohort study2008
Chen et al[46]N/AN/ASR: 86.6%RFA: 85.9%SR: 93.3%RFA: 95.8%1 (100%, 100%) ≤ 5_SR: 90RFA: 71RCT2006
et al[47]N/AN/ASR: 82.4%RFA: 78.5%SR: 91.3%RFA: 93.5%1 (96, 4)> 1 (88%, 12%)3.2_SR: 54RFA: 51RCT2006
Chen et al[48]N/AN/AN/ASR: 93.2%RFA: 92.8%1 (100%, 100%) ≤ 5_SR: 65RFA: 47RCT2005

It is unlikely that any future RCT will address this issue, because it will require very large sample size to show significant differences between two modalities. RFA is less expensive, less invasive, with lower complication rates and shorter hospital stay than surgical resection. In patients with tumor > 3 cm but < 5 cm in size, the success rate of RFA alone is decreased. In these cases combination with TACE could be considered. Recently, the results of an RCT[49] aimed at evaluating the therapeutic efficacy of combining RFA with TACE for treating intermediate-size (3.1-5.0 cm) HCCs have been published. Local tumor progression rate was significantly lower in the TACE and RFA-treated group than in the RFA-only group (6% vs 39%).

RFA should be considered the first option for the treatment of small HCC. However, RFA is size-dependent. RFA can produce a necrotic area of about 4 cm, so it can be effective in HCC measuring up to 3 cm or smaller. In future technical developments allowing achieving ablation areas of 5 cm or more in diameter will make percutaneous thermal ablation an effective alternative to surgery even for tumors measuring 3 cm or more[50].

TACE

TACE is currently considered a standard treatment for the patients with intermediate-stage HCC. Patients with compensated liver function (Child B up to 8 points), with large single nodule (< 5 cm) or multifocal HCC without evidence of vascular invasion or extra hepatic spread are considered candidates for TACE. The recommendation for TACE as the standard of care for intermediate-stage HCC is based on the demonstration of improved survival, as compared with the best supportive care or suboptimal therapies[51]. Llovet et al[52] reported a meta-analysis of six randomized controlled trials, comparing TACE with the best supportive care or suboptimal therapies. There was considerable heterogeneity between the individual study designs (including patient populations and TACE technique), as well as the study results, with only two of the six individual studies reporting 2-year survival with a statistically significant improvement[53,54]. TACE has been reported to achieve a partial response in 15%-62% patients, with significantly delayed tumor progression and improvement in median survival from 16-20 mo[52].

Intermediate-stage HCC includes a heterogeneous population of the patients with variable tumor burden and liver function (Child-Pugh class A or B). The risk of TACE-associated complications may be greater in patients with more extensive disease requiring non-selective embolization, with portal vein thrombosis and with poor residual liver function. The patients with Child-Pugh class C and some with Child-Pugh class B should be excluded from TACE. In the studies reported in literature, MELD score has not been used to select patients. Patients with total bilirubin > 3 mg/dL were excluded. TACE benefits should be balanced with risk of treatment-induced liver failure.

There is lack of standardized therapy regimen for TACE. The optimal schedule, choice of antineoplastic agents (e.g., mitomycin, cisplatin, and doxorubicin alone or in combination), embolizing agent (e.g., gelatin sponge particles or polyvinyl alcohol particles) use of iodized oil, or bland embolization vs chemoembolization, has not been fully established. From a technical point of view, while there is a general consensus about the fact that TACE should be as selective as possible, more standardization of TACE protocols is still needed. Selective TACE comprises the injection of chemotherapeutic agents into the segmental or sub segmental branches feeding the tumors. Golfieri et al[55] compared the effectiveness of selective or super-selective TACE vs standard TACE in determining tumor necrosis in a prospective study of 67 consecutive patients (122 nodules, all < 5 cm). When compared with the standard TACE, selective/super-selective TACE was associated with higher mean levels of necrosis. A direct relationship was reported between the tumor diameter and the mean tumor necrosis level (59.6% for lesions < 2 cm, 68.4% for lesions 2.1-3 cm and 76.2% for lesions > 3 cm). These findings suggest that selective/super selective TACE may determine a higher rate of tumor necrosis than the standard TACE; however, very small nodules (< 2 cm) may not respond as 3-4 cm nodules[56].

The ideal TACE procedure should allow maximum and sustained concentration of chemotherapeutic drug in the tumor, with minimal systemic exposure combined with calibrated tumor vessel obstruction. Lipiodol has been widely adopted in TACE protocols because HCC tumors have great avidity to lipiodol. However there is no data showing that lipiodol allows slow release of chemotherapeutic agents and achieves higher or sustained concentration of chemotherapeutic agents in tumor. One recent survey from multiple eastern and western centers in Europe showed that surgical resection is widely in practice among patients with multinodular, large, and macro-vascular invasive HCC, and provides acceptable short- and long-term results[57,58].

TACE with drug-eluting beads

The recent introduction of embolic microspheres that have the ability to actively sequester doxorubicin hydro-chloride from solution and release it in a controlled fashion has been shown to substantially diminish the amount of chemotherapeutic agent that reaches the systemic circulation, as compared with ethiodized oil-based regimens. This significantly increases the local concentration of the drug and the antitumor efficacy[59].

Recently published results from the PRECISION V trial indicate that TACE with drug-eluting beads is a valuable alternative to ethiodized oil-based conventional TACE. Compared with conventional TACE, the TACE with drug-eluting beads (DEB-TACE) with doxorubicin-eluted beads was associated with improved outcomes[60]. At 6 mo, the DEB-TACE group showed higher rates of complete response, objective response and disease control compared with the conventional TACE group. Although the predefined hypothesis of superiority was not met in the overall population, patients with Child-Pugh B, bi-lobar disease and recurrent disease showed a significant increase in objective response. In addition, DEB-TACE was associated with a reduction in serious liver toxicity and lower rate of doxorubicin-related side effects when compared with the standard TACE[60].

In some patients, there is a risk of systemic toxicity of chemotherapeutic agents used in conventional TACE or DEB-TACE. In these patients bland embolization can be performed. Trans-arterial bland embolization achieves tumor necrosis, but much less compared to DEB-TACE[61].

At present TACE is the standard of care for treating patients with intermediate-stage HCC, but due to heterogeneity of the patient population in this stage, all patients do not achieve the same response. DEB-TACE is preferred over conventional TACE. Repetition of TACE with aggressive schedule increases the adverse events. Repeat TACE should be considered based on objective evidence of tumor progression. Patient at risk of adverse outcome should be identified based on response to first TACE and effect on underlying liver disease. Recently describe ART score may help in identifying patients at high risk for poor outcome after repeated TACE[62,63].

RADIOEMBOLIZATION

Radioembolization or selective internal radiation therapy (SIRT) has recently emerged as a therapeutic option for intermediate-stage HCC and its role in unresectable liver disease is still being refined[56,64-66]. In radioembolization, implantable radioactive microspheres are delivered into the arteries that feed the tumor so that tumor nodules are treated irrespective of their number, size or location. Radioembolization is different from the TACE. In TACE, the embolizing particles or drug eluting particles are usually 100-500 μm in size, which cause ischemia of tumor; but in radioembolization the microspheres are usually smaller (35 μm) in diameter and deliver radiation to tumor without ischemia to the tumor or liver tissue. Currently, the most popular radioembolization technique uses microspheres coated with Y90 b-emitting isotope (TheraSphere and SIR Sphere). The safety of Y90 radioembolization has been documented in phase I and phase II clinical investigations[67]. A few observational studies and retrospective analyses have reported the efficacy of radioembolization in the treatment of HCC[68,69]. Median survivals for intermediate stage HCC, however, vary widely (between 7 and 27 mo) between phase II studies, depending on the PS, extent of the disease and the degree of hepatic functional reserve. Salem et al[70] reported a large prospective study in 291 patients treated with glass-based Y90 microspheres (TheraSphere) showing that liver function and portal vein thrombosis were main predictors of survival. Recently, a comparative analysis of radioembolization or TACE reported fewer side effects, better response rate and longer time to progression (13.3 mo vs 8.4 mo) in radioembolization group, but median survival time was not different (20.5 mo vs 17.5 mo)[68]. In another similar study by European Network on radioembolization with Y90 resin microspheres. Sangro et al[69] reported similar safety profile and response rates. Results of RCTs would provide the highest level of evidence, but based on these studies, it has been estimated that more than 1000 patients would be required to confirm the statistical equivalence or superiority of one treatment over other. Moreover, the relevant cost associated with radioembolization may limit a wide use of this technique. At present radioembolization appears to be safer in more advanced stage HCC including portal vein thrombosis and large tumor burden[69,71,72].

HCC TREATMENT AS A BRIDGE TO TRANSPLANT

Patients with HCC receiving LT within Milan Criteria have a low rate of recurrence and excellent long-term survival.

In recent years, waiting time for LT has progressively increased and despite priority for HCC within the Milan criteria, a significant rate of dropout from the waiting list occurs due to tumor progression. Hence treatment of HCC in patients awaiting LT has become routine, primarily in an effort to prevent tumor progression, reduce dropout rate and to decrease the post-transplant HCC recurrence.

The risk of dropout for HCC within the Milan criteria correlates with the length of waiting time and initial tumor characteristics. In patients initially presenting with solitary HCC < 2 cm, risk of progression is low and only tumors > 2 cm receive priority on waiting list. Hence most transplant centers observe rather than treat these lesions until they grow to 2 cm. The cumulative dropout rates at 6 and 12 mo for patients with single HCC > 3 cm or with 2-3 nodules have been reported 12% and 56% vs 0% and 10% with solitary HCC ≤ 3 cm. These patients are often considered for treatment while awaiting LT.

Chemoembolization, radiofrequency ablation and ethanol injection all are effective in controlling tumor growth; however, there is no high level evidence that these modalities are effective in stopping tumor progression in patients on the waiting list, reducing dropout rate or decreasing post-transplant recurrence. TACE has been widely used, as a bridge to transplant but there is no evidence-based data to support this practice. TACE has not been shown to decrease the dropout rates on waiting list[14,73], but most of the studies addressing this issue were heterogeneous in patient selection, TACE-related protocols and had variable waiting time on LT list. It is unlikely that well-designed RCTs will address this issue in the future. Nevertheless, particularly in the United States, where continued waiting list priority depends on maintaining HCC within Milan criteria, use of nonsurgical HCC treatment will likely continue in an effort to prevent tumor progression and waiting list dropout.

TACE alone or combination with other treatments is recommended to bridge patients to transplant specifically when the waiting list time is more than six mo.

TARGETED SYSTEMIC CHEMOTHERAPY

Hepatocarcinogenesis is the result of genetic alterations affecting multiple signaling cascades resulting in uncontrolled growth of the hepatocytes. Systemic targeted therapies focus on the critical steps of the carcinogenic pathways, limiting widespread systemic toxicity. No single dominant or pathognomic pathway exists in the hepatocarcinogenesis. Overexpression of multiple signaling pathways have been implicated in the pathogenesis of HCC including Vascular endothelial growth factor (VEGF), epidermal growth factor, Ras mitogen-activated protein kinase (MAPK), insulin-like growth factor receptor, hepatocyte growth factor/c-MET, PI3K/PTEN/Akt/mammalian target of rapamycin (mTOR) and Wnt/β-Catenin pathways[74-79]. Targeted molecular agents may block one or more steps in a targeted pathway or potentially more than one pathway to provide suitable results. Currently, sorafenib is approved for the treatment of HCC and represents a paradigm shift in the systemic treatment of HCC, and many new molecular therapies are under investigation.

Sorafenib

Multiple cellular kinases are involved in the development and progression of the HCC by promoting angiogenesis, cellular differentiation, proliferation and survival. Sorafenib is an oral bi-aryl urea, which inhibits multiple cell surface and downstream kinases involved in tumor progression. Cell surface tyrosine kinases inhibited by Sorafenib include VEGF receptor- (VEGFR-) 1, VEGFR-2, VEGFR-3, platelet-derived growth factor receptor- (PDGFR-) β, RET, c-KIT and FMS-like tyrosine kinase-3. Sorafenib also inhibits Ras/MAPK pathway, this pathway involves extracellular signal-regulated kinases and multiple intracellular serine/ threonine kinases including Raf-1 (C-Raf) and B-Raf (wild and mutant-types). Ras/MAPK pathway activation could be due to the mutational activation of Ras oncogene or over expression of surface tyrosine kinases. Overexpression of these kinases is important in HCC proliferation and angiogenesis[80,81]. Two phase III randomized placebo-controlled trials, the SHARP trial conducted mainly in America and Europe[82] and a similar trial conducted in Asia[83] reported improved overall survival with sorafenib. In the SHARP trial, the median overall survival was 10.7 mo with sorafenib and 7.9 mo with placebo. In the Asian study, the median overall survival was 6.5 mo with sorafenib and 4.2 mo with placebo. Sorafenib was generally well tolerated; toxicities were mild to moderate in severity, predominantly including diarrhea, fatigue, and hand-foot skin reaction.

These two, phase III trials have established sorafenib as the preferred systemic therapy for advanced HCC although the role of sorafenib in intermediate HCC is less clear. Moreover, only small numbers of patients with Child-Pugh B have been included in clinical trials, so it is not possible to assess efficacy and safety of sorafenib in this group of patients. Various phase III trials reporting the overall survival in patients with advanced HCC treated with sorafenib, sunitinib, erlotinib, linifanib and brivanib are shown in Table 2.

Table 2 Phase III clinical trials of systemic targeted agents.
Ref.YearPatients (n)Overall survival (mo)
Llovet et al[82]2008Sorafenib: 299Sorafenib: 10.7
(SHARP trial)Placebo: 303Placebo: 7.9
Cheng et al[83]2009Sorafenib: 150Sorafenib: 6.5
(NCT00492752)Placebo: 76Placebo: 4.2
Zhu et al[100]2012Sorafenib: 358Sorafenib: 8.5
(SEARCH trial)Sorafenib +Sorafenib +
Erlotinib: 362Erlotinib: 9.5
Cheng et al[99]2013Sorafenib: 544Sorafenib: 10.2
(SUN1170 trial)Sunitinib: 530Sunitinib: 7.9
Cainap et al[101]2013Sorafenib: N/ASorafenib: 9.8
(LIGHT trial)Linifanib: N/ALinifanib: 9.1
Johnson et al[95]2013Sorafenib: 578Sorafenib: 9.9
(BRISK-FL trial)Brivanib: 577Brivanib: 9.5
Llovet et al[94]2013Brivanib: 263Brivanib: 9.4
(BRISK-PS trial)Placebo: 132Placebo: 8.3

Sorafenib has also been used in combination with other systemic chemotherapeutic agents with a goal to improve efficacy. Sorafenib in combination with doxorubicin[84], octreotide[85] and oxaliplatin[86], tegafur/uracil[87], cisplatin and gemcitabine[88] and AVE 1642 (a human monoclonal antibody inhibiting the insulin-like growth factor-1 receptor)[89] has been used. All of these studies report some survival advantage over sorafenib alone. But most of the studies looking at the combination of sorafenib with other systemic therapies have small sample size. Large randomized double-blind studies are needed to establish the role and toxicity profile of these combination regimens.

Other chemotherapeutic agents

Sunitinib: Sunitinib is a multi-kinase blocker that targets VEGFR and PDGFR. Sunitinib was used in phase II clinical trials for HCC treatment, which led to an open-label phase III trial comparing it with sorafenib[90]. A total of 1073 patients were randomized to receive either sorafenib (544) or sunitinib (529). This trial was terminated early due to increased side effects and futility concerns.

Linifanib: Linifanib is a multi-kinase inhibitor targeting VEGFR and PDGFR along with other kinases. It was found to be effective in the treatment of the HCC with an acceptable safety profile in a single arm phase II clinical trial[91].

Brivanib: Brivanib is a selective inhibitor of fibroblastic growth factor receptor and VEGFR. It showed somewhat promising results in the phase II trials as first line (median overall survival: 10 mo) and second line (median overall survival: 9.5 mo) treatment agent for HCC[92,93]. Brivanib was tried in a phase III BRISK-PS trial as a secondary treatment agent (failed prior systemic treatment due to side effects or progression of the disease) for the treatment of HCC. The median length of overall survival was 9.4 mo for brivanib recipients vs 8.2 mo in the placebo group, which was not statistically significant (P = 0.33)[94]. Another phase III trial, BRISK-FL, compared brivanib with sorafenib as first line treatment agent for HCC[95]. Median survival was 9.5 mo in the brivanib group compared with 9.9 mo in the sorafenib group, which was not statistically significant. Sorafenib was better tolerated than brivanib leading to lesser discontinuation rate (33% vs 43% respectively).

Tivantinib: Tivantinib is an oral MET receptor tyrosine kinase inhibitor. When added to sorafenib, it had synergistic effect against HCC as noted in a phase I clinical trial[96]. In a randomized, placebo-controlled, double-blind, phase II trial, tivantinib was used as a second line agent for the treatment of HCC in previously unresectable HCC who progressed or could not tolerate the first line systemic therapy[97]. The patients were randomly assigned to receive tivantinib (n = 71) or placebo (n = 36). Time to progression of HCC was longer in tivantinib group (1.6 mo) than the placebo group (1.4 mo) (HR = 0.64; P = 0.04). The subgroup of patients who received tivantinib and expressed high tissue MET levels (n = 22) had even longer median time to progression of HCC (2.7 mo). A randomized, double-blinded, controlled phase III study (METIV-HCC trial) is currently underway to determine the efficacy and safety of tivantinib plus sorafenib vs sorafenib alone in the patients with previously unresectable cancer as a first line treatment agent.

Everolimus: Everolimus is an inhibitor of mTOR. A phase I/II single arm trial using everolimus in advanced HCC patients (unresectable) with and without prior systemic therapy for HCC showed that the median progression free survival of 28 patients was 3.8 mo (95%CI: 2.1-4.6) and overall survival was 8.4 mo (95%CI: 3.9-21.1)[98]. And phase III clinical trials of systemic targeted agents is shown in Table 2[82,83,94,95,99-101]. A randomized, double blind, placebo control phase III trial (EVOLVE-1) is underway to assess the role of everolimus in unresectable HCC patients who failed prior treatment with sorafenib.

Sorafenib and TACE combination

As previously discussed, TACE works by blocking the hyper-vascular arterial blood supply of the tumor with the help of an embolic agent and injecting the chemotherapeutic drug. As a result of TACE, a hypoxic environment is created around the surviving tumor cells. Hypoxia stimulates the expression of VEGF and hence the neovascularization of the surviving cells. Sorafenib appears to be a good choice to block the neovascularization at that stage. A phase III trial comparing linifanib to sorafenib as a first line targeted agent has recently been reported[101]. Recently Gadani et al[102] presented their results of a retrospective analysis of 19 patients with Child-Pugh class A and B patients with HCC. Most of the patients were Child-Pugh class A (n = 16) and BCLC stage C (n = 13).

Various studies have looked at the combination of TACE with sorafenib, where sorafenib was introduced few days to weeks after the first TACE (sequential introduction) or it was started prior to the planned TACE and only interrupted for few days around the procedure (interrupted scheduling). There has been reluctance to use combination of TACE and sorafenib due to fear of increased toxicity. In a prospective study patients with unresectable HCC received a combination of sorafenib (started 2-4 wk prior to TACE) and TACE with LC beads[103]. The authors reported safety of concurrent sorafenib and transarterial therapy but without clear benefit of survival.

The efficacy of combination treatment has been assessed in few prospective studies. In a prospective, placebo controlled, randomized, double-blind study Sansonno et al[104] randomized 31 patients with Child-Pugh class A and BCLC-B HCC to receive conventional TACE plus sorafenib and similar number of patients to receive TACE plus placebo. Sorafenib was added 30 d after the first TACE procedure and the patients received more than one TACE procedures. The median time to progression was 9.2 and 4.9 mo in the TACE plus sorafenib and the TACE plus placebo groups respectively.

In another study Kudo et al[105] did not find a difference in overall survival or time to progression benefit with TACE plus sorafenib combination compared with TACE plus placebo. But this effect was likely due to the fact that sorafenib was started late after the first TACE procedure (> 50% of the patients starting it more than 9 wk post-TACE) and there were significant dose reductions and multiple dose interruptions. The START trial[106] was conducted to assess the combination of sorafenib with conventional TACE procedure. One subgroup analysis of the Chinese patients (n = 62) in the START trial was recently published[107]. Patients with unresectable HCC were enrolled and they received conventional TACE and sorafenib 400 mg twice a day. Sorafenib was continued until 4 d prior to the next TACE and was resumed 4 d after TACE procedure for safety reasons. The preliminary results of START indicate concurrent sorafenib and TACE therapy is safe and effective with no unexpected side effects. Similar results were produced in another subgroup analysis of the START trial in Asia-Pacific region, without any un-expected side effects[108]. Currently DEB-TACE has shown superiority over conventional TACE. DEB-TACE in combination with sorafenib has been studied in clinical trails (SPACE, and TACE-2 trials)[109,110]. Recently reported data from the randomized phase II SPACE trial suggest that DEB-TACE in combination with sorafenib met the predefined primary endpoint of improving time to radiologic progression compared with DEB-TACE in combination with placebo[109]. The results of ongoing phase III trials will determine whether there is a role to implement this combination in clinical practice.

The results of concurrent TACE and sorafenib in intermediate stage appear promising but at present it is difficult to recommend combination therapy. There are uncertainties regarding dose, frequency and duration of sorafenib when used in combination with TACE.

Sorafenib and radio-embolization

Several on-going clinical trials are looking at the combination of radio-embolization and sorafenib in patients with HCC. Recently Gadani et al[102] presented their results of a retrospective analysis of patients with Child-Pugh class A and B patients with HCC. The patients were on sorafenib prior to yttrium-90 treatment, which was resumed post- treatment. The overall survival of the patients was higher than the previously reported studies that only used sorafenib. Further prospective studies are being conducted to evaluate the combination of radiation therapy and sorafenib.

CONCLUSION

Management of HCC depends on the tumor stage, liver function reserve, and patient performance status (BCLC stage), and requires a multidisciplinary approach for optimal treatment. LT and hepatic resection are the only curative options in early stage of disease. There have been significant advances in local ablative and trans-arterial therapies. In the early stage HCC, RFA is equivalent to surgical resection in well-selected patients. Drug-eluting beads have improved the efficacy and safety of conventional TACE. Radioembolization with use of resin or glass sphere appear promising. Molecular studies of HCC have identified aberrant activation of different signaling pathways, which represent key targets for novel molecular therapies. For patients with advanced disease, sorafenib is the only approved therapy, but novel targeted agents and their combinations are emerging.

Footnotes

P- Reviewers: Cong WM, Cucchetti A S- Editor: Gou SX L- Editor: A E- Editor: Liu XM

References
1.  El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142:1264-1273.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2183]  [Cited by in F6Publishing: 2332]  [Article Influence: 194.3]  [Reference Citation Analysis (0)]
2.  Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet. 2012;379:1245-1255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3249]  [Cited by in F6Publishing: 3472]  [Article Influence: 289.3]  [Reference Citation Analysis (3)]
3.  Marrero JA. Current Treatment Approaches in HCC. Clin Adv Hematol Oncol. 2013;11 Suppl 5:15-18.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Bruix J, Sherman M. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53:1020-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5972]  [Cited by in F6Publishing: 6326]  [Article Influence: 486.6]  [Reference Citation Analysis (1)]
5.  European Association For The Study Of The Liver, European Organisation For Research And Treatment Of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56:908-943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4059]  [Cited by in F6Publishing: 4336]  [Article Influence: 361.3]  [Reference Citation Analysis (2)]
6.  Lencioni R, Chen XP, Dagher L, Venook AP. Treatment of intermediate/advanced hepatocellular carcinoma in the clinic: how can outcomes be improved? Oncologist. 2010;15 Suppl 4:42-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 117]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
7.  Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, Montalto F, Ammatuna M, Morabito A, Gennari L. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996;334:693-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5110]  [Cited by in F6Publishing: 4991]  [Article Influence: 178.3]  [Reference Citation Analysis (0)]
8.  Mazzaferro V. Results of liver transplantation: with or without Milan criteria? Liver Transpl. 2007;13:S44-S47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
9.  Mazzaferro V, Bhoori S, Sposito C, Bongini M, Langer M, Miceli R, Mariani L. Milan criteria in liver transplantation for hepatocellular carcinoma: an evidence-based analysis of 15 years of experience. Liver Transpl. 2011;17 Suppl 2:S44-S57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 384]  [Cited by in F6Publishing: 399]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
10.  Taniguchi M. Liver transplantation in the MELD era--analysis of the OPTN/UNOS registry. Clin Transpl. 2012;41-65.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Prasad KR, Young RS, Burra P, Zheng SS, Mazzaferro V, Moon DB, Freeman RB. Summary of candidate selection and expanded criteria for liver transplantation for hepatocellular carcinoma: a review and consensus statement. Liver Transpl. 2011;17 Suppl 2:S81-S89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
12.  Washburn K, Halff G. Hepatocellular carcinoma and liver transplantation. Curr Opin Organ Transplant. 2011;16:297-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
13.  Gordon-Weeks AN, Snaith A, Petrinic T, Friend PJ, Burls A, Silva MA. Systematic review of outcome of downstaging hepatocellular cancer before liver transplantation in patients outside the Milan criteria. Br J Surg. 2011;98:1201-1208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 75]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
14.  Cescon M, Cucchetti A, Ravaioli M, Pinna AD. Hepatocellular carcinoma locoregional therapies for patients in the waiting list. Impact on transplantability and recurrence rate. J Hepatol. 2013;58:609-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 108]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
15.  Mazzaferro V, Llovet JM, Miceli R, Bhoori S, Schiavo M, Mariani L, Camerini T, Roayaie S, Schwartz ME, Grazi GL. Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet Oncol. 2009;10:35-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1267]  [Cited by in F6Publishing: 1421]  [Article Influence: 88.8]  [Reference Citation Analysis (1)]
16.  Cucchetti A, Cescon M, Bigonzi E, Piscaglia F, Golfieri R, Ercolani G, Cristina Morelli M, Ravaioli M, Daniele Pinna A. Priority of candidates with hepatocellular carcinoma awaiting liver transplantation can be reduced after successful bridge therapy. Liver Transpl. 2011;17:1344-1354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 77]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
17.  Yao FY, Kerlan RK, Hirose R, Davern TJ, Bass NM, Feng S, Peters M, Terrault N, Freise CE, Ascher NL. Excellent outcome following down-staging of hepatocellular carcinoma prior to liver transplantation: an intention-to-treat analysis. Hepatology. 2008;48:819-827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 386]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
18.  Clavien PA, Lesurtel M, Bossuyt PM, Gores GJ, Langer B, Perrier A. Recommendations for liver transplantation for hepatocellular carcinoma: an international consensus conference report. Lancet Oncol. 2012;13:e11-e22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 673]  [Cited by in F6Publishing: 728]  [Article Influence: 56.0]  [Reference Citation Analysis (1)]
19.  Vitale A, Volk ML, De Feo TM, Burra P, Frigo AC, Ramirez Morales R, De Carlis L, Belli L, Colledan M, Fagiuoli S. A method for establishing allocation equity among patients with and without hepatocellular carcinoma on a common liver transplant waiting list. J Hepatol. 2014;60:290-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 80]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
20.  Volk ML, Vijan S, Marrero JA. A novel model measuring the harm of transplanting hepatocellular carcinoma exceeding Milan criteria. Am J Transplant. 2008;8:839-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 150]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
21.  Ravaioli M, Grazi GL, Piscaglia F, Trevisani F, Cescon M, Ercolani G, Vivarelli M, Golfieri R, D’Errico Grigioni A, Panzini I. Liver transplantation for hepatocellular carcinoma: results of down-staging in patients initially outside the Milan selection criteria. Am J Transplant. 2008;8:2547-2557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 271]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
22.  Schwartz M, Dvorchik I, Roayaie S, Fiel MI, Finkelstein S, Marsh JW, Martignetti JA, Llovet JM. Liver transplantation for hepatocellular carcinoma: extension of indications based on molecular markers. J Hepatol. 2008;49:581-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 72]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
23.  Bhoori S, Schiavo M, Russo A, Mazzaferro V. First-line treatment for hepatocellular carcinoma: resection or transplantation? Transplant Proc. 2007;39:2271-2273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
24.  Beard RE, Hanto DW, Gautam S, Miksad RA. A comparison of surgical outcomes for noncirrhotic and cirrhotic hepatocellular carcinoma patients in a Western institution. Surgery. 2013;154:545-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
25.  Bruix J, Castells A, Bosch J, Feu F, Fuster J, Garcia-Pagan JC, Visa J, Bru C, Rodés J. Surgical resection of hepatocellular carcinoma in cirrhotic patients: prognostic value of preoperative portal pressure. Gastroenterology. 1996;111:1018-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 656]  [Cited by in F6Publishing: 590]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
26.  Ruzzenente A, Valdegamberi A, Campagnaro T, Conci S, Pachera S, Iacono C, Guglielmi A. Hepatocellular carcinoma in cirrhotic patients with portal hypertension: is liver resection always contraindicated? World J Gastroenterol. 2011;17:5083-5088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
27.  Kawano Y, Sasaki A, Kai S, Endo Y, Iwaki K, Uchida H, Shibata K, Ohta M, Kitano S. Short- and long-term outcomes after hepatic resection for hepatocellular carcinoma with concomitant esophageal varices in patients with cirrhosis. Ann Surg Oncol. 2008;15:1670-1676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 92]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
28.  Cucchetti A, Ercolani G, Vivarelli M, Cescon M, Ravaioli M, Ramacciato G, Grazi GL, Pinna AD. Is portal hypertension a contraindication to hepatic resection? Ann Surg. 2009;250:922-928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 155]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
29.  Santambrogio R, Kluger MD, Costa M, Belli A, Barabino M, Laurent A, Opocher E, Azoulay D, Cherqui D. Hepatic resection for hepatocellular carcinoma in patients with Child-Pugh’s A cirrhosis: is clinical evidence of portal hypertension a contraindication? HPB (Oxford). 2013;15:78-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 109]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
30.  Barbier L, Fuks D, Pessaux P, Muscari F, Le Treut YP, Faivre S, Belghiti J. Safety of liver resection for hepatocellular carcinoma after sorafenib therapy: a multicenter case-matched study. Ann Surg Oncol. 2013;20:3603-3609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
31.  Wong TC, Lo CM. Resection strategies for hepatocellular carcinoma. Semin Liver Dis. 2013;33:273-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
32.  Cucchetti A, Piscaglia F, Caturelli E, Benvegnù L, Vivarelli M, Ercolani G, Cescon M, Ravaioli M, Grazi GL, Bolondi L. Comparison of recurrence of hepatocellular carcinoma after resection in patients with cirrhosis to its occurrence in a surveilled cirrhotic population. Ann Surg Oncol. 2009;16:413-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 138]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
33.  Mazzaferro V, Romito R, Schiavo M, Mariani L, Camerini T, Bhoori S, Capussotti L, Calise F, Pellicci R, Belli G. Prevention of hepatocellular carcinoma recurrence with alpha-interferon after liver resection in HCV cirrhosis. Hepatology. 2006;44:1543-1554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 279]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
34.  Xia Y, Qiu Y, Li J, Shi L, Wang K, Xi T, Shen F, Yan Z, Wu M. Adjuvant therapy with capecitabine postpones recurrence of hepatocellular carcinoma after curative resection: a randomized controlled trial. Ann Surg Oncol. 2010;17:3137-3144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
35.  Nault JC, De Reyniès A, Villanueva A, Calderaro J, Rebouissou S, Couchy G, Decaens T, Franco D, Imbeaud S, Rousseau F. A hepatocellular carcinoma 5-gene score associated with survival of patients after liver resection. Gastroenterology. 2013;145:176-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 247]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
36.  Nishikawa H, Kimura T, Kita R, Osaki Y. Radiofrequency ablation for hepatocellular carcinoma. Int J Hyperthermia. 2013;29:558-568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
37.  Livraghi T, Meloni F, Di Stasi M, Rolle E, Solbiati L, Tinelli C, Rossi S. Sustained complete response and complications rates after radiofrequency ablation of very early hepatocellular carcinoma in cirrhosis: Is resection still the treatment of choice? Hepatology. 2008;47:82-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 774]  [Cited by in F6Publishing: 783]  [Article Influence: 48.9]  [Reference Citation Analysis (0)]
38.  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;104:514-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 193]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
39.  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;49:453-459.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 328]  [Cited by in F6Publishing: 360]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
40.  Shen A, Zhang H, Tang C, Chen Y, Wang Y, Zhang C, Wu Z. Systematic review of radiofrequency ablation versus percutaneous ethanol injection for small hepatocellular carcinoma up to 3 cm. J Gastroenterol Hepatol. 2013;28:793-800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
41.  Huang J, Yan L, Cheng Z, Wu H, Du L, Wang J, Xu Y, Zeng Y. A randomized trial comparing radiofrequency ablation and surgical resection for HCC conforming to the Milan criteria. Ann Surg. 2010;252:903-912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 580]  [Cited by in F6Publishing: 588]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
42.  Feng K, Yan J, Li X, Xia F, Ma K, Wang S, Bie P, Dong J. A randomized controlled trial of radiofrequency ablation and surgical resection in the treatment of small hepatocellular carcinoma. J Hepatol. 2012;57:794-802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 471]  [Cited by in F6Publishing: 529]  [Article Influence: 44.1]  [Reference Citation Analysis (0)]
43.  Hasegawa K, Kokudo N, Makuuchi M, Izumi N, Ichida T, Kudo M, Ku Y, Sakamoto M, Nakashima O, Matsui O. Comparison of resection and ablation for hepatocellular carcinoma: a cohort study based on a Japanese nationwide survey. J Hepatol. 2013;58:724-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 274]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
44.  Tohme S, Geller DA, Cardinal JS, Chen HW, Packiam V, Reddy S, Steel J, Marsh JW, Tsung A. Radiofrequency ablation compared to resection in early-stage hepatocellular carcinoma. HPB (Oxford). 2013;15:210-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
45.  Hasegawa K, Makuuchi M, Takayama T, Kokudo N, Arii S, Okazaki M, Okita K, Omata M, Kudo M, Kojiro M. Surgical resection vs. percutaneous ablation for hepatocellular carcinoma: a preliminary report of the Japanese nationwide survey. J Hepatol. 2008;49:589-594.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Chen MS, Li JQ, Zheng Y, Guo RP, Liang HH, Zhang YQ, Lin XJ, Lau WY. A prospective randomized trial comparing percutaneous local ablative therapy and partial hepatectomy for small hepatocellular carcinoma. Ann Surg. 2006;243:321-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1006]  [Cited by in F6Publishing: 1039]  [Article Influence: 57.7]  [Reference Citation Analysis (0)]
47.  Lü MD, Kuang M, Liang LJ, Xie XY, Peng BG, Liu GJ, Li DM, Lai JM, Li SQ. [Surgical resection versus percutaneous thermal ablation for early-stage hepatocellular carcinoma: a randomized clinical trial]. Zhonghua Yixue Zazhi. 2006;86:801-805.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Chen MS, Li JQ, Liang HH, Lin XJ, Guo RP, Zheng Y, Zhang YQ. [Comparison of effects of percutaneous radiofrequency ablation and surgical resection on small hepatocellular carcinoma]. Zhonghua Yixue Zazhi. 2005;85:80-83.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Morimoto M, Numata K, Kondou M, Nozaki A, Morita S, Tanaka K. Midterm outcomes in patients with intermediate-sized hepatocellular carcinoma: a randomized controlled trial for determining the efficacy of radiofrequency ablation combined with transcatheter arterial chemoembolization. Cancer. 2010;116:5452-5460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 239]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
50.  Cucchetti A, Piscaglia F, Cescon M, Colecchia A, Ercolani G, Bolondi L, Pinna AD. Cost-effectiveness of hepatic resection versus percutaneous radiofrequency ablation for early hepatocellular carcinoma. J Hepatol. 2013;59:300-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 277]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
51.  Forner A, Llovet JM, Bruix J. Chemoembolization for intermediate HCC: is there proof of survival benefit? J Hepatol. 2012;56:984-986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 94]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
52.  Llovet JM, Bruix J. Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology. 2003;37:429-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2207]  [Cited by in F6Publishing: 2176]  [Article Influence: 103.6]  [Reference Citation Analysis (0)]
53.  Llovet JM, Real MI, Montaña X, Planas R, Coll S, Aponte J, Ayuso C, Sala M, Muchart J, Solà R. Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet. 2002;359:1734-1739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2502]  [Cited by in F6Publishing: 2474]  [Article Influence: 112.5]  [Reference Citation Analysis (0)]
54.  Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, Poon RT, Fan ST, Wong J. Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology. 2002;35:1164-1171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1904]  [Cited by in F6Publishing: 1900]  [Article Influence: 86.4]  [Reference Citation Analysis (0)]
55.  Golfieri R, Cappelli A, Cucchetti A, Piscaglia F, Carpenzano M, Peri E, Ravaioli M, D’Errico-Grigioni A, Pinna AD, Bolondi L. Efficacy of selective transarterial chemoembolization in inducing tumor necrosis in small (& lt; 5 cm) hepatocellular carcinomas. Hepatology. 2011;53:1580-1589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 201]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
56.  Golfieri R, Bilbao JI, Carpanese L, Cianni R, Gasparini D, Ezziddin S, Paprottka PM, Fiore F, Cappelli A, Rodriguez M. Comparison of the survival and tolerability of radioembolization in elderly vs. younger patients with unresectable hepatocellular carcinoma. J Hepatol. 2013;59:753-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 77]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
57.  Ishizawa T, Hasegawa K, Aoki T, Takahashi M, Inoue Y, Sano K, Imamura H, Sugawara Y, Kokudo N, Makuuchi M. Neither multiple tumors nor portal hypertension are surgical contraindications for hepatocellular carcinoma. Gastroenterology. 2008;134:1908-1916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 513]  [Cited by in F6Publishing: 543]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
58.  Torzilli G, Belghiti J, Kokudo N, Takayama T, Capussotti L, Nuzzo G, Vauthey JN, Choti MA, De Santibanes E, Donadon M. A snapshot of the effective indications and results of surgery for hepatocellular carcinoma in tertiary referral centers: is it adherent to the EASL/AASLD recommendations?: an observational study of the HCC East-West study group. Ann Surg. 2013;257:929-937.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 387]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
59.  Song MJ, Chun HJ, Song do S, Kim HY, Yoo SH, Park CH, Bae SH, Choi JY, Chang UI, Yang JM. Comparative study between doxorubicin-eluting beads and conventional transarterial chemoembolization for treatment of hepatocellular carcinoma. J Hepatol. 2012;57:1244-1250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 179]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
60.  Lammer J, Malagari K, Vogl T, Pilleul F, Denys A, Watkinson A, Pitton M, Sergent G, Pfammatter T, Terraz S. Prospective randomized study of doxorubicin-eluting-bead embolization in the treatment of hepatocellular carcinoma: results of the PRECISION V study. Cardiovasc Intervent Radiol. 2010;33:41-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1063]  [Cited by in F6Publishing: 1115]  [Article Influence: 74.3]  [Reference Citation Analysis (0)]
61.  Nicolini D, Svegliati-Baroni G, Candelari R, Mincarelli C, Mandolesi A, Bearzi I, Mocchegiani F, Vecchi A, Montalti R, Benedetti A. Doxorubicin-eluting bead vs conventional transcatheter arterial chemoembolization for hepatocellular carcinoma before liver transplantation. World J Gastroenterol. 2013;19:5622-5632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 45]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
62.  Hucke F, Sieghart W, Pinter M, Graziadei I, Vogel W, Müller C, Heinzl H, Waneck F, Trauner M, Peck-Radosavljevic M. The ART-strategy: sequential assessment of the ART score predicts outcome of patients with hepatocellular carcinoma re-treated with TACE. J Hepatol. 2014;60:118-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 94]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
63.  Sieghart W, Hucke F, Pinter M, Graziadei I, Vogel W, Müller C, Heinzl H, Trauner M, Peck-Radosavljevic M. The ART of decision making: retreatment with transarterial chemoembolization in patients with hepatocellular carcinoma. Hepatology. 2013;57:2261-2273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 253]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
64.  Sangro B, Carpanese L, Cianni R, Golfieri R, Gasparini D, Ezziddin S, Paprottka PM, Fiore F, Van Buskirk M, Bilbao JI. Survival after yttrium-90 resin microsphere radioembolization of hepatocellular carcinoma across Barcelona clinic liver cancer stages: a European evaluation. Hepatology. 2011;54:868-878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 476]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
65.  Lau WY, Sangro B, Chen PJ, Cheng SQ, Chow P, Lee RC, Leung T, Han KH, Poon RT. Treatment for hepatocellular carcinoma with portal vein tumor thrombosis: the emerging role for radioembolization using yttrium-90. Oncology. 2013;84:311-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 119]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
66.  Shah RP, Brown KT, Sofocleous CT. Arterially directed therapies for hepatocellular carcinoma. AJR Am J Roentgenol. 2011;197:W590-W602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
67.  Mazzaferro V, Sposito C, Bhoori S, Romito R, Chiesa C, Morosi C, Maccauro M, Marchianò A, Bongini M, Lanocita R. Yttrium-90 radioembolization for intermediate-advanced hepatocellular carcinoma: a phase 2 study. Hepatology. 2013;57:1826-1837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 373]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
68.  Salem R, Lewandowski RJ, Kulik L, Wang E, Riaz A, Ryu RK, Sato KT, Gupta R, Nikolaidis P, Miller FH. Radioembolization results in longer time-to-progression and reduced toxicity compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology. 2011;140:497-507.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 451]  [Cited by in F6Publishing: 473]  [Article Influence: 36.4]  [Reference Citation Analysis (0)]
69.  Sangro B, Iñarrairaegui M, Bilbao JI. Radioembolization for hepatocellular carcinoma. J Hepatol. 2012;56:464-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 216]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
70.  Salem R, Lewandowski RJ, Mulcahy MF, Riaz A, Ryu RK, Ibrahim S, Atassi B, Baker T, Gates V, Miller FH. Radioembolization for hepatocellular carcinoma using Yttrium-90 microspheres: a comprehensive report of long-term outcomes. Gastroenterology. 2010;138:52-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 738]  [Cited by in F6Publishing: 728]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
71.  Salem R, Mazzaferro V, Sangro B. Yttrium 90 radioembolization for the treatment of hepatocellular carcinoma: biological lessons, current challenges, and clinical perspectives. Hepatology. 2013;58:2188-2197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 134]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
72.  Kim YH, Kim do Y. Yttrium-90 radioembolization for hepatocellular carcinoma: what we know and what we need to know. Oncology. 2013;84 Suppl 1:34-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
73.  Lesurtel M, Müllhaupt B, Pestalozzi BC, Pfammatter T, Clavien PA. Transarterial chemoembolization as a bridge to liver transplantation for hepatocellular carcinoma: an evidence-based analysis. Am J Transplant. 2006;6:2644-2650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 138]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
74.  Chiang DY, Villanueva A, Hoshida Y, Peix J, Newell P, Minguez B, LeBlanc AC, Donovan DJ, Thung SN, Solé M. Focal gains of VEGFA and molecular classification of hepatocellular carcinoma. Cancer Res. 2008;68:6779-6788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 509]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
75.  Ito Y, Takeda T, Sakon M, Tsujimoto M, Higashiyama S, Noda K, Miyoshi E, Monden M, Matsuura N. Expression and clinical significance of erb-B receptor family in hepatocellular carcinoma. Br J Cancer. 2001;84:1377-1383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 216]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
76.  Calvisi DF, Ladu S, Gorden A, Farina M, Conner EA, Lee JS, Factor VM, Thorgeirsson SS. Ubiquitous activation of Ras and Jak/Stat pathways in human HCC. Gastroenterology. 2006;130:1117-1128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 538]  [Cited by in F6Publishing: 528]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
77.  Sahin F, Kannangai R, Adegbola O, Wang J, Su G, Torbenson M. mTOR and P70 S6 kinase expression in primary liver neoplasms. Clin Cancer Res. 2004;10:8421-8425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 286]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
78.  Takami T, Kaposi-Novak P, Uchida K, Gomez-Quiroz LE, Conner EA, Factor VM, Thorgeirsson SS. Loss of hepatocyte growth factor/c-Met signaling pathway accelerates early stages of N-nitrosodiethylamine induced hepatocarcinogenesis. Cancer Res. 2007;67:9844-9851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 75]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
79.  Breuhahn K, Longerich T, Schirmacher P. Dysregulation of growth factor signaling in human hepatocellular carcinoma. Oncogene. 2006;25:3787-3800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 432]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
80.  Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, Schwartz B, Simantov R, Kelley S. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov. 2006;5:835-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1230]  [Cited by in F6Publishing: 1280]  [Article Influence: 71.1]  [Reference Citation Analysis (0)]
81.  Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004;64:7099-7109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2966]  [Cited by in F6Publishing: 3027]  [Article Influence: 151.4]  [Reference Citation Analysis (0)]
82.  Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9016]  [Cited by in F6Publishing: 9474]  [Article Influence: 592.1]  [Reference Citation Analysis (1)]
83.  Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, Luo R, Feng J, Ye S, Yang TS. 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;10:25-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3854]  [Cited by in F6Publishing: 4327]  [Article Influence: 270.4]  [Reference Citation Analysis (0)]
84.  Abou-Alfa GK, Johnson P, Knox JJ, Capanu M, Davidenko I, Lacava J, Leung T, Gansukh B, Saltz LB. Doxorubicin plus sorafenib vs doxorubicin alone in patients with advanced hepatocellular carcinoma: a randomized trial. JAMA. 2010;304:2154-2160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 327]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
85.  Prete SD, Montella L, Caraglia M, Maiorino L, Cennamo G, Montesarchio V, Piai G, Febbraro A, Tarantino L, Capasso E. Sorafenib plus octreotide is an effective and safe treatment in advanced hepatocellular carcinoma: multicenter phase II So.LAR. study. Cancer Chemother Pharmacol. 2010;66:837-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
86.  Yau TCP, Chan P, Cheung FY, Lee AS, Yau TK, Choo SP, Lau J, Wong JS, Fan ST, Poon RT. Phase II trial of sorafenib with capecitabine and oxaliplatin (SECOX) in patients with locally advanced or metastatic hepatocellular carcinoma. EJC. 2009;Suppl 7:20-21.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Hsu CH, Shen YC, Lin ZZ, Chen PJ, Shao YY, Ding YH, Hsu C, Cheng AL. Phase II study of combining sorafenib with metronomic tegafur/uracil for advanced hepatocellular carcinoma. J Hepatol. 2010;53:126-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 105]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
88.  Giuliana FAR, Addeo R, Febbraio A, Rizzi D, Macello E, del Prete S, Pisconti S, Fico M, Colucci G. Sorafenib plus cisplatin and gemcitabine in the treatment of advanced hepatocellular carcinoma: a phase II study by the Grupo Oncologico Dell’Italia Meridonale. (PROT. GOIM 2705). Cancer Treat Rev. 2010;36 Suppl 4:S96.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Faivre SFL, Fartoux L, Bumsel F, Bouattor M, Dreyer C, Raymond E, Rosmorduc O. Phase I safety, pharmacokinetic, and pharmacodynamic study of AVE 1642, a human monoclonal antibody inhibiting the insulin-like grouth factor-1 receptor (IGF-1R/CD221), administered as single agent and in combination with sorafinib as first line therapy in patients with advanced hepatocellular carcinoma. Hepatology. 2010;52 Suppl:466A, 288.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Cheng A, Kang Y, Lin D, Park J, Kudo M, Qin S, Omata M, Lowenthal SW, Lanzalone S, Yang L, Lechuga M, Raymond E and SUN1170 HCC Study Group. Phase III trial of sunitinib (Su) versus sorafenib (So) in advanced hepatocellular carcinoma (HCC). J Clin Oncol. 2011;29 Suppl 15:4000.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Toh HC, Chen PJ, Carr BI, Knox JJ, Gill S, Ansell P, McKeegan EM, Dowell B, Pedersen M, Qin Q. Phase 2 trial of linifanib (ABT-869) in patients with unresectable or metastatic hepatocellular carcinoma. Cancer. 2013;119:380-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 87]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
92.  Park JW, Finn RS, Kim JS, Karwal M, Li RK, Ismail F, Thomas M, Harris R, Baudelet C, Walters I. Phase II, open-label study of brivanib as first-line therapy in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2011;17:1973-1983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 132]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
93.  Finn RS, Kang YK, Mulcahy M, Polite BN, Lim HY, Walters I, Baudelet C, Manekas D, Park JW. Phase II, open-label study of brivanib as second-line therapy in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2012;18:2090-2098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 124]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
94.  Llovet JM, Decaens T, Raoul JL, Boucher E, Kudo M, Chang C, Kang YK, Assenat E, Lim HY, Boige V. Brivanib versus placebo in patients with advanced hepatocellular carcinoma (HCC) who failed or were intolerant to sorafenib: results from the phase 3 BRISK-PS study. EASL. 2012;Abstract 1398.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Johnson PJ, Qin S, Park JW, Poon RT, Raoul JL, Philip PA, Hsu CH, Hu TH, Heo J, Xu J. Brivanib versus sorafenib as first-line therapy in patients with unresectable, advanced hepatocellular carcinoma: results from the randomized phase III BRISK-FL study. J Clin Oncol. 2013;31:3517-3524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 557]  [Cited by in F6Publishing: 564]  [Article Influence: 51.3]  [Reference Citation Analysis (0)]
96.  Martell RE, Puzanov I, Ma WW, Santoro A, Dy GK, Goff LW, Fetterly GJ, Michael SA, Means-Powell JA, Chai F. Safety and efficacy of MET inhibitor tivantinib (ARQ 197) combined with sorafenib in patients (pts) with hepatocellular carcinoma (HCC) from a phase I study. Proc Am Soc Clin Oncol. 2012;30 Suppl:abstract 4117.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Santoro A, Rimassa L, Borbath I, Daniele B, Salvagni S, Van Laethem JL, Van Vlierberghe H, Trojan J, Kolligs FT, Weiss A. Tivantinib for second-line treatment of advanced hepatocellular carcinoma: a randomised, placebo-controlled phase 2 study. Lancet Oncol. 2013;14:55-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 474]  [Article Influence: 43.1]  [Reference Citation Analysis (0)]
98.  Zhu AX, Abrams TA, Miksad R, Blaszkowsky LS, Meyerhardt JA, Zheng H, Muzikansky A, Clark JW, Kwak EL, Schrag D. Phase 1/2 study of everolimus in advanced hepatocellular carcinoma. Cancer. 2011;117:5094-5102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 155]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
99.  Cheng AL, Kang YK, Lin DY, Park JW, Kudo M, Qin S, Chung HC, Song X, Xu J, Poggi G. Sunitinib versus sorafenib in advanced hepatocellular cancer: results of a randomized phase III trial. J Clin Oncol. 2013;31:4067-4075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 523]  [Cited by in F6Publishing: 563]  [Article Influence: 51.2]  [Reference Citation Analysis (0)]
100.  Zhu AX, Rosmorduc O, Evans J, Ross P, Santoro A, Carriho FJ, Leberre M, Jensen M, Meinhardt G, Kang Y. SEARCH: a phase III, randomized, double-blind, placebo-controlled trial of sorafenib plus erlotinib in patients with hepatocellular carcinoma (HCC). Ann Oncol. 2012;23 Suppl:LBA2.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Cainap C, Qin S, Huang WT, Chung IJ, Pan H, Cheng Y, Kudo M, Kang YK, PChen PJ, Toh HC. Phase III trial of linifanib versus sorafenib in patients with advanced hepatocellular carcinoma (HCC). J Clin Oncol. 2012;30:abstr 249.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Gadani S, Mahvash A, Avritscher R, Chasen B, Kaseb A, Murthy R.  Yttirum-90 resin microspheres as an adjunct to sorafenib in patients with unresectable HCC: A retrospective study for evaluation of survival benefit and adverse events. Society of Interventional Radiology Annual Scientific Meeting. Presented at: April 15 2013; Abstract 62.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Cabrera R, Pannu DS, Caridi J, Firpi RJ, Soldevila-Pico C, Morelli G, Clark V, Suman A, George TJ, Nelson DR. The combination of sorafenib with transarterial chemoembolisation for hepatocellular carcinoma. Aliment Pharmacol Ther. 2011;34:205-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 77]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
104.  Sansonno D, Lauletta G, Russi S, Conteduca V, Sansonno L, Dammacco F. Transarterial chemoembolization plus sorafenib: a sequential therapeutic scheme for HCV-related intermediate-stage hepatocellular carcinoma: a randomized clinical trial. Oncologist. 2012;17:359-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 127]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
105.  Kudo M, Imanaka K, Chida N, Nakachi K, Tak WY, Takayama T, Yoon JH, Hori T, Kumada H, Hayashi N. Phase III study of sorafenib after transarterial chemoembolisation in Japanese and Korean patients with unresectable hepatocellular carcinoma. Eur J Cancer. 2011;47:2117-2127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 417]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
106.  Chao Y, Lee HC, Lee TY, Yoon J, Han G, Yang J, Wang J, Kim B, Shao G, Chung YH.  START (Study in Asia of the Combination of TACE (transcatheter arterial chemoembolization) with Sorafenib in Patients with Hepatocellular Carcinoma) Trial: 5th International Liver Cancer Association (ILCA) Annual Conference. Hong Kong: Wanchai; Abstract 0-Abstract 026.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Han G, Yang J, Shao G, Teng G, Wang M, Yang J, Liu Z, Feng G, Yang R, Lu L. Sorafenib in combination with transarterial chemoembolization in Chinese patients with hepatocellular carcinoma: a subgroup interim analysis of the START trial. Future Oncol. 2013;9:403-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
108.  Chung YH, Han G, Yoon JH, Yang J, Wang J, Shao GL, Kim BI, Lee TY, Chao Y. Interim analysis of START: Study in Asia of the combination of TACE (transcatheter arterial chemoembolization) with sorafenib in patients with hepatocellular carcinoma trial. Int J Cancer. 2013;132:2448-2458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
109.  Pawlik TM, Reyes DK, Cosgrove D, Kamel IR, Bhagat N, Geschwind JF. Phase II trial of sorafenib combined with concurrent transarterial chemoembolization with drug-eluting beads for hepatocellular carcinoma. J Clin Oncol. 2011;29:3960-3967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 243]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
110.  Meyer T, Fox R, Bird D, Watkinson A, Hacking N, Stocken D, Johnson PJ, Palmer DH. TACE 2: A randomized placebo-controlled, double-blinded, phase III trial evaluating sorafenib in combination with transarterial chemoembolization (TACE) in patients with unresectable hepatocellular carcinoma (HCC). J Clin Oncol. 2012;30 suppl:TPS4150.  [PubMed]  [DOI]  [Cited in This Article: ]