Review Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Feb 14, 2012; 18(6): 498-506
Published online Feb 14, 2012. doi: 10.3748/wjg.v18.i6.498
Targeted systemic therapies for hepatocellular carcinoma: Clinical perspectives, challenges and implications
Catherine Frenette, Robert Gish, California Pacific Medical Center, San Francisco, CA 94115, United States
Author contributions: Frenette C and Gish R contributed equally to the writing of this manuscript.
Correspondence to: Catherine Frenette, MD, Medical Director of Liver Transplantation, Director of Liver Cancer Program, The Methodist Hospital, Houston, TX 77030, United States. ctfrenette@tmhs.org
Telephone: +1-713-4410179 Fax: +1-731-4415248
Received: February 21, 2011
Revised: June 6, 2011
Accepted: June 13, 2011
Published online: February 14, 2012

Abstract

Hepatocellular carcinoma (HCC) is a lethal disease in most patients, due to its aggressive course and a lack of effective systemic therapies for advanced disease. Surgical resection and liver transplantation remain the only curative options for a small subset of patients. Few patients with HCC are diagnosed early enough to be eligible for curative treatment. Angiogenesis inhibition is a natural therapeutic target for all solid tumors, but particularly for the highly vascularized HCC tumors. With the approval of the targeted agent sorafenib, there are now additional options for patients with HCC. Although sorafenib does produce some improvement in survival in HCC patients, the responses are not durable. In addition, there are significant dermatologic, gastrointestinal, and metabolic toxicities, and, as importantly, there is still limited knowledge of its usefulness in special subpopulations with HCC. Other angiogenesis inhibitors are in development to treat HCC both in the first-line setting and for use following sorafenib failure; the furthest in development is brivanib, a dual fibroblast growth factor pathway and vascular endothelial growth factor receptor inhibitor. Additional agents with antiangiogenic properties also in phase II and III development for the treatment of patients with HCC include bevacizumab, ramucirumab, ABT-869, everolimus and ARQ 197.

Key Words: Hepatocellular carcinoma, Angiogenesis, Vascular endothelial growth factor, Fibroblast growth factor, Sorafenib, Tumor response, Brivanib



INTRODUCTION

Primary liver cancer, including hepatoma and hepatocellular carcinoma (HCC) is diagnosed in more than 560 000 people worldwide each year[1], including more than 24 000 Americans[2]. HCC accounts for up to 90% of all primary liver cancers[3]. HCC can be treated curatively with surgical resection or liver transplantation if diagnosed at an early stage; however, since most HCC patients present with advanced disease, only 15% are eligible for curative treatments[4]. Even for patients undergoing surgical resection, recurrence rates may be as high as 50% after 2 years and 76% by 10 years[5,6]. Patients meeting the Milan criteria who undergo liver transplantation can achieve a 5-year cancer-free survival rate greater than 60%[7].

As most patients with HCC are diagnosed with advanced disease, they generally have a poor prognosis, with median survival times of less than 1 year[3]. This is due, at least in part, to the absence of effective systemic therapies. Systemic therapies examined in the past, including both cytotoxic and hormonal agents, have provided limited or no benefit for these patients[6]. In late 2007, the angiogenesis inhibitor sorafenib was approved for use in advanced HCC based on an improvement in survival compared with placebo[8,9]. While initial responses are observed, over time a loss of efficacy is apparent that may be due to “resistance”via escape/compensatory mechanisms. Like other angiogenesis inhibitors, sorafenib also has known class side effects, including skin-related toxicities, hypertension, proteinuria, diarrhea, and an increased risk for thromboembolism and bleeding events[10-12]. While most are manageable, certain rare events can be life-threatening (i.e., gastrointestinal perforation, fatal hemoptysis, thromboembolic events). Thus, the balance between risk and benefit in every clinical setting is an integral part of the differentiation and evaluation of targeted agents.

RATIONALE FOR ANGIOGENESIS INHIBITION IN HCC

Angiogenesis is a ubiquitous process that is required for tumor growth[13,14]. Angiogenic processes are also indirectly involved in tumor invasion and metastasis through the secretion of matrix-degrading proteinases by vascular endothelial cells[15] and the ability of tumor cells to travel to distant sites via the vascular network[16].

Proangiogenic factors are attractive therapeutic targets because they stimulate cancer formation, growth, and proliferation via angiogenesis using a number of distinct mechanisms. Established proangiogenic factors and their receptor signaling pathways include vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2, platelet-derived growth factor (PDGF), angiogenin, and angiopoietin-2 (Ang-2)[17]. Other mediators, such as c-MET and mTOR, although not directly related to new vessel formation, affect angiogenesis via influences on downstream signaling.

HCC tumors are generally hypervascularized[18], suggesting that they may be especially vulnerable to angiogenesis inhibition. Several endogenous proangiogenic factors are expressed in HCC[19-22], and evidence indicates they play a role in HCC pathogenesis. For instance, serum VEGF levels increase with advancing HCC stages, being highest in patients with metastatic disease[23]. Elevated VEGF levels after locoregional therapy also are associated with poor prognosis and diminished response to therapy[24,25]. Expression of the proangiogenic factor FGF-2, the target of newer agents, is also elevated in patients with HCC[22] and its expression in HCC correlates with tumor microvessel density[26] and postoperative recurrence rate[27]. Tumor angiogenin expression correlates with microvascular density in patients with HCC, and high serum angiogenin levels are associated with decreased survival at 5 years[28]. Finally, mRNA angiopoietin expression level (via Ang-2/Ang-1 ratio) is positively correlated with tumor portal vein invasion, diameter, microvascular density, and poor prognosis[29]. Taken together, this evidence provides a strong rationale for targeting angiogenesis and related proangiogenic factors to provide more effective therapies for the treatment of HCC.

CHALLENGES AND LIMITATIONS OF SORAFENIB IN TREATING HCC

Sorafenib was the first systemic targeted therapy to be approved by the US Food and Drug Administration for patients with unresectable HCC[30], based on a 2.8-mo survival advantage over best supportive care (BSC) [hazard ratio (HR) 0.69, P = 0.00058] in the Sorafenib Hepatocellular carcinoma Assessment Randomized Protocol (SHARP) trial[31]. Sorafenib, which is also approved for advanced renal cell carcinoma (RCC)[30] inhibits the following receptor tyrosine kinases: VEGFR-2, VEGFR-3, PDGFR-β, c-KIT, and Flt-3[32]. It also binds to the serine-threonine kinases Raf, MEK, and ERK[32,33]. The VEGFR and PDGFR pathways and Raf-1 have all been implicated in the pathogenesis of HCC[34,35], providing a rationale for sorafenib activity in HCC.

Although sorafenib represents a much needed treatment option for patients with HCC, it also produces toxicities that may significantly affect patients’ quality of life. High rates of dermatologic side effects are commonly reported with sorafenib, the most clinically significant being hand-foot skin reaction (HFSR)[36]. HFSR typically develops in the first few weeks of therapy, with painful hyperkeratotic lesions on the palms and soles that are surrounded by a ring of erythema localized on areas of pressure or flexure[37]. A meta-analysis examining the incidence of HFSR in phase II and III trials across solid tumors reported an incidence of 33.8% with sorafenib use, 8.9% of which were grade 3[38]. In the SHARP trial, sorafenib-associated HFSR had an overall incidence of 21% and a grade 3 incidence of 8%[9]. Of particular concern, HSFR appears to be more prevalent in Asian patients, the population most affected worldwide by HCC. A phase III trial conducted solely in Asian patients with HCC reported a doubling of the overall incidence of HFSR compared with the SHARP trial[8,9]. Similarly, a phase III trial of sorafenib monotherapy following transarterial chemoembolization in Asian patients reported an HFSR incidence of 82%[39].

Bleeding events, albeit rare, are also a toxicity of sorafenib and other angiogenesis inhibitors. In a meta-analysis of 2109 patients enrolled in sorafenib clinical trials or expanded-access programs, the relative risk of bleeding events was 1.86 (P < 0.001) and the incidence of grade 3 or higher bleeding events was 2.2%[40]. It is notable that patients with HCC have not demonstrated an increased incidence of bleeding events with sorafenib; for example, in the SHARP trial, grade 3/4 bleeding events were 1% in the sorafenib arm.

By design, SHARP was conducted in the Americas, Europe, and Australasia, and thus generated limited data in Asian patients. Patients included in SHARP had preserved liver function and were of mostly Child-Pugh (C-P) A status[9]. In order to gain additional data, a randomized phase III trial in Asian patients with advanced HCC was completed, but again C-P B and C patients were excluded[8,39]. The overall trend of response to sorafenib from this Asian population was similar to those of SHARP, with time to progression (TTP) and overall survival (OS) improved compared with placebo, and similar treatment effects (HRs of 0.68-0.69). However, median OS appeared to be shorter (6.2 mo) vs SHARP (10.7 mo), most likely due to more advanced disease based on performance status, number of tumor sites, and presence of lung metastases compared with SHARP[8,9]. A subsequent single-arm, phase II trial was conducted in Asian patients with worse prognosis, including C-P B and C status (29%) and portal vein thrombosis (43%)[41]. In this trial, 26% of patients derived clinical benefit from sorafenib. Again, the incidence of toxicities was higher than in SHARP, with grade 3/4 diarrhea occurring in 20% and grade 3/4 HFSR in 16% of patients. As the majority of HCC cases are found in Asia[42], the development of safe and more effective therapies for this population represents a significant unmet need.

Hence, while the sorafenib trials have provided valuable information in patients with preserved liver function[8,9,41], determining efficacy and safety in the substantial portion of patients with advanced HCC remains a challenge. Other recent phase II trials are beginning to provide preliminary evidence of sorafenib safety and efficacy in patients with more advanced disease. A trial of sorafenib monotherapy in 59 patients with unresectable HCC, including 39% with C-P B status and 17% of C-P C status, showed promising activity regardless of disease stage and liver function[43]. Responses for patients of C-P B status were similar to those from SHARP. Median OS, however, declined with more advanced C-P status, most likely due to underlying cirrhosis, and because drug toxicities are more prevalent with compromised liver function, causing liver-related or systemic complications that lead to early treatment discontinuations. In a second phase II trial, pharmacokinetic profiles of sorafenib were similar in both C-P A and B subgroups, while median TTP and OS appeared shorter and adverse events related to poorer liver function were more frequent in C-P B patients[44]. Finally, another single-arm, phase II trial in 51 Asian patients, including 15 (29%) with C-P B/C status, found no significant differences between C-P B/C status patients and C-P A patients in disease control rate, median OS, grade 3/4 hematologic toxicities, or grade 3/4 nonhematologic toxicities[41].

Despite initial responses to sorafenib, and similar to other targeted agents, most HCC patients experience a loss of efficacy. Furthermore, across clinical trials, 20%-38% of patients discontinued sorafenib due to adverse events[8,9,41]. Similar to what has been reported with bevacizumab, some data indicate that patients who discontinue sorafenib therapy may experience “rebound,” whereby symptom and tumor progression develops rapidly upon discontinuation[45]. While this accelerated growth effect appears to be temporarily curtailed with re-initiation of therapy, insensitivity to treatment returns quickly. No effective second-line treatment options currently exist outside of clinical trials for patients who are resistant or refractory to and/or intolerant of sorafenib.

THE FAILED PROMISE OF SUNITINIB

Next to sorafenib, sunitinib is the most studied multitargeted tyrosine kinase inhibitor. Like sorafenib, sunitinib is an inhibitor of VEGFR and PDGFR and is currently indicated for the treatment of RCC, as well as for gastrointestinal stromal tumors[46]. While early indications were that sunitinib would have efficacy in HCC, the phase III SUN 1170 trial comparing sunitinib with sorafenib in patients with advanced HCC was discontinued due to increased serious sunitinib-related adverse events and the improbability of achieving noninferior efficacy[47]. As a result, sunitinib is no longer in development for the treatment of HCC.

CHALLENGES IN ASSESSING TUMOR RESPONSE

The 2 traditional imaging criteria widely used for measuring tumor responses to treatment are Response Evaluation Criteria In Solid Tumors (RECIST)[48], which are used primarily in the United States, and the World Health Organization (WHO) criteria[49], which are used internationally. With the advent of targeted therapies that are cytostatic, and in particular regarding HCC tumors, these traditional criteria are limited in their usefulness to assess treatment response because (1) cirrhotic livers may not remodel around a necrotic tumor; (2) HCC tumors are occasionally diffuse and infiltrative in cirrhotic livers; (3) arterial phase enhancement of premalignant dysplastic nodules can be mistaken for progression; and (4) cytostatic targeted agents alter tumor vascularity without affecting tumor size[50]. Both criteria have been updated in the past decade to account for HCC tumor viability in order to appropriately evaluate the extent of tumor necrosis and/or viability to quantify treatment response. The European Association for the Study of the Liver updated the WHO criteria in 2000[51], and in 2008 the American Association for the Study of Liver Diseases updated RECIST[52] and then further clarified the modified criteria in 2010[53]. Specific differences between these current criteria-modified WHO (mWHO) and modified RECIST (mRECIST)-for treatment response in HCC are listed in Table 1.

Table 1 Response assessment by modified World Health Organization criteria and modified Response Evaluation Criteria in Solid Tumors.
ParameterModified WHOModified RECIST
Type of assessmentSpiral CTSpiral CT or dynamic MRI
Frequency of assessment≥ 4 wk6-8 wk
Measurement of tumor volumeBidimensional measurementUnidimensional measurement
Tumor necrosis measurementReduction in viable tumor area using contrast-enhanced radiological imagingReduction in viable tumor area using contrast-enhanced radiological imaging
Viable tumor definitionEnhanced areas inside treatment lesionsUptake of contrast agent in the arterial phase
Complete responseComplete disappearance of tumor enhancement determined by 2 observations ≥ 4 wk apartDisappearance of any intratumoral arterial enhancement in all target lesions
Partial response> 50% reduction in total area of tumor enhancement determined by 2 observations ≥ 4 wk apart≥ 30% decrease in the sum of diameters of viable target lesions, taking as reference the baseline sum of the diameters of target lesions
Stable diseaseInsufficient shrinkage to qualify for partial response and insufficient increase to qualify for progressive diseaseAny cases that do not qualify for either partial response or progressive disease
Progressive disease> 25% increase in total area of tumor enhancement or the appearance of new lesions≥ 20% increase in the sum of the diameters of viable target lesions, taking as reference the smallest sum of the diameters of viable target lesions recorded since the treatment started or the appearance of 1 or more new lesions

As an example of the variability in response assessment that can arise according to which of the 2 criteria is used, Finn et al[54] retrospectively compared response assessment by the necrosis-adjusted mRECIST (mRECIST has not yet been validated) for HCC with the prospective use of mWHO criteria in 101 patients with HCC who received brivanib, an antiangiogenic targeted agent. They found that mWHO criteria underreported treatment benefit compared with mRECIST for HCC, with 31 patients classified as having progressive disease under the mWHO criteria but stable disease or partial response under mRECIST. This suggests that these patients may have been prematurely discontinued from treatment while still deriving benefit. These discrepancies can be explained by differences in the methodologies of mRECIST and mWHO, which employ different measurements and calculations of tumor size, thereby producing distinct interpretations of treatment response. In addition, there are differences in how progression is defined, with cytologic confirmation of ascites required with mRECIST and small new lesions (< 1 cm) unlikely representing progression with the mRECIST. While not conclusive, these results suggest that treatment response based on assessment criteria vary widely and indicate the need for further clinical validation.

LOSS OF EFFICACY TO ANTIANGIOGENIC THERAPY: ESCAPE AND RESISTANCE

Clinical trials of antiangiogenic agents have shown that most patients with advanced tumors eventually experience progression, including those who initially respond to treatment[8,9,55-57]. Recent evidence suggests that relapse during treatment with antiangiogenic agents occurs due to VEGF inhibition-driven hypoxia, which induces upregulation of alternate proangiogenic signals such as FGF, which overrides the VEGFR inhibition[58]. This was demonstrated preclinically in murine tumors that initially responded to treatment with an anti-VEGFR-2 antibody, but relapsed after 2 wk, showing higher levels of other proangiogenic signals, including FGF-1 and FGF-2, than untreated tumors[59]. Moreover, blockage of FGF signaling in this model slowed tumor growth and attenuated its revascularization during the relapse phase. Clinically, this has been observed in patients with HCC[26,27,60] and, more recently, in glioblastoma patients treated with the pan-VEGFR inhibitor AZD2171, in whom increased plasma levels of FGF were detected upon relapse[61]. Another study showed that approximately half of patients with metastatic colorectal cancer who received bevacizumab plus chemotherapy had more than a 5-fold increase in either placental growth factor or FGF prior to progression[62]. Patients with late-stage breast cancer have been reported to express a large number of proangiogenic factors, including FGF-2, in contrast to earlier stage lesions, which primarily express VEGF[58,63]. Taken together, these data support the hypothesis that tumor progression during inhibition of angiogenesis may be facilitated via activation of compensatory proangiogenic and tumorigenic mechanisms.

COMPOUNDS IN DEVELOPMENT FOR TREATMENT OF HCC

Several compounds in development stand to address the challenges and limitations of targeted therapy in the treatment of HCC. These are discussed in the following sections and summarized in Table 2.

Table 2 Agents with antiangiogenic properties in development for the treatment of hepatocellular carcinoma.
AgentTherapeutic targetPhase of study
BrivanibVEGFR, FGFRIII
BevacizumabVEGFII
RamucirumabVEGFR-2III
ABT-869VEGFR-1, VEGFR-2, PDGFR-β, c-KIT, Flt-3III
EverolimusmTORIII
ARQ 197c-METII
Brivanib

Brivanib is currently in phase III trials in HCC. It is distinct from both sorafenib and sunitinib in that it is an oral, selective, dual inhibitor of the FGF and VEGF signalling pathways[64,65]. Since FGF signaling may contribute to acquired “resistance,” or compensatory signaling, during anti-VEGFR therapy[58], the simultaneous inhibition of these[64,65] 2 pathways by brivanib may both delay initial progression in response to antiangiogenic therapy (as first-line treatment) and successfully treat tumors that have already progressed during anti-VEGFR therapy (as second-line treatment). With respect to its potential as first-line therapy, brivanib has delayed initial progression compared with sorafenib in preclinical studies[66]. It has also shown specific inhibitory activity in patient-derived HCC xenografts implanted in mice[67]. Clinically, brivanib has demonstrated a disease control rate of 51%, a median TTP of 2.8 mo, and an OS of 10 mo as first-line monotherapy in a phase II trial of predominantly Asian patients with HCC[68]. A retrospective analysis using mRECIST for HCC criteria also demonstrated an objective response rate of 25%, with 9% complete responses[68]. Brivanib was also associated with a low incidence of grade 3/4 adverse events, including hypertension (10.9%), diarrhea (3.6%), and HFSR (1.8%)[68]. Due to its unique mechanism of action and favorable safety profile, brivanib is currently under phase III investigation as first-line therapy vs sorafenib in patients with advanced HCC (BRISK-FL). As a potential second-line agent following antiangiogenic therapy, brivanib has demonstrated activity against xenograft tumors that were nonresponsive to bevacizumab[69]. Allen et al[66] also used a mouse model of pancreatic neuroendocrine cancer to show that brivanib administered after sorafenib failure could delay tumor growth modestly, despite showing evidence of revascularization. In a phase II trial of brivanib in patients with HCC who had been treated with sorafenib, brivanib produced a median TTP of 2.7 mo and an OS of 9.8 mo[70]; in a retrospective analysis of paired TTP, at least 40% of patients had longer TTP with brivanib than with prior sorafenib[71]. Brivanib is currently under investigation in 2 second-line phase III trials-1 in Asian patients following sorafenib failure (progression or intolerance; BRISK-APS), and another similar trial that is enrolling an ethnically unselected patient population (BRISK-PS).

Bevacizumab

The anti-VEGF monoclonal antibody bevacizumab was the first angiogenesis inhibitor to be approved as an antineoplastic agent[72]. Bevacizumab has shown activity in phase II HCC testing in combination with chemotherapy[72-75], with the epidermal growth factor receptor inhibitor erlotinib[76], and as monotherapy[77]. Despite initial safety concerns, particularly gastrointestinal bleeding and thrombosis, phase II trials in HCC have shown toxicities to be manageable[78]. New bevacizumab combinations are under investigation in ongoing phase II HCC trials, including combination with sorafenib, everolimus, temsirolimus, chemoembolization, and hepatic arterial infusion of floxuridine and dexamethasone.

Ramucirumab

The monoclonal antibody ramucirumab is a specific inhibitor of VEGFR-2[79]. A phase II study of 42 patients with advanced HCC and primarily well-preserved liver function (75% C-P A status) showed that first-line ramucirumab monotherapy produced a disease control rate of 50% and a median progression-free survival (PFS) of 4.3 mo[80]. This positive study prompted the initiation of the phase III REACH trial in HCC, which is comparing ramucirumab/supportive care with placebo/supportive care for second-line treatment after sorafenib.

ABT-869

ABT-869 is a multitargeted tyrosine kinase inhibitor that inhibits multiple members of the VEGFR and PDGFR families[81]. In a xenograft model of HCC, ABT-869 significantly reduced tumor burden, either alone or in combination with rapamycin[82]. Interim phase II results in patients with advanced HCC showed a median TTP of 3.7 mo with ABT-869 treatment and a safety profile consistent with angiogenesis inhibition[83]. ABT-869 is in phase III testing as a first-line treatment for advanced HCC versus sorafenib.

INHIBITION OF OTHER ANGIOGENIC AND TUMORIGENIC PATHWAYS
mTOR inhibitors: Everolimus and sirolimus

mTOR inhibitors are not traditionally considered direct angiogenesis inhibitors; rather, they have well-known immunosuppressive properties. In fact, 2 of these agents, sirolimus and everolimus, are used to prevent rejection in organ transplant recipients[84]. mTOR inhibitors also have antineoplastic properties, via mTOR regulation of tumor proliferation and metabolism[85]. MTOR indirectly modulates angiogenesis through regulation of VEGF expression and translation of proteins involved in angiogenesis[86]. Clinically, there is growing evidence to suggest that mTOR inhibitors may reduce de novo malignant growth[87] and recurrence in the liver post-transplant[88]. In patients with advanced HCC, everolimus produced a median PFS of 3.8 mo and a disease control rate of 44% in phase I/II testing[81]. Consequently, the ongoing phase III EVOLVE-1 trial has been initiated to compare everolimus with BSC in patients with HCC who progressed on or after sorafenib or who were intolerant to sorafenib.

ARQ 197

Similar to everolimus, ARQ 197 has antiangiogenic properties, but is not considered an angiogenesis inhibitor. ARQ 197 is an inhibitor of the oncogene c-MET, which stimulates tumor growth, invasion, metastasis, and angiogenesis via binding of its ligand, hepatocyte growth factor[89]. In phase I testing in cirrhotic patients with HCC, ARQ 197 demonstrated some activity and was well tolerated, with serious adverse events that were primarily hematologic[90]. ARQ 197 is currently in phase II testing in second-line advanced HCC.

CONCLUSION

Unmet needs for HCC remain, despite the availability of sorafenib. Indeed, sorafenib has some significant limitations, including modest, transient benefits, and toxicity challenges; and its use in patients with more advanced liver disease and in Asian patients has not yet been fully defined. The development of newer targeted therapies that inhibit angiogenesis simultaneously with inhibition of other key proangiogenic factors in HCC, such as FGFR or c-MET signaling, is providing further insights into the underlying pathogenesis of HCC tumors. Compounds that directly block angiogenesis and tumorigenesis via dual inhibition of FGFR and VEGFR, such as brivanib, and other compounds that indirectly modulate angiogenesis, such as mTOR inhibitors, are providing novel mechanisms that exploit critical pathways in HCC tumor progression and may have the potential to improve clinical outcomes both as monotherapy and in the case of escape from sorafenib. In the coming years, a number of phase III clinical trials examining these angiogenesis inhibitors will be mature, providing a better picture of the clinical utility and treatment options for patients with HCC.

ACKNOWLEDGMENTS

We thank Lena Prisco, PhD, and Mark English, PhD, of PAREXEL and Bristol-Myers Squibb for their editorial assistance with this paper.

Footnotes

Peer reviewer: Jean Paul Galmiche, MD, Professor, Department of Gastroenterology and Hepatology, Hôpital Hôtel Dieu, 44093 Nantes cedex, France

S- Editor Gou SX L- Editor O’Neill M E- Editor Xiong L

References
1.  Stewart BW, Kleihues P.  World cancer report. Lyon: IARC Press 2003; .  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10002]  [Cited by in F6Publishing: 10353]  [Article Influence: 739.5]  [Reference Citation Analysis (0)]
3.  Nordenstedt H, White DL, El-Serag HB. The changing pattern of epidemiology in hepatocellular carcinoma. Dig Liver Dis. 2010;42:S206-S214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 367]  [Cited by in F6Publishing: 394]  [Article Influence: 28.1]  [Reference Citation Analysis (0)]
4.  Finn RS. Development of molecularly targeted therapies in hepatocellular carcinoma: where do we go now? Clin Cancer Res. 2010;16:390-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
5.  Poon RT, Fan ST, Lo CM, Liu CL, Wong J. Long-term survival and pattern of recurrence after resection of small hepatocellular carcinoma in patients with preserved liver function: implications for a strategy of salvage transplantation. Ann Surg. 2002;235:373-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 647]  [Cited by in F6Publishing: 664]  [Article Influence: 30.2]  [Reference Citation Analysis (0)]
6.  Zhu AX. Systemic therapy of advanced hepatocellular carcinoma: how hopeful should we be? Oncologist. 2006;11:790-800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 209]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
7.  Pelletier SJ, Fu S, Thyagarajan V, Romero-Marrero C, Batheja MJ, Punch JD, Magee JC, Lok AS, Fontana RJ, Marrero JA. An intention-to-treat analysis of liver transplantation for hepatocellular carcinoma using organ procurement transplant network data. Liver Transpl. 2009;15:859-868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 119]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
8.  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: 4350]  [Article Influence: 271.9]  [Reference Citation Analysis (0)]
9.  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: 9552]  [Article Influence: 597.0]  [Reference Citation Analysis (1)]
10.  Nexavar [prescribing information]. Wayne, NJ: Bayer HealthCare Pharmaceuticals Inc. 2009; .  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Launay-Vacher V, Deray G. Hypertension and proteinuria: a class-effect of antiangiogenic therapies. Anticancer Drugs. 2009;20:81-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 79]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
12.  Roodhart JM, Langenberg MH, Witteveen E, Voest EE. The molecular basis of class side effects due to treatment with inhibitors of the VEGF/VEGFR pathway. Curr Clin Pharmacol. 2008;3:132-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 158]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
13.  Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6437]  [Cited by in F6Publishing: 6276]  [Article Influence: 261.5]  [Reference Citation Analysis (0)]
14.  Ribatti D, Vacca A, Dammacco F. The role of the vascular phase in solid tumor growth: a historical review. Neoplasia. 1999;1:293-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 107]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
15.  Mignatti P, Rifkin DB. Biology and biochemistry of proteinases in tumor invasion. Physiol Rev. 1993;73:161-195.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Aznavoorian S, Murphy AN, Stetler-Stevenson WG, Liotta LA. Molecular aspects of tumor cell invasion and metastasis. Cancer. 1993;71:1368-1383.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Maruotti N, Cantatore FP, Crivellato E, Vacca A, Ribatti D. Angiogenesis in rheumatoid arthritis. Histol Histopathol. 2006;21:557-566.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Ribatti D, Vacca A, Nico B, Sansonno D, Dammacco F. Angiogenesis and anti-angiogenesis in hepatocellular carcinoma. Cancer Treat Rev. 2006;32:437-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 61]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
19.  Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor. Endocr Rev. 1997;18:4-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1070]  [Cited by in F6Publishing: 1015]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
20.  Mochida S, Ishikawa K, Toshima K, Inao M, Ikeda H, Matsui A, Shibuya M, Fujiwara K. The mechanisms of hepatic sinusoidal endothelial cell regeneration: a possible communication system associated with vascular endothelial growth factor in liver cells. J Gastroenterol Hepatol. 1998;13 Suppl:S1-S5.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Tanaka S, Mori M, Sakamoto Y, Makuuchi M, Sugimachi K, Wands JR. Biologic significance of angiopoietin-2 expression in human hepatocellular carcinoma. J Clin Invest. 1999;103:341-345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 194]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
22.  Uematsu S, Higashi T, Nouso K, Kariyama K, Nakamura S, Suzuki M, Nakatsukasa H, Kobayashi Y, Hanafusa T, Tsuji T. Altered expression of vascular endothelial growth factor, fibroblast growth factor-2 and endostatin in patients with hepatocellular carcinoma. J Gastroenterol Hepatol. 2005;20:583-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 57]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
23.  Jinno K, Tanimizu M, Hyodo I, Nishikawa Y, Hosokawa Y, Doi T, Endo H, Yamashita T, Okada Y. Circulating vascular endothelial growth factor (VEGF) is a possible tumor marker for metastasis in human hepatocellular carcinoma. J Gastroenterol. 1998;33:376-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 102]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
24.  Montella L, Addeo R, Caraglia M, Faiola V, Guarrasi R, Vincenzi B, Palmeri A, Capasso E, Nocera V, Tarantino L. Vascular endothelial growth factor monitoring in advanced hepatocellular carcinoma patients treated with radiofrequency ablation plus octreotide: a single center experience. Oncol Rep. 2008;20:385-390.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Tamesa T, Iizuka N, Mori N, Okada T, Takemoto N, Tangoku A, Oka M. High serum levels of vascular endothelial growth factor after hepatectomy are associated with poor prognosis in hepatocellular carcinoma. Hepatogastroenterology. 2009;56:1122-1126.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  El-Assal ON, Yamanoi A, Ono T, Kohno H, Nagasue N. The clinicopathological significance of heparanase and basic fibroblast growth factor expressions in hepatocellular carcinoma. Clin Cancer Res. 2001;7:1299-1305.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Poon RT, Ng IO, Lau C, Yu WC, Fan ST, Wong J. Correlation of serum basic fibroblast growth factor levels with clinicopathologic features and postoperative recurrence in hepatocellular carcinoma. Am J Surg. 2001;182:298-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 110]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
28.  Hisai H, Kato J, Kobune M, Murakami T, Miyanishi K, Takahashi M, Yoshizaki N, Takimoto R, Terui T, Niitsu Y. Increased expression of angiogenin in hepatocellular carcinoma in correlation with tumor vascularity. Clin Cancer Res. 2003;9:4852-4859.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Mitsuhashi N, Shimizu H, Ohtsuka M, Wakabayashi Y, Ito H, Kimura F, Yoshidome H, Kato A, Nukui Y, Miyazaki M. Angiopoietins and Tie-2 expression in angiogenesis and proliferation of human hepatocellular carcinoma. Hepatology. 2003;37:1105-1113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 146]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
30.  US Food and Drug Administration. FDA approval for sorafenib tosylate. 2010-10-01.  Available from: http: //www.cancer.gov/cancertopics/druginfo/fda-sorafenib-tosylate.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Kane RC, Farrell AT, Madabushi R, Booth B, Chattopadhyay S, Sridhara R, Justice R, Pazdur R. Sorafenib for the treatment of unresectable hepatocellular carcinoma. Oncologist. 2009;14:95-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 148]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
32.  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: 3039]  [Article Influence: 152.0]  [Reference Citation Analysis (0)]
33.  Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, Wilhelm S, Lynch M, Carter C. 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;66:11851-11858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1042]  [Cited by in F6Publishing: 1129]  [Article Influence: 66.4]  [Reference Citation Analysis (0)]
34.  Mukherjee B, Das T, Ghosh S, Datta S. Changes in the antioxidant defense and hepatic drug metabolizing enzyme and isoenzyme levels, 8-hydroxydeoxyguanosine formation and expressions of c-raf.1 and insulin-like growth factor II genes during the stages of development of hepatocellular carcinoma in rats. Eur J Cancer Prev. 2007;16:363-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
35.  Yang ZF, Poon RT. Vascular changes in hepatocellular carcinoma. Anat Rec (Hoboken). 2008;291:721-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 145]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
36.  Lacouture ME, Wu S, Robert C, Atkins MB, Kong HH, Guitart J, Garbe C, Hauschild A, Puzanov I, Alexandrescu DT. Evolving strategies for the management of hand-foot skin reaction associated with the multitargeted kinase inhibitors sorafenib and sunitinib. Oncologist. 2008;13:1001-1011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 259]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
37.  Robert C, Soria JC, Spatz A, Le Cesne A, Malka D, Pautier P, Wechsler J, Lhomme C, Escudier B, Boige V. Cutaneous side-effects of kinase inhibitors and blocking antibodies. Lancet Oncol. 2005;6:491-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 424]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
38.  Chu D, Lacouture ME, Fillos T, Wu S. Risk of hand-foot skin reaction with sorafenib: a systematic review and meta-analysis. Acta Oncol. 2008;47:176-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 131]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
39.  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: ]
40.  Je Y, Schutz FA, Choueiri TK. Risk of bleeding with vascular endothelial growth factor receptor tyrosine-kinase inhibitors sunitinib and sorafenib: a systematic review and meta-analysis of clinical trials. Lancet Oncol. 2009;10:967-974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 215]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
41.  Yau T, Chan P, Ng KK, Chok SH, Cheung TT, Fan ST, Poon RT. Phase 2 open-label study of single-agent sorafenib in treating advanced hepatocellular carcinoma in a hepatitis B-endemic Asian population: presence of lung metastasis predicts poor response. Cancer. 2009;115:428-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 126]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
42.  Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer. 2001;94:153-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2651]  [Cited by in F6Publishing: 2570]  [Article Influence: 111.7]  [Reference Citation Analysis (0)]
43.  Pinter M, Sieghart W, Graziadei I, Vogel W, Maieron A, Königsberg R, Weissmann A, Kornek G, Plank C, Peck-Radosavljevic M. Sorafenib in unresectable hepatocellular carcinoma from mild to advanced stage liver cirrhosis. Oncologist. 2009;14:70-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 128]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
44.  Abou-Alfa GK, Amadori D, Santoro A, Figer A, De Greve J, Lathia C, Voliotis D, Anderson S, Moscovici M, Ricci S. Is sorafenib (S) safe and effective in patients (pts) with hepatocellular carcinoma (HCC) and Child-Pugh B (CPB) cirrhosis? [abstract]. J Clin Oncol. 2008;26:4518.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Desar IM, Mulder SF, Stillebroer AB, van Spronsen DJ, van der Graaf WT, Mulders PF, van Herpen CM. The reverse side of the victory: flare up of symptoms after discontinuation of sunitinib or sorafenib in renal cell cancer patients. A report of three cases. Acta Oncol. 2009;48:927-931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
46.  US Food and Drug Administration. FDA approval for sunitinib malate. 7, 2010-10-07.  Available from: http: //www.cancer.gov/cancertopics/druginfo/fda-sunitinib-malate..  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Pfizer discontinues phase 3 trial of Sutent in advanced hepatocellular carcinoma [press release]. Business Wire Web site. 2010-10-13.  Available from: http: //www.businesswire.com/news/home/20100422006892/en/Pfizer-Discontinues-Phase-3-Trial-Sutent-Advanced..  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92:205-216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12751]  [Cited by in F6Publishing: 12904]  [Article Influence: 537.7]  [Reference Citation Analysis (0)]
49.  Miller AB, Hoogstraten B, Staquet M, Winkler A. Reporting results of cancer treatment. Cancer. 1981;47:207-214.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Thomas MB, Jaffe D, Choti MM, Belghiti J, Curley S, Fong Y, Gores G, Kerlan R, Merle P, O'Neil B. Hepatocellular carcinoma: consensus recommendations of the National Cancer Institute Clinical Trials Planning Meeting. J Clin Oncol. 2010;28:3994-4005.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 306]  [Article Influence: 21.9]  [Reference Citation Analysis (1)]
51.  Bruix J, Sherman M, Llovet JM, Beaugrand M, Lencioni R, Burroughs AK, Christensen E, Pagliaro L, Colombo M, Rodés J. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL conference. European Association for the Study of the Liver. J Hepatol. 2001;35:421-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3252]  [Cited by in F6Publishing: 3170]  [Article Influence: 137.8]  [Reference Citation Analysis (0)]
52.  Llovet JM, Di Bisceglie AM, Bruix J, Kramer BS, Lencioni R, Zhu AX, Sherman M, Schwartz M, Lotze M, Talwalkar J. Design and endpoints of clinical trials in hepatocellular carcinoma. J Natl Cancer Inst. 2008;100:698-711.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1232]  [Cited by in F6Publishing: 1298]  [Article Influence: 81.1]  [Reference Citation Analysis (0)]
53.  Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30:52-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2583]  [Cited by in F6Publishing: 2928]  [Article Influence: 209.1]  [Reference Citation Analysis (36)]
54.  Finn RS, Raoul J, Manekas D, Baudelet C, Walters IB. Optimal assessment of treatment benefit with targeted agents for hepatocellular carcinoma (HCC): an analysis of brivanib phase II data [abstract]. J Clin Oncol. 2010;28:4096.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335-2342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7832]  [Cited by in F6Publishing: 7527]  [Article Influence: 376.4]  [Reference Citation Analysis (1)]
56.  Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, Shenkier T, Cella D, Davidson NE. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007;357:2666-2676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2393]  [Cited by in F6Publishing: 2249]  [Article Influence: 132.3]  [Reference Citation Analysis (0)]
57.  Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, Lilenbaum R, Johnson DH. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006;355:2542-2550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4457]  [Cited by in F6Publishing: 4355]  [Article Influence: 241.9]  [Reference Citation Analysis (0)]
58.  Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8:592-603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2122]  [Cited by in F6Publishing: 2186]  [Article Influence: 136.6]  [Reference Citation Analysis (0)]
59.  Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8:299-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1215]  [Cited by in F6Publishing: 1231]  [Article Influence: 64.8]  [Reference Citation Analysis (0)]
60.  Poon RT, Lau CP, Cheung ST, Yu WC, Fan ST. Quantitative correlation of serum levels and tumor expression of vascular endothelial growth factor in patients with hepatocellular carcinoma. Cancer Res. 2003;63:3121-3126.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Batchelor TT, Sorensen AG, di Tomaso E, Zhang WT, Duda DG, Cohen KS, Kozak KR, Cahill DP, Chen PJ, Zhu M. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1442]  [Cited by in F6Publishing: 1325]  [Article Influence: 77.9]  [Reference Citation Analysis (0)]
62.  Kopetz S, Hoff PM, Eng C, Overman MJ, Glover Y, Chang DZ, Wolff RA, Abbruzzee L, Ellis LM, Heymach JV. Levels of angiogenic cytokines prior to disease progression in metastatic colorectal cancer patients treated with bevacizumab. ASCO-GI. 2009;.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Relf M, LeJeune S, Scott PA, Fox S, Smith K, Leek R, Moghaddam A, Whitehouse R, Bicknell R, Harris AL. Expression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor beta-1, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesis. Cancer Res. 1997;57:963-969.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Bhide RS, Cai ZW, Zhang YZ, Qian L, Wei D, Barbosa S, Lombardo LJ, Borzilleri RM, Zheng X, Wu LI. Discovery and preclinical studies of (R)-1-(4-(4-fluoro-2-methyl-1H-indol-5-yloxy)-5- methylpyrrolo[2,1-f][1,2,4]triazin-6-yloxy)propan- 2-ol (BMS-540215), an in vivo active potent VEGFR-2 inhibitor. J Med Chem. 2006;49:2143-2146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
65.  Tille JC, Wood J, Mandriota SJ, Schnell C, Ferrari S, Mestan J, Zhu Z, Witte L, Pepper MS. Vascular endothelial growth factor (VEGF) receptor-2 antagonists inhibit VEGF- and basic fibroblast growth factor-induced angiogenesis in vivo and in vitro. J Pharmacol Exp Ther. 2001;299:1073-1085.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Allen E, Walters IB, Rivera IC, Hanahan D. Abstract B16: Anti-angiogenic therapy using brivanib, a combined VEGF and FGF pathway inhibitor, in a mouse model of pancreatic neuroendocrine cancer (PNET), results in sustained vascular blockade, without evidence for evasive/acquired resistance in the form of VEGF-independent revascularization, in contrast to other VEGF inhibitors. Mol Cancer Ther. 2009;8:B16.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Huynh H, Ngo VC, Fargnoli J, Ayers M, Soo KC, Koong HN, Thng CH, Ong HS, Chung A, Chow P. Brivanib alaninate, a dual inhibitor of vascular endothelial growth factor receptor and fibroblast growth factor receptor tyrosine kinases, induces growth inhibition in mouse models of human hepatocellular carcinoma. Clin Cancer Res. 2008;14:6146-6153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 175]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
68.  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: ]
69.  Ayers M, Dito G, Henley B, Jeyaseelan R, Yoganthan S, Han X, Wu Q, Platero S, Wu S, Feltquate D. Comparison of a dual inhibitor of VEGF and FGF signaling, BMS-582664, to the activity of bevacizumab, an inhibitor exclusively of VEGF signaling, in xenograft models of colon carcinoma. Proc Am Assoc Cancer Res. 2007;48:Abstract 1618.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  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;[Epub ahead of print].  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Finn RS, Park JW, Kang YK, Baudelet C, Walters I.  Time-to-progression sub-analysis of second-line treatment with brivanib after failure of prior antiangiogenic therapy in patients with unresectable, locally advanced, or metastatic hepatocellular carcinoma (Abstract). Boston, MA: The 60th Annual Meeting of the American Association for the Study of Liver Diseases 2009; .  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Avastin [prescribing information]. South San Francisco, CA: Genentech, Inc. 2009; .  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Sun W, Haller DG, Mykulowycz K, Rosen M, Soulen M, Capparo M, Faust T, Giantonia B, Olthoff K. Combination of capecitabine, oxaliplatin with bevacizumab in treatment of advanced hepatocellular carcinoma (HCC): A phase II study [abstract]. J Clin Oncol. 2007;25:4574.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Hsu CH, Yang TS, Hsu C, Toh HC, Epstein RJ, Hsiao LT, Chen PJ, Lin ZZ, Chao TY, Cheng AL. Efficacy and tolerability of bevacizumab plus capecitabine as first-line therapy in patients with advanced hepatocellular carcinoma. Br J Cancer. 2010;102:981-986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 101]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
75.  Zhu AX, Blaszkowsky LS, Ryan DP, Clark JW, Muzikansky A, Horgan K, Sheehan S, Hale KE, Enzinger PC, Bhargava P. Phase II study of gemcitabine and oxaliplatin in combination with bevacizumab in patients with advanced hepatocellular carcinoma. J Clin Oncol. 2006;24:1898-1903.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 285]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
76.  Thomas MB, Morris JS, Chadha R, Iwasaki M, Kaur H, Lin E, Kaseb A, Glover K, Davila M, Abbruzzese J. Phase II trial of the combination of bevacizumab and erlotinib in patients who have advanced hepatocellular carcinoma. J Clin Oncol. 2009;27:843-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 236]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
77.  Siegel AB, Cohen EI, Ocean A, Lehrer D, Goldenberg A, Knox JJ, Chen H, Clark-Garvey S, Weinberg A, Mandeli J. Phase II trial evaluating the clinical and biologic effects of bevacizumab in unresectable hepatocellular carcinoma. J Clin Oncol. 2008;26:2992-2998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 378]  [Cited by in F6Publishing: 376]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
78.  Finn RS, Zhu AX. Targeting angiogenesis in hepatocellular carcinoma: focus on VEGF and bevacizumab. Expert Rev Anticancer Ther. 2009;9:503-509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 84]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
79.  Spratlin JL, Cohen RB, Eadens M, Gore L, Camidge DR, Diab S, Leong S, O'Bryant C, Chow LQ, Serkova NJ. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol. 2010;28:780-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 424]  [Cited by in F6Publishing: 421]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
80.  Zhu AX, Finn RS, Mulcahy MF, Gurtler JS, Sun W, Schwartz JD, Rojas P, Dontabhaktuni A, Youssoufian H, Stuart KE. A phase II study of ramucirumab as first-line monotherapy in patients (pts) with advanced hepatocellular carcinoma (HCC) [abstract]. J Clin Oncol. 2010;28:4083.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Albert DH, Tapang P, Magoc TJ, Pease LJ, Reuter DR, Wei RQ, Li J, Guo J, Bousquet PF, Ghoreishi-Haack NS. Preclinical activity of ABT-869, a multitargeted receptor tyrosine kinase inhibitor. Mol Cancer Ther. 2006;5:995-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 157]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
82.  Jasinghe VJ, Xie Z, Zhou J, Khng J, Poon LF, Senthilnathan P, Glaser KB, Albert DH, Davidsen SK, Chen CS. ABT-869, a multi-targeted tyrosine kinase inhibitor, in combination with rapamycin is effective for subcutaneous hepatocellular carcinoma xenograft. J Hepatol. 2008;49:985-997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
83.  Toh H, Chen P, Carr BI, Knox JJ, Gill S, Steinberg J, Carlson DM, Qian J, Qin Q, Yong W. A phase II study of ABT-869 in hepatocellular carcinoma (HCC): Interim analysis [abstract]. J Clin Oncol. 2009;27:A4581.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Geissler EK, Schlitt HJ, Thomas G. mTOR, cancer and transplantation. Am J Transplant. 2008;8:2212-2218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 94]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
85.  Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124:471-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4260]  [Cited by in F6Publishing: 4276]  [Article Influence: 237.6]  [Reference Citation Analysis (0)]
86.  Huynh H. Molecularly targeted therapy in hepatocellular carcinoma. Biochem Pharmacol. 2010;80:550-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 101]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
87.  Campistol JM, Eris J, Oberbauer R, Friend P, Hutchison B, Morales JM, Claesson K, Stallone G, Russ G, Rostaing L. Sirolimus therapy after early cyclosporine withdrawal reduces the risk for cancer in adult renal transplantation. J Am Soc Nephrol. 2006;17:581-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 379]  [Cited by in F6Publishing: 399]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
88.  Chinnakotla S, Davis GL, Vasani S, Kim P, Tomiyama K, Sanchez E, Onaca N, Goldstein R, Levy M, Klintmalm GB. Impact of sirolimus on the recurrence of hepatocellular carcinoma after liver transplantation. Liver Transpl. 2009;15:1834-1842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 124]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
89.  You WK, McDonald DM. The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Rep. 2008;41:833-839.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Zucali P, Santoro A, Rodriguez-Lope C, Simonelli M, Camacho LH, Senzer NN, Bolondi L, Lamar M, Abbadessa G, Schwartz BE. Final results from ARQ 197-114: A phase Ib safety trial evaluating ARQ 197 in cirrhotic patients (pts) with hepatocellular carcinoma (HCC). J Clin Oncol. 2010;28 suppl:Abstract 4137.  [PubMed]  [DOI]  [Cited in This Article: ]