Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jul 18.
Published in final edited form as: J Hepatol. 2008 Apr 28;49(1):1–5. doi: 10.1016/j.jhep.2008.04.006

Molecular targeted therapies in hepatocellular carcinoma: From pre-clinical models to clinical trials

Pippa Newell 1, Augusto Villanueva 2, Josep M Llovet 1,2,*
PMCID: PMC12272936  NIHMSID: NIHMS2089823  PMID: 18486262

1. Introduction

Hepatocellular carcinoma (HCC) is one of the world’s most common and deadly cancers. Less than one-third of patients can currently benefit from potentially curative therapies in the West [1]. However, a new era has dawned in oncology with novel and promising drugs emerging in parallel with a better understanding of the pathogenesis of cancer [2].

The advent of sorafenib – a multikinase inhibitor – as an effective therapy in advanced HCC has enhanced the interest in testing new molecular therapies in experimental and clinical studies [3]. Integrative genomic studies in human HCC samples have begun to identify subgroups of patients with characteristic molecular features such as mutations, gene expression profiles and chromosomal aberrations [4,5]. These studies have underlined the fact that a number of molecular pathways are disrupted in almost all tumors, involving critical functions for the progression or dissemination of the disease. Such is the case of three main cellular functions: (1) Activation of pro-angiogenic signals mediated by VEGF, PDGER, angiopoeitin-2, and others [4,6]; (2) Mitosis checkpoint disruption and activation of pro-apoptotic mediated by mutations of critical tumor suppressors (e.g. p53, inactivation of Rb) or activation of oncogenes (e.g. Cyclin D1) [7]; (3) Acquisition of limitless replicative potential through the activation of TERT at preneoplastic and early HCC stages [8]. Nonetheless, the main driving force ensuring tumor viability in HCC depends on the activation of specific signal transduction pathways leading to tumor proliferation. Nonetheless, the main driving force ensuring tumor viability in HCC depends on the activation of specific signaling pathways leading to tumor proliferation. From the molecular classifications published so far, one-third of HCCs are driven by proliferative signals generated from Tyrosine Kinase Receptor (e.g. EGFR, IGF-IR), RAS/MAPK, PI3K-Akt-mTOR or c-MET signaling transduction pathways [5,9,10]. In another third of HCC patients, cell proliferation is lead by activation of Wnt pathway, mostly as a result of b-catenin mutations [11,12]. Genomic abnormalities driving proliferation in the remaining cases are still unclear. Therefore, there is rationale to combine drugs abrogating potent signals at different levels of one of the main pathways (e.g. blocking EGFR with erlotinib and Raf-Ras with sorafenib) or abrogating signals of two different pathways (e.g. VEGF with bevacizumab and mTOR with rapamycin, as in the study by Huynh et al.) [13]. Since there is not a single dominant molecular pathogenesis underlying all HCCs, it is increasingly clear that different models will be ultimately required to mimic different subclasses of the neoplasm.

Investigators at the front-line of drug development of targeted therapies in HCC are now facing two challenging questions. First, what is the best experimental model to assess new molecular targeted therapies in HCC, and second, if there are data to support a direct correlation between experimental findings and clinical outcomes in phase II–III studies in oncology and HCC.

2. Testing new drugs in pre-clinical HCC models

The demonstration that concentrated cancer cells grown in vitro could form tumors when implanted subcutaneously into an immunocompromised mouse was first established in 1969 [14]. This xenograft model has since demonstrated several advantages that explain its persistence as the mainstay of pre-clinical studies of antineoplastic drugs in vivo: the tumors are rapidly and easily induced, and their subcutaneous location enables direct measurement of tumor growth. More recently, however, several critical differences between xenograft- and patient-derived specimens have become apparent. Cancer is now appreciated as a complex disease dependent upon the interaction between transformed cells harboring oncogenic mutations and their surrounding tumor environment made up of normal cells, stromal cells, and immune cells [15].

One of the challenges we face in pre-clinical testing of targeted therapies in HCC is the lack of models that accurately recapitulate the disease in humans. Several key mouse models have been instrumental in defining the pathogenesis of HCC by introducing genetic alterations into one or more etiologic pathways that can be targeted exclusively to the liver [16]. Nonetheless, substantial challenges persist in modeling liver diseases whose natural history requires a chronic inflammatory milieu. Although these genetically modified mice have been employed to investigate the molecular pathways dysregulated in HCC, they are not commonly employed for pre-clinical drug testing, using either cytotoxic chemotherapeutic or molecularly targeted agents [16].

Pre-clinical testing, in HCC as in the majority of cancers, is typically performed in immune deficient mice using human tumor xenografts grown subcutaneously [16,17]. In the study published this month in the Journal by Huynh et al., the authors assess the efficacy of Bevacizumab and Rapamycin in two different nude mouse animal HCC culture models (Fig. 1). The first is a typical xenograft model in which the authors test 4 different HCC cell lines and 2 immortalized cirrhotic cell lines. This allows them to compare the sensitivity of the cancer cells and the non-cancer cells to the combined treatment, relative to the control and monotherapy treated groups. More importantly, they demonstrate that certain cell lines with oncogenic mutations are more susceptible to the drugs blocking the activated pathways than other cell lines, and describe an additive effect of the combination in tumor growth inhibition. The second model assesses the ability of cells to implant and metastasize to liver after intraperitoneal injection whilst undergoing the treatment regimens in question. Here the authors demonstrate a significant survival benefit of the bevacizumab/rapamycin combination, as opposed to the control and monotreatment groups. This experiment represents a useful departure from ectopic xenograft models in which metastases are rare. Overall, this study improves upon the routine xenograft model and demonstrates quite convincingly that the combination of the two therapies could merit further investigation in clinical trials.

Fig. 1.

Fig. 1.

Molecular targets of bevacizumab and rapamycin. Bevacizumab is a monoclonal antibody against VEGF, a ligand implicated in the proangiogenic response in human malignancies. Rapamycin is a small molecule blocking mTOR, an important downstream molecule of the Akt pathway. MTOR has been implicated in protein synthesis, cell growth and transition from G1 to S phase of the cell cycle. It can be activated through various growth factor tyrosine kinase receptors.

Novel models are emerging to test new drugs. One solution to the disparity between cancer cell lines and human tumors is surgical orthotopic implantation, in which intact fragments of human cancer taken directly from a patient are transplanted into the corresponding organ of immunodeficient rodents, as reviewed elsewhere [18]. Another alternative is to test new drugs in xenograft models generated from cultured cancer stem cells, the key target cells to assess efficacious drugs. Further possibilities include the use of mouse cell lines in immunocompetent mice with underlying liver fibrosis [19], a model that provides a unique tool for testing efficacy of drug combinations within the context of liver fibrosis, not likely possible in immune deficient mice. Finally, a more ambitious approach would be to test novel drugs in genetically engineered mice recapitulating specific pathway abnormalities (such as double transgenic TGF/c-MYC [20], transgenic of PDGFR [21], or transgenic for b-catenin [22]) in animals with an underlying fibrotic milieu. None of the latest models are currently ready for the conventional experimental studies [16].

3. Correlation between experimental findings and clinical trials

The validity of xenografts as a predictive indicator of probable clinical activity is limited, with the most success seen in cytotoxic agents [17]. A retrospective analysis performed by the NCI for 39 compounds in which both xenograft testing and phase II clinical data were available showed that in vivo activity in a particular tumor histology did not closely correlate with activity in the same human cancer, and that less than 50% of agents with activity in more than one-third of xenografts showed clinical activity [23]. A similar study from the NCI of Canada comparing drug activity in phase II clinical trials, human xenograft and mouse allografts showed that the human xenograft model was predictive for non-small cell lung cancer and ovarian cancers when panels of xenografts were used, but that these same models were not predictive for breast and colon cancers [24]. More recent reviews emphasize the predictive nature of the xenograft models when pharmacokinetically clinically equivalent drug doses are tested [25].

Targeted drugs tested in pre-clinical studies and the subsequent data in clinical trials in HCC are summarized in Table 1 [2646]. The only positive survival data reported with molecular therapies with sorafenib in HCC were preceded by strong positive pre-clinical experiments including evaluation in xenografts [27]. The remaining drugs with positive pre-clinical data have only been tested in the setting of small phase II studies, and thus the correlation between pre-clinical data and final clinical benefit (only coming from phase III studies) is difficult to predict. Although all drugs listed in Table 1 demonstrated pre-clinicial positive results, only some of them are likely to move forward according to phase II data (combinations with sorafenib, bevacizumab, erlotinib and rapamycin analogs), whereas others have shown limited results (gefitinib, cetuximab and bortezomib). Several variables may impact on the divergent outcomes compared to human disease. These include degree of heterogeneity of tumors in humans versus in cell lines; the molecular aberrations of the cell line chosen, ectopic versus orthotopic location of tumor, dosage and scheduling of the two compounds, and variability in selected end-points [47]. The greatest discrepancies between success of cancer therapies in xenograft models and in human clinical trials are likely due to critical differences in both the tumor cells and their microenvironment; this is a particularly relevant to HCC, which arises in an environment of inflammation and fibrosis.

Table 1. Targeted agents in pre-clinical models in HCC and clinical trial outcomes.

Target Cancer Agent or combination Pre-clinical studies with (+) outcomes Clinical trial outcomes in HCC
VEGFR/Raf HCC Sorafenib [2628] Phase III: survival benefit [29]
RCC
Breast
VEGFR/PDGFR HCC Sunitinib [30,31] Phase II [32,33] – survival: 11.6mo, TTR: 4.1mo
mTOR Gallbladder, ovarian, breast Rapamycin and its analogs everolimus, temsirolimus [3436] No data in HCC. RCC phase III: survival benefit [37]
EGFR HCC Erlotinib [38] Phase II [39,40]: Survival: 13mo; TTR: 3.2mo
EGFR HCC Gefitinib [41] Phase II [42]: negative
EGFR HCC Cetuximab [43] Phase II [32,44]: negative
VEGF HCC Bevacizumab No data Phase II [30,45,46]: TTR: 6.5mo
VEGF and EGFR HCC Bevacizumab and Erlotinib No data Phase II [40]: survival: 19mo

In conclusion, as in other malignancies, we are in dire need of accurate pre-clinical models of HCC that allow us to choose which molecularly targeted therapies and combinations thereof to advance to clinical trials. However, HCC is unique in two important ways: in the heterogeneity of the tumors amongst individuals and in the microenvironment of cirrhosis in the vast majority of affected patients. The paper by Huynh et al. addresses the first need by employing a number of cell lines with known mutations and dysregulated signaling pathways. It also addresses the need for testing in a metastatic model, although a model in which the metastatic disease burden was pre-established would more accurately mimic advanced HCC in humans [25].

In order to truly justify translation of a combination therapy study into clinical trials, strong pre-clinical support is essential. The best model to test these new compounds has not yet been defined in HCC, although some novel approaches are being proposed. In parallel, serum or tissue biomarkers of molecular signatures from tumors in humans should be obtained in early trials to understand their tumor biology [2], as was recently recommended by the panel of experts in trial design in HCC [48].

Grant Support:

P.N. is a recipient of an American Liver Foundation Fellowship. A.V. is supported by a grant from Fundación Caixa Galicia and a Sheila Sherlock Grant. J.M.L is supported by grants from the U.S. National Institute of Diabetes and Digestive and Kidney Diseases (1R01DK076986–01), The Samuel Waxman Cancer Research Foundation and the Spanish National Health Institute (SAF-2007–61898).

Footnotes

J.M.L. is consulting for Bayer Healthcare.

References

  • [1].Farazi PA, DePinho RA. Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer 2006;6:674–687. [DOI] [PubMed] [Google Scholar]
  • [2].Dancey JE, Chen HX. Strategies for optimizing combinations of molecularly targeted anticancer agents. Nat Rev Drug Discov 2006;5:649–659. [DOI] [PubMed] [Google Scholar]
  • [3].Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Raoul J, Zeuzem S, et al. for the SHARP Investigators. Sorafenib improves survival in advanced hepatocellular carcinoma (HCC): results of a phase III randomized placebo-controlled trial. J Clin Oncol 2007;25: LBA1. [Google Scholar]
  • [4].Chiang D, Villanueva A, Peix J, Newell P, Wurmbach E, Donovan D, et al. Integrative genomic classification of hepatitis C virus positive hepatocellular carcinoma. Hepatology 2007;46 (Suppl). Abstract #657. [Google Scholar]
  • [5].Villanueva A, Newell P, Chiang DY, Friedman SL, Llovet JM. Genomics and signaling pathways in hepatocellular carcinoma. Semin Liver Dis 2007;27:55–76. [DOI] [PubMed] [Google Scholar]
  • [6].Semela D, Dufour JF. Angiogenesis and hepatocellular carcinoma. J Hepatol 2004;41:864–880. [DOI] [PubMed] [Google Scholar]
  • [7].Hussain SP, Schwank J, Staib F, Wang XW, Harris CC. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene 2007;26:2166–2176. [DOI] [PubMed] [Google Scholar]
  • [8].Satyanarayana A, Manns MP, Rudolph KL. Telomeres and telomerase: a dual role in hepatocarcinogenesis. Hepatology 2004;40:276–283. [DOI] [PubMed] [Google Scholar]
  • [9].Calvisi DF, Ladu S, Gorden A, Farina M, Conner EA, Lee JS, et al. Ubiquitous activation of Ras and Jak/Stat pathways in human HCC. Gastroenterology 2006;130:1117–1128. [DOI] [PubMed] [Google Scholar]
  • [10].Lee JS, Heo J, Libbrecht L, Chu IS, Kaposi-Novak P, Calvisi DF, et al. A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progenitor cells. Nat Med 2006;12:410–416. [DOI] [PubMed] [Google Scholar]
  • [11].Boyault S, Rickman DS, de Reynies A, Balabaud C, Rebouissou S, Jeannot E, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 2007;45:42–52. [DOI] [PubMed] [Google Scholar]
  • [12].Buendia MA. Genetics of hepatocellular carcinoma. Semin Cancer Biol 2000;10:185–200. [DOI] [PubMed] [Google Scholar]
  • [13].Llovet J, Bruix J. Molecular targeted therapies in HCC. Hepatology 2008, in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Rygaard J, Povlsen CO. Heterotransplantation of a human malignant tumour to ‘‘Nude” mice. Acta Pathol Microbiol Scand 1969;77:758–760. [DOI] [PubMed] [Google Scholar]
  • [15].Frese KK, Tuveson DA. Maximizing mouse cancer models. Nat Rev Cancer 2007;7:645–658. [DOI] [PubMed] [Google Scholar]
  • [16].Newell P, Villanueva A, Friedman SL, Koike K, Llovet JM. Experimental models of hepatocellular carcinoma. J Hepatol 2008;48:858–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Huynh H, Soo KC, Chow PK, Panasci L, Tran E. Xenografts of human hepatocellular carcinoma: a useful model for testing drugs. Clin Cancer Res 2006;12:4306–4314. [DOI] [PubMed] [Google Scholar]
  • [18].Hoffman RM. Orthotopic metastatic (MetaMouse) models for discovery and development of novel chemotherapy. Methods Mol Med 2005;111:297–322. [DOI] [PubMed] [Google Scholar]
  • [19].Kornek M, Raskopf E, Tolba R, Becker U, Klockner M, Sauerbruch T, et al. Accelerated orthotopic hepatocellular carcinomas growth is linked to increased expression of pro-angiogenic and prometastatic factors in murine liver fibrosis. Liver Int 2008;28:509–518. [DOI] [PubMed] [Google Scholar]
  • [20].Lee JS, Chu IS, Mikaelyan A, Calvisi DF, Heo J, Reddy JK, et al. Application of comparative functional genomics to identify best-fit mouse models to study human cancer. Nat Genet 2004;36:1306–1311. [DOI] [PubMed] [Google Scholar]
  • [21].Campbell JS, Hughes SD, Gilbertson DG, Palmer TE, Holdren MS, Haran AC, et al. Platelet-derived growth factor C induces liver fibrosis, steatosis, and hepatocellular carcinoma. Proc Natl Acad Sci USA 2005;102:3389–3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Thompson MD, Monga SP. WNT/beta-catenin signaling in liver health and disease. Hepatology 2007;45:1298–1305. [DOI] [PubMed] [Google Scholar]
  • [23].Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti JM, Schepartz S, et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br J Cancer 2001;84:1424–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Voskoglou-Nomikos T, Pater JL, Seymour L. Clinical predictive value of the in vitro cell line, human xenograft, and mouse allograft preclinical cancer models. Clin Cancer Res 2003;9:4227–4239. [PubMed] [Google Scholar]
  • [25].Kerbel RS. Human tumor xenografts as predictive preclinical models for anticancer drug activity in humans: better than commonly perceived-but they can be improved. Cancer Biol Ther 2003;2:S134–S139. [PubMed] [Google Scholar]
  • [26].Chang YS, Adnane J, Trail PA, Levy J, Henderson A, Xue D, et al. Sorafenib (BAY 43–9006) inhibits tumor growth and vascularization and induces tumor apoptosis and hypoxia in RCC xenograft models. Cancer Chemother Pharmacol 2007;59:561–574. [DOI] [PubMed] [Google Scholar]
  • [27].Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, et al. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res 2006;66:11851–11858. [DOI] [PubMed] [Google Scholar]
  • [28].Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, et al. 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. [DOI] [PubMed] [Google Scholar]
  • [29].Zhu AX. Development of sorafenib and other molecularly targeted agents in hepatocellular carcinoma. Cancer 2008;112:250–259. [DOI] [PubMed] [Google Scholar]
  • [30].Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 2003;9:327–337. [PubMed] [Google Scholar]
  • [31].Bergers G, Song S, Meyer-Morse N, Bergsland E, Hanahan D. Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. J Clin Invest 2003;111:1287–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Zhu AX, Sahani DV, di Tomaso E, Duda D, Sindhwani V, Yoon SS, et al. A phase II study of sunitinib in patients with advanced hepatocellular carcinoma. J Clin Oncol 2007;25:4637. [Google Scholar]
  • [33].Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, et al. Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol 2006;24:25–35. [DOI] [PubMed] [Google Scholar]
  • [34].Rini BI. Temsirolimus, an inhibitor of mammalian target of rapamycin. Clin Cancer Res 2008;14:1286–1290. [DOI] [PubMed] [Google Scholar]
  • [35].Seeliger H, Guba M, Kleespies A, Jauch KW, Bruns CJ. Role of mTOR in solid tumor systems: a therapeutical target against primary tumor growth, metastases, and angiogenesis. Cancer Metastasis Rev 2007;26:611–621. [DOI] [PubMed] [Google Scholar]
  • [36].Sieghart W, Fuereder T, Schmid K, Cejka D, Werzowa J, Wrba F, et al. Mammalian target of rapamycin pathway activity in hepatocellular carcinomas of patients undergoing liver transplantation. Transplantation 2007;83:425–432. [DOI] [PubMed] [Google Scholar]
  • [37].Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007;356:2271–2281. [DOI] [PubMed] [Google Scholar]
  • [38].Huether A, Hopfner M, Sutter AP, Schuppan D, Scherubl H. Erlotinib induces cell cycle arrest and apoptosis in hepatocellular cancer cells and enhances chemosensitivity towards cytostatics. J Hepatol 2005;43:661–669. [DOI] [PubMed] [Google Scholar]
  • [39].Philip PA, Mahoney MR, Allmer C, Thomas J, Pitot HC, Kim G, et al. Phase II study of Erlotinib (OSI-774) in patients with advanced hepatocellular cancer. J Clin Oncol 2005;23:6657–6663. [DOI] [PubMed] [Google Scholar]
  • [40].Thomas MB, Iwasaki M, Glover K, Abbruzzese JL. The combination of bevacizumab (B) and erlotinib (E) shows significant biological activity in patients with advanced hepatocellular carcinoma. J Clin Oncol 2007;25:4567. [Google Scholar]
  • [41].Schiffer E, Housset C, Cacheux W, Wendum D, Desbois-Mouthon C, Rey C, et al. Gefitinib, an EGFR inhibitor, prevents hepatocellular carcinoma development in the rat liver with cirrhosis. Hepatology 2005;41:307–314. [DOI] [PubMed] [Google Scholar]
  • [42].O’Dwyer PJ, Levy DE, Kauh JS, Fitzgerald DB, Benson III AB. Gefitinib in advanced unresectable hepatocellular carcinoma: results from the Eastern Cooperative Oncology Group’s Study E1203. J Clin Oncol 2006;24:4143.16896003 [Google Scholar]
  • [43].Huether A, Hopfner M, Baradari V, Schuppan D, Scherubl H. EGFR blockade by cetuximab alone or as combination therapy for growth control of hepatocellular cancer. Biochem Pharmacol 2005;70:1568–1578. [DOI] [PubMed] [Google Scholar]
  • [44].Gruenwald V, Wilkens L, Gebel M, Greten TF, Kubicka S, Ganser A, et al. A phase II open-label study of cetuximab in unresectable hepatocellular carcinoma: final results. J Clin Oncol 2007;25:4598. [Google Scholar]
  • [45].Malka DC, Dromain C, Farace F, Horn S, Pignon J, Ducreux M, et al. Bevacizumab in patients with advanced hepatocellular carcinoma (HCC): preliminary results of a phase II study with circulating endothelial cell (CEC) monitoring. J Clin Oncol 2007;25:4570. [Google Scholar]
  • [46].Schwartz JD, Lehrer D. Bevacizumab in unresectable hepatocellular carcinoma (HCC) for patients without metastasis and without invasion of the portal vein. J Clin Oncol 2006;25:4144. [Google Scholar]
  • [47].Kelland LR. Of mice and men: values and liabilities of the athymic nude mouse model in anticancer drug development. Eur J Cancer 2004;40:827–836. [DOI] [PubMed] [Google Scholar]
  • [48].Llovet JM, Di Bisceglie A, Bruix J, Kramer B, Lencioni R, Zhu A, et al. Design and end-points of clinical trials in HCC. J Natl Cancer Inst 2008, in press. [DOI] [PubMed] [Google Scholar]

RESOURCES