Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma (Review)

  • Authors:
    • Sufang Yang
    • Guohua Liu
  • View Affiliations

  • Published online on: January 2, 2017     https://doi.org/10.3892/ol.2017.5557
  • Pages: 1041-1047
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although the biological basis of hepatocellular carcinoma (HCC) remains unclear, effective treatments and improvement of the survival rate remain worthwhile research goals. Abnormal protein signaling pathways contributing to uncontrolled cell proliferation, differentiation, survival and apoptosis are biomarkers of the carcinogenic process. Certain mutated components or overexpression of the rat sarcoma virus (Ras)/rapidly accelerated fibrosarcoma (Raf)/mitogen‑activated protein kinase kinase (MEK)/extracellular signal‑regulated kinase (ERK) signaling pathway are increasingly being studied in HCC carcinogenesis. The present review addresses the effect of the Ras/Raf/MEK/ERK signaling pathway on the pathogenesis of HCC, and provides an update on the preclinical and clinical development of various inhibitors targeting this core signaling pathway, which include various Ras inhibitors, Raf inhibitors and MEK inhibitors for HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common cancers, with an incidence rate ranking sixth highest and a mortality rate ranking third highest, accounting for 7% of all cancers in the world (1). In particular, there is a high incidence in China, with ~4/10,000 cases per year. Current treatment options for HCC, including surgical approaches, locoregional ablative techniques and interventional ablation treatments, could increase the 5-year-survival rate to 75% (vs. 30% prior to these treatments), however, <20% of HCC patients qualify for these treatments (2). Although the promising 5-year-survival rate of HCC cases has been increased due to advances in surgical techniques, nutritional support and perioperative management, long-term survival after surgical resection remains low due to the high rate of recurrence and metastasis (3,4). Novel evidence-based therapies for HCC are urgently required. Recently, biological studies have pointed to aberrant rapidly accelerated fibrosarcoma (Raf)/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway activation as being central for cancer growth, survival and motility, as well as for targeted therapy resistance mechanisms (57). For example, sorafenib is a Raf-1 kinase inhibitor and is the only approved drug therapy for HCC. In patients with advanced or metastatic HCC and compensated cirrhosis, sorafenib offers disease control in ~40% of treated patients, with a time to progression of 5.5 months and a median survival time of 10.7 months, ~3 months longer than that of placebo-treated patients (8). Hence, there is an eagerness to dissect the molecular mechanisms of invasion and metastasis for novel insights and interventions against the recurrence of HCC.

Undoubtedly, the rat sarcoma virus (Ras)/Raf/MEK/ERK signaling pathway contributes a core effect in regulating cell proliferation, differentiation and survival in the signaling networks (9). On this account, it has been studied and discussed to determine the pathogenesis of several types of human cancers, including HCC (10). Not merely the Ras/Raf/MEK/ERK signaling pathway, but also the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway has been studied to determine whether it is connected with the pathogenesis of HCC (11). It is notable that the interaction between the Ras/Raf/MEK/ERK and PI3K/Akt signaling pathways may lead to the regulation of cell growth and development, more than either alone.

In the present review, the function of the Ras/Raf/MEK/ERK signaling pathway in HCC is elaborated on and its therapeutic potential as a target for the intervention and treatment of HCC is expounded.

Ras/Raf/MEK/ERK pathway

The mitogen-activated protein kinase (MAPK) cascade consists of serine/threonine kinases, which convert extracellular molecules such as growth factors, hormones, tumor-promoting substances and differentiation factors, into intracellular signals for regulating cell proliferation, differentiation and survival (12,13). There are four core protein kinases, Ras, Raf, MEK and ERK, in the Ras/Raf/MEK/ERK signaling pathway. Ras, Raf and MEK are members of multi-gene families; of those, Ras has three members (Ki-Ras, N-Ras and Ha-Ras), Raf has three members (A-Raf, B-Raf and Raf-1) and MEK has five gene family members (MEK1, MEK2, MEK3, MEK4 and MEK5). At the cell surface, the activation of the Ras/Raf/MEK/ERK signaling pathway is initiated by ligand binding to receptor tyrosine kinases (RTK), then, in the nucleus, the phosphorylation of four core protein kinases, Ras, Raf, MEK and ERK, in turn regulating gene transcription (14). The specific activation pathways are as follows.

There are a number of extracellular signals, including growth factors, hormones, tumor-promoting substances and differentiation factors, which activate the Ras/Raf/MEK/ERK signaling pathway. When extracellular signals bond with an appropriate RTK (an Src homology 2 domain-containing protein), the C-terminus of the growth factor receptor [for example, FGFR, Flt-3, platelet-derived growth factor receptor (PDGFR), insulin-like growth factor receptor-1 (IGFR-1) and macrophage colony stimulating receptor among others] that has been activated is linked with the RTK. The tyrosine kinase domain of the excessive phosphorylation of RTK acting as a carrier protein, such as sex muscle abnormal protein-5 or organization control-1, recruits guanine nucleotide exchange factors [for example, mammalian son-of-sevenless (SOS)] to the cytomembrane where they stimulate Ras-GDP conversion to Ras-GTP, resulting in Ras protein activation (14,15). Ras phosphorylation then recruits Raf to the membrane where it becomes activated, likely via an Src-family tyrosine (Y) kinase. Raf is responsible for the serine/threonine phosphorylation of MEK1. The MEK family has five genes, namely MEK1, MEK2, MEK3, MEK4 and MEK5. The five genes are all double specificity kinases, which means that they can phosphorylate serine/threonine residues along with tyrosine residues. Of those, Ras and Raf activate downstream target proteins of MEK1 and MEK2 through phosphorylating the activation domain of two serine residues. MEK1 phosphorylates ERK1/2 at specific T and Y residues. The ERK family has four members, namely JNK1/2/3, ERK1/2, ERK5 and p38 MAPK. ERK1/2 is the only downstream protein target of MEK1/2 phosphorylation. When activating ERK1/2 serine/threonine kinases, they will generate a series of effects (for example, the phosphorylation and activation p90 ribosomal six kinase-1) (16,17). There are 460 ERK1/2 targets, including downstream and upstream substrates (18,19). Therefore, the regulation of the Ras/Raf/MEK/ERK signaling pathway plays an important role in cell proliferation, differentiation and survival by suppressing MEK and ERK activities.

There is a feedback pathway regulating the activity of B-Raf, Raf-1 and MEK1 through the activation of ERK. With regard to Raf-1, ERK phosphorylation can improve or lower the its activity, which depends on the site phosphorylated. With regard to B-Raf and MEK1, ERK phosphorylation can lower their activity. There is also a negative feedback pathway preventing the activation of Ras through the phosphorylation of SOS by ERK. Target protein phosphorylation, such as that of Ras, Raf, MEK and ERK, can enhance or inhibit the associated signaling pathway, even phosphorylating different sites of a target protein playing a different role in regulating the pathway (20,21). Therefore, regulating the Ras/Raf/MEK/ERK signaling pathway is a complex process, which plays an important role in cell proliferation, differentiation and survival (Fig. 1).

Ras/Raf/MEK/ERK pathway activation in HCC

A large amount of preclinical and clinical evidence has shown that the abnormal activation of the Ras/Raf/MEK/ERK signaling pathway frequently results in HCC. Ito et al showed that MAPK/ERK is activated and its associated gene expression is upregulated in 58% of HCC cases (22). Hoffmann et al demonstrated that the mRNA of Ras, MEK, ERK and MAPK was overexpressed in 33, 40, 50 and 50% of HCC patients, respectively (23). Similarly, H-ras has been found to be activated in ~93.8% of HCC cases (24). A study further showed that the expression Raf and its downstream genes, MEK and ERK, were upregulated in samples of hepatocirrhosis and HCC (25). Western blot analysis demonstrated the overexpression of Raf-1 in 91.2% of hepatocirrhosis and 100% of HCC patients. Furthermore, the Raf-1 expression level in HCC patients was significantly high compared with that of hepatocirrhosis patients (26). All research results showed that the activation of the Ras/Raf/MEK/ERK pathway may lead to HCC development functionally.

The cellular mechanisms behind the activation of the Ras/Raf/MEK/ERK signaling pathway are not yet completely clear in HCC. However, activation by RTKs is hypothesized to be the main mechanism. It has been verified that EGFR, IGFR, vascular endothelial growth factor receptor (VEGFR) and c-Met are overexpressed. Of those, EGFR accounts for 47.1% of cases in HCC, and the overexpression of EGFR is responsible for the invasiveness and recurrence of HCC (27). Wiedmann and Mössner showed that the EGFR inhibitors erlotinib and lapatinib inhibit not only RTKs, but also serine/threonine kinases along the Ras/Raf/MEK/ERK pathway, in two phase III placebo-controlled trials (28). Similarly, the overexpression of VEGFR has been found in HCC cell lines and in the serum and tissues of HCC patients (29,30). Furthermore, the overexpression of c-MET accounts for 20–48% of cases in HCC (31). Activation of c-MET plays a role in bringing the growth factor receptor-bound protein 2/SOS complex to the plasma membrane. As a result, GTP along with Ras activates a protein cascade that contributes to the downstream protein kinase phosphorylation of ERK by Raf and MEK (32).

Hepatitis virus infections also play an important role in the activation of the Ras/Raf/MEK/ERK pathway in HCC. Hepatitis B virus X protein has also been demonstrated to have an essential effect on the progression of HCC through activating the Ras/Raf/MEK/ERK cascade (33) and then contributing to the loss of function of the tumor suppressor p53 (34). Hepatitis C virus (HCV) core protein activates the kinase Raf-1 and MAPK/ERK pathway through interacting with 14–3-3 protein (35). Schmitz et al confirmed that the mechanism of HCC carcinogenesis may be via the activation of the Ras/Raf/MEK/ERK pathway by HCV infection (36). There are at least two functions of the HCV core protein, including the activation of the Ras/Raf/MEK/ERK pathway and an anti-apoptotic effect. HCV core protein can activate ERK phosphorylation alone without hepatocyte mitogen-mediated signaling. However, HCV core protein along with tissue plasminogen activator may contribute to the effect on MEK1 or further upstream of the protein kinase (37).

In recent years, genomic sequencing research has revealed the associated gene changes of the Ras/Raf/MEK/ERK signaling pathway in HCC. The B-Raf gene, one of the human isoforms of Raf, has been found to be mutated or deleted in HCC, accounting for ~23% of cases. As a result, this may lead to activating oncogenic Ras in HCC (38). In addition to the mutation or deletion of B-Raf, codon 12 of the K-Ras and N-Ras genes is also mutated or deleted in HCC, accounting for 4.69 and 41.37% of cases (24,38,39). Challen et al found that mutations of the K-ras and N-ras genes in codon 61 occurs in 5.3 and 15.8% of HCC cases (40). All the reported mutations of K-Ras, N-Ras and H-ras are somatic missense mutations (for example, changes to the amino acids of codons 12, 13 and 61) in HCC. Mutations in Ras family genes can phosphorylate the Ras/Raf/MEK/ERK signaling pathway, then deregulate signal transduction (41). However, Taketomi et al examined 61 patients through a dot-blot and elaborated that the mutations of Ras proto-oncogenes in codons 12, 13 and 61 had little effect in HCC (42). Therefore, further study is required to clarify the controversy.

Targeting Ras/Raf/MEK/ERK pathway in HCC

As the abnormal activation of the Ras/Raf/MEK/ERK signaling pathway plays a major role in HCC cell proliferation, differentiation, survival and apoptosis, a number of studies have been focused on the inhibitors of the core protein kinases Ras, Raf, MEK and ERK in the Ras/Raf/MEK/ERK signaling pathway (Fig. 1); a number of these studies are preclinical, while others are clinical studies (Table I).

Table I.

Inhibitors of the Raf/MEK/ERK signaling pathway for HCC in preclinical and clinical studies.

Table I.

Inhibitors of the Raf/MEK/ERK signaling pathway for HCC in preclinical and clinical studies.

DrugTargetsPhaseOutcomeRef.
SalirasibRasPreclinicalIC50 in µM range, therapeutic potential of HCC was further confirmed in a xenograft model(48)
SorafenibRafPhase IIISignificantly improved OS and TTP(55)
NovartisRafPreclinicalInhibition the growth rates of HCC cells and xenograft tumors(56)
U0126MEKPreclinicalNot favorable for clinical use due to poor solubility and low bioavailability(62)
PD098059MEKPreclinicalNot favorable for clinical use due to poor solubility and low bioavailability(59)
CI-1040MEKPhase I28% SD for 5.5 months(64)
PD0325901MEKPhase IIAll patients experienced at least one adverse event(76)
SelumetinibMEKPhase II35% SD for 6 weeks; PFS, 1.4 months; TTP, 1.4 months; OS, 4.2 months(73)

[i] MEK, mitogen-activated protein kinase kinase; IC50, half maximal inhibitory concentration; HCC, hepatoceullar carcinoma; OS, overall survival; TTP, time to progression; SD, stable disease; PFS, progression-free survival; ERK, extracellular-regulated protein kinase.

Targeting Ras

Activating Ras mutations have been observed in ~30% of all cancers. However, according to studies of the pathogenesis of cancer at present, the specific function of Ras is not yet settled. Salirasib [also known as S-trans, trans-farnesylthiosalycilic acid (FTS)] is a synthetic low-weight molecule of a S-farnesyl cysteine analog that expresses a potent inhibitory effect on Ras. Its mechanism of action is likely to be associated primarily with the dislodgment of the mature protein from membrane domains that interact with Ras and with the subsequent accelerated degradation of the dislodged Ras proteins (43,44). These effects of FTS are manifested by a decrease in the amount of cellular Ras accompanied by interruption of the Ras-dependent Raf-1/ERK signaling cascade (45). FTS plays an antitumoral role in several non-hepatic cancer cell lines (46), and a phase I clinical trial of gemcitabine and FTS has demonstrated that FTS is well tolerated in patients with solid non-liver tumors (47). As its expression is well tolerated, salirasib may become a drug of choice for the treatment of HCC by targeting Ras and mammalian target of rapamycin (mTOR) protein kinase (48). A study by da Silva Morais et al indicated that a high concentration of salirasib can inhibit liver cancer cell proliferation in vivo in rats following a partial liver resection (49). Its mechanism of inhibitory effects is mediated, at least in part, by the inhibition of ERK phosphorylation. Furthermore, the study by Nicolas et al demonstrated that salirasib injection can prevent the development of liver tumors in a subcutaneous xenograft model (50).

Targeting Raf

Certain special distinct classes of compounds have been developed as potential Raf kinase inhibitors. However, thus far, sorafenib (Nexavar) is the most successful anti-Raf inhibitor. Sorafenib, an orally available anti-Raf compound, is the only small molecular target kinase to receive Food and Drug Administration approval for the treatment of advanced HCC (51). Sorafenib suppresses the serine/threonine kinase subtypes of Raf, which are well known to regulate the Raf/MEK/ERK signaling pathway and inhibit tyrosine kinase receptors, including VEGFR2, PDGFR and IGFR. For this reason, sorafenib inhibits angiogenesis and tumor growth (52). Sorafenib has demonstrable preclinical and clinical activity against certain types of cancer (for example, HCC, and ovarian, breast and pancreatic cancer). However, in pharmacokinetics, pharmacodynamics and adverse events, sorafenib has exhibited individual differences in clinical research (53,54). An updated meta-analysis evaluated sorafenib administration and found that it could significantly increase OS time and TTP in patients with advanced HCC. Additional large-scale, well-designed randomized controlled trials are planned to further evaluate the efficacy of treating advanced HCC with sorafenib (55).

NVP-AAL881 (Novartis, Basel, Switzerland) is an oral Raf and VEGFR2 small molecule inhibitor that has been shown to inhibit cell proliferation and tumor growth in a subcutaneous xenograft model of HCC (56). NVP-AAL881 can abnormally activate ERK and STAT3, and inhibits the migration of HCC cells. NVP-AAL881 inhibits the growth of HCC cells in a dose-dependent manner, as observed through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. In HCC cells under low serum culture conditions, the inhibition effect of NVP-AAL881 is enhanced. Furthermore, with increasing concentrations of NVP-AAL881, the inhibition of ERK, MEK and STAT3 phosphorylation is enhanced in HCC cell lines (57). More recently, it has been shown that NVP-AAL881 administration markedly inhibits the growth of HCC xenograft tumors compared with controls (58).

Targeting MEK

As Ras inhibitors are challenging to identify and almost no biological functions of B-Raf inhibitors are known, more attention is being focused on the study of MEK and ERK inhibitors. Numerous MEK inhibitors have been developed; PD98059 was the first MEK1/2 inhibitor to be found, which combines with the inactive forms of MEK1/2 to prevent its phosphorylation, then inhibits the phosphorylation of ERK1/2 and blocks cell signal transduction (59). U0126 is the second MEK1/2 inhibitor to be identified, and its inhibitive effect is greater than that of PD98059. U0126 is widely used in in vitro experiments (60). In HepG2 cell cultures in vitro, the phosphorylation of p38 and ERK1/2 was inhibited by PD98059, and this effect was also demonstrated in vivo (61). The two types of MEK1/2 kinase inhibitors are non-ATP-competitive, acting by inhibiting the MEK excitation instead of directly inhibiting the activity of MEK. Due to the low solubility and bioavailability of PD98059 and U0126, they have not entered into clinical trials and are only applied in experiments in vitro (62).

CI-1040 is the first MEK1/2 inhibitor to enter into a clinical trial (63). Despite effective inhibition of the MEK pathway and demonstration of antitumor activity in HCC cell models, its development was halted after phase II trials due to poor clinical efficacy (64,65). However, the reduced adverse events associated with CI-1040 has led to more investigation of potent analogues. PD0325901 is the congener of CI-1040, which was found through replacing the hydroxyl and fluorine of CI-1040. It was also to optimize diphenylamine of CI-1040 through pharmacological probe in the study of the Ras/Raf/MEK/ERK signaling pathway (66). To improve the poor solubility and rapid clearance of CI-1040, the diphenylamine core and the hydroxamate side chain were optimized with resulting improvements in solubility, potency in cell-based assays and oral bioavailability. Compared with CI-1040, this compound represents a 100-fold improvement in potency in cell-based assays (0.33 nM in colon 26 cells) and, perhaps more significantly, greater solubility and better stability in human liver microsomes and hepatocytes. Due to its increased solubility, compared with CI-1040, the pharmacological effect and biopharmaceutical properties are significantly improved (63). However, as all patients treated in the phase II trial of the drug experienced at least one adverse event, PD0325901 could not be developed further (67,68).

The oral, non-ATP competitive MEK1/2 inhibitor selumetinib (previously known as ARRY-142886 and AZD6244), is a benzimidazole derivative (69). The drug is the second MEK inhibitor to enter into clinical trials (70). A large quantity of effective results have been shown in preclinical studies only, using cell cultures and animal models (71). Selumetinib has been demonstrated to play a role in contributing to the inhibition of ERK1/2 phosphorylation in a number of cancer cell lines, with a half maximal inhibitory concentration as low as 8 nmol/l and the sustained inhibition of ERK activity achieved with a concentration of 10 mg/kg/day in a subcutaneous xenograft model of HCC (72). In a multi-center, single-arm phase II study of selumetinib in advanced or metastatic HCC patients, the study was stopped at the interim analysis due to a lack of radiographic reaction. The drug has not yet been shown to significantly decrease the time to progression. Selumetinib is well tolerated, but the treatment effect is not ideal in advanced HCC (73). Another phase II study showed that 14 out of 17 evaluable HCC patients succumbed; of the remaining 3 patients, 2 experienced progression and 1 remained alive without progression (74). The median progression-free survival time was 1.4 months. The median time to progression was the same. The median survival time was 4.2 months (75).

Conclusion

Studying the Ras/Raf/MEK/ERK signaling pathway has provided novel insights and novel target drugs for HCC treatment. On the one hand, although such drugs exhibit improved therapeutic effects compared with conventional chemotherapeutic drugs, they present potential problems and challenges for HCC therapy, such as adverse events and resistance. On the other hand, although the Ras/Raf/MEK/ERK signaling pathway plays an important role in the regulation of HCC cell proliferation, differentiation, survival and apoptosis, its exact functional relevance in the settings of this complex signaling network and HCC tumorigenesis are far from being fully understood. Furthermore, a key challenge for Ras/Raf/MEK/ERK pathway inhibition will likely be the level of cross-talk and negative feedback along parallel pathways (such as the PI3K/AKT/mTOR pathway). Preclinical data suggest that certain problems and challenges may be overcome by combining Ras/Raf/MEK/ERK pathway inhibitors with other pathway inhibitors, but this must be confirmed in clinical studies.

References

1 

He W, Zeng Q, Zheng Y, Chen M, Shen J, Qiu J, Chen M, Zou R, Liao Y, Li Q, et al: The role of clinically significant portal hypertension in hepatic resection for hepatocellular carcinoma patients: A propensity score matching analysis. BMC Cancer. 15:2632015. View Article : Google Scholar : PubMed/NCBI

2 

Yang B, Zan RY, Wang SY, Li XL, Wei ML, Guo WH, You X, Li J and Liao ZY: Radiofrequency ablation versus percutaneous ethanol injection for hepatocellular carcinoma: A meta-analysis of randomized controlled trials. World J Surg Oncol. 13:962015. View Article : Google Scholar : PubMed/NCBI

3 

Baek YH, Kim KT, Lee SW, Jeong JS, Park BH, Nam KJ, Cho JH, Kim YH, Roh YH, Lee HS, et al: Efficacy of hepatic arterial infusion chemotherapy in advanced hepatocellular carcinoma. World J Gastroenterol. 18:3426–3434. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Kim JY, Chung SM, Choi BO and Kay CS: Hepatocellular carcinoma with portal vein tumor thrombosis: Improved treatment outcomes with external beam radiation therapy. Hepatol Res. 41:813–824. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Buscà R, Pouysségur J and Lenormand P: ERK1 and ERK2 Map Kinases: Specific Roles or Functional Redundancy? Front Cell Dev Biol. 4:532016. View Article : Google Scholar : PubMed/NCBI

6 

Wang Y, Nie H, Zhao X, Qin Y and Gong X: Bicyclol induces cell cycle arrest and autophagy in HepG2 human hepatocellular carcinoma cells through the PI3K/AKT and Ras/Raf/MEK/ERK pathways. BMC Cancer. 16:7422016. View Article : Google Scholar : PubMed/NCBI

7 

Asati V, Mahapatra DK and Bharti SK: PI3K/Akt/mTOR and Ras/Raf/MEK/ERK signaling pathways inhibitors as anticancer agents: Structural and pharmacological perspectives. Eur J Med Chem. 109:314–341. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Lee SH, Song IH, Noh R, Kang HY, Kim SB, Ko SY, Lee ES, Kim SH, Lee BS, Kim AN, et al: Clinical outcomes of patients with advanced hepatocellular carcinoma treated with sorafenib: A retrospective study of routine clinical practice in multi-institutions. BMC Cancer. 15:2362015. View Article : Google Scholar : PubMed/NCBI

9 

Yu P, Ye L, Wang H, Du G, Zhang J, Zhang J and Tian J: NSK-01105 inhibits proliferation and induces apoptosis of prostate cancer cells by blocking the Raf/MEK/ERK and PI3K/Akt/mTOR signal pathways. Tumour Biol. 36:2143–2153. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Huynh H, Ngo VC, Koong HN, Poon D, Choo SP, Toh HC, Thng CH, Chow P, Ong HS, Chung A, et al: AZD6244 enhances the anti-tumor activity of sorafenib in ectopic and orthotopic models of human hepatocellular carcinoma (HCC). J Hepatol. 52:79–87. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Zhou Q, Lui VW and Yeo W: Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Future Oncol. 7:1149–1167. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Jiang M, Wen F, Cao J, Li P, She J and Chu Z: Genome-wide exploration of the molecular evolution and regulatory network of mitogen-activated protein kinase cascades upon multiple stresses in Brachypodium distachyon. BMC Genomics. 16:2282015. View Article : Google Scholar : PubMed/NCBI

13 

Ward AF, Braun BS and Shannon KM: Targeting oncogenic Ras signaling in hematologic malignancies. Blood. 120:3397–3406. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Knight T and Irving JA: Ras/Raf/MEK/ERK pathway activation in childhood acute lymphoblastic leukemia and its therapeutic targeting. Front Oncol. 4:1602014. View Article : Google Scholar : PubMed/NCBI

15 

Chang YS, Liu JC, Fu HQ, Yu BT, Zou SB, Wu QC and Wan L: Roles of targeting Ras/Raf/MEK/ERK signaling pathways in the treatment of esophageal carcinoma. Yao Xue Xue Bao. 48:635–641. 2013.(In Chinese). PubMed/NCBI

16 

Zebisch A and Troppmair J: Back to the roots: The remarkable RAF oncogene story. Cell Mol Life Sci. 63:1314–1330. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Moodie SA and Wolfman A: The 3Rs of life: Ras, Raf and growth regulation. Trends Genet. 10:44–48. 1994. View Article : Google Scholar : PubMed/NCBI

18 

De Luca A, Maiello MR, D'Alessio A, Pergameno M and Normanno N: The RAS/RAF/MEK/ERK and the PI3K/AKT signaling pathways: Role in cancer pathogenesis and implications for therapeutic approaches. Expert Opin Ther Targets. 16:(Suppl 2). S17–S27. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Steelman LS, Franklin RA, Abrams SL, Chappell W, Kempf CR, Bäsecke J, Stivala F, Donia M, Fagone P, Nicoletti F, et al: Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy. Leukemia. 25:1080–1094. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Trujillo JI: MEK inhibitors: A patent review 2008–2010. Expert Opin Ther Pat. 21:1045–1069. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Chung E and Kondo M: Role of Ras/Raf/MEK/ERK signaling in physiological hematopoiesis and leukemia development. Immunol Res. 49:248–268. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Ito Y, Sasaki Y, Horimoto M, Wada S, Tanaka Y, Kasahara A, Ueki T, Hirano T, Yamamoto H, Fujimoto J, et al: Activation of mitogen-activated protein kinases/extracellular signal-regulated kinases in human hepatocellular carcinoma. Hepatology. 27:951–958. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Hoffmann K, Shibo L, Xiao Z, Longerich T, Büchler MW and Schemmer P: Correlation of gene expression of ATP-binding cassette protein and tyrosine kinase signaling pathway in patients with hepatocellular carcinoma. Anticancer Res. 31:3883–3890. 2011.PubMed/NCBI

24 

Zuo Q, Huang H, Shi M, Zhang F, Sun J, Bin J, Liao Y and Liao W: Multivariate analysis of several molecular markers and clinicopathological features in postoperative prognosis of hepatocellular carcinoma. Anat Rec (Hoboken). 295:423–431. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, Wilhelm S, Lynch M and 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. 66:11851–11858. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Gollob JA, Wilhelm S, Carter C and Kelley SL: Role of Raf kinase in cancer: Therapeutic potential of targeting the Raf/MEK/ERK signal transduction pathway. Semin Oncol. 33:392–406. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Tang Z, Qin L, Wang X, Zhou G, Liao Y, Weng Y, Jiang X, Lin Z, Liu K and Ye S: Alterations of oncogenes, tumor suppressor genes and growth factors in hepatocellular carcinoma: With relation to tumor size and invasiveness. Chin Med J (Engl). 111:313–318. 1998.PubMed/NCBI

28 

Wiedmann MW and Mössner J: Molecular targeted therapy of hepatocellular carcinoma-results of the first clinical studies. Curr Cancer Drug Targets. 11:714–733. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Poon RT, Ho JW, Tong CS, Lau C, Ng IO and Fan ST: Prognostic significance of serum vascular endothelial growth factor and endostatin in patients with hepatocellular carcinoma. Br J Surg. 91:1354–1360. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Dhar DK, Naora H, Yamanoi A, Ono T, Kohno H, Otani H and Nagasue N: Requisite role of VEGF receptors in angiogenesis of hepatocellular carcinoma: A comparison with angiopoietin/Tie pathway. Anticancer Res. 22:379–386. 2002.PubMed/NCBI

31 

Tavian D, De Petro G, Benetti A, Portolani N, Giulini SM and Barlati S: u-PA and c-MET mRNA expression is co-ordinately enhanced while hepatocyte growth factor mRNA is down-regulated in human hepatocellular carcinoma. Int J Cancer. 87:644–649. 2000. View Article : Google Scholar : PubMed/NCBI

32 

Whittaker S, Marais R and Zhu AX: The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene. 29:4989–5005. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Giambartolomei S, Covone F, Levrero M and Balsano C: Sustained activation of the Raf/MEK/Erk pathway in response to EGF in stable cell lines expressing the Hepatitis C Virus (HCV) core protein. Oncogene. 20:2606–2610. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Wang XW: Microinjection technique used to study functional interaction between p53 and hepatitis B virus X gene in apoptosis. Mol Biotechnol. 18:169–177. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Nakamura H, Aoki H, Hino O and Moriyama M: HCV core protein promotes heparin binding EGF-like growth factor expression and activates Akt. Hepatol Res. 41:455–462. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Schmitz KJ, Wohlschlaeger J, Lang H, Sotiropoulos GC, Malago M, Steveling K, Reis H, Cicinnati VR, Schmid KW and Baba HA: Activation of the ERK and AKT signaling pathway predicts poor prognosis in hepatocellular carcinoma and ERK activation in cancer tissue is associated with hepatitis C virus infection. J Hepatol. 48:83–90. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Hayashi J, Aoki H, Kajino K, Moriyama M, Arakawa Y and Hino O: Hepatitis C virus core protein activates the MAPK/ERK cascade synergistically with tumor promoter TPA, but not with epidermal growth factor or transforming growth factor alpha. Hepatology. 32:958–961. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Colombino M, Sperlongano P, Izzo F, Tatangelo F, Botti G, Lombardi A, Accardo M, Tarantino L, Sordelli I, Agresti M, et al: BRAF and PIK3CA genes are somatically mutated in hepatocellular carcinoma among patients from South Italy. Cell Death Dis. 3:e2592012. View Article : Google Scholar : PubMed/NCBI

39 

Kalinina O, Marchio A, Urbanskii AI, Tarkova AB, Rebbani K, Granov DA, Dejean A, Generalov MI and Pineau P: Somatic changes in primary liver cancer in Russia: A pilot study. Mutat Res. 755:90–99. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Challen C, Guo K, Collier JD, Cavanagh D and Bassendine MF: Infrequent point mutations in codons 12 and 61 of ras oncogenes in human hepatocellular carcinomas. J Hepatol. 14:342–346. 1992. View Article : Google Scholar : PubMed/NCBI

41 

Saini KS, Loi S, de Azambuja E, Metzger-Filho O, Saini ML, Ignatiadis M, Dancey JE and Piccart-Gebhart MJ: Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev. 39:935–946. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Taketomi A, Shirabe K, Muto J, Yoshiya S, Motomura T, Mano Y, Ikegami T, Yoshizumi T, Sugio K and Maehara Y: A rare point mutation in the Ras oncogene in hepatocellular carcinoma. Surg Today. 43:289–292. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Marom M, Haklai R, Ben-Baruch G, Marciano D, Egozi Y and Kloog Y: Selective inhibition of Ras-dependent cell growth by farnesylthiosalisylic acid. J Biol Chem. 270:22263–22270. 1995. View Article : Google Scholar : PubMed/NCBI

44 

Haklai R, Weisz MG, Elad G, Paz A, Marciano D, Egozi Y, Ben-Baruch G and Kloog Y: Dislodgment and accelerated degradation of Ras. Biochemistry. 37:1306–1314. 1998. View Article : Google Scholar : PubMed/NCBI

45 

McMahon LP, Yue W, Santen RJ and Lawrence JC Jr: Farnesylthiosalicylic acid inhibits mammalian target of rapamycin (mTOR) activity both in cells and in vitro by promoting dissociation of the mTOR-raptor complex. Mol Endocrinol. 19:175–183. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Blum R, Elkon R, Yaari S, Zundelevich A, Jacob-Hirsch J, Rechavi G, Shamir R and Kloog Y: Gene expression signature of human cancer cell lines treated with the ras inhibitor salirasib (S-farnesylthiosalicylic acid). Cancer Res. 67:3320–3328. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Tsimberidou AM, Rudek MA, Hong D, Ng CS, Blair J, Goldsweig H and Kurzrock R: Phase 1 first-in-human clinical study of S-trans, trans-farnesylthiosalicylic acid (salirasib) in patients with solid tumors. Cancer Chemother Pharmacol. 65:235–241. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Charette N, De Saeger C, Lannoy V, Horsmans Y, Leclercq I and Stärkel P: Salirasib inhibits the growth of hepatocarcinoma cell lines in vitro and tumor growth in vivo through ras and mTOR inhibition. Mol Cancer. 9:2562010. View Article : Google Scholar : PubMed/NCBI

49 

da Silva Morais A, Saliez A, Leclercq I, Horsmans Y and Stärkel P: Inhibition of the Ras oncoprotein reduces proliferation of hepatocytes in vitro and in vivo in rats. Clin Sci (Lond). 114:73–83. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Schneider-Merck T, Borbath I, Charette N, De Saeger C, Abarca J, Leclercq I, Horsmans Y and Stärkel P: The Ras inhibitor farnesylthiosalicyclic acid (FTS) prevents nodule formation and development of preneoplastic foci of altered hepatocytes in rats. Eur J Cancer. 45:2050–2060. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Wecksler AT, Hwang SH, Liu JY, Wettersten HI, Morisseau C, Wu J, Weiss RH and Hammock BD: Biological evaluation of a novel sorafenib analogue, t-CUPM. Cancer Chemother Pharmacol. 75:161–171. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Fucile C, Marenco S, Bazzica M, Zuccoli ML, Lantieri F, Robbiano L, Marini V, Di Gion P, Pieri G, Stura P, et al: Measurement of sorafenib plasma concentration by high-performance liquid chromatography in patients with advanced hepatocellular carcinoma: Is it useful the application in clinical practice? A pilot study. Med Oncol. 32:3352015. View Article : Google Scholar : PubMed/NCBI

53 

Guan YS and He Q: Sorafenib: Activity and clinical application in patients with hepatocellular carcinoma. Expert Opin Pharmacother. 12:303–313. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Arrondeau J, Mir O, Boudou-Rouquette P, Coriat R, Ropert S, Dumas G, Rodrigues MJ, Rousseau B, Blanchet B and Goldwasser F: Sorafenib exposure decreases over time in patients with hepatocellular carcinoma. Invest New Drugs. 30:2046–2049. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Peng S, Zhao Y, Xu F, Jia C, Xu Y and Dai C: An updated meta-analysis of randomized controlled trials assessing the effect of sorafenib in advanced hepatocellular carcinoma. PLoS One. 9:e1125302014. View Article : Google Scholar : PubMed/NCBI

56 

Sathornsumetee S, Hjelmeland AB, Keir ST, McLendon RE, Batt D, Ramsey T, Yusuff N, Rasheed BK, Kieran MW, Laforme A, et al: AAL881, a novel small molecule inhibitor of RAF and vascular endothelial growth factor receptor activities, blocks the growth of malignant glioma. Cancer Res. 66:8722–8730. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Lang SA, Schachtschneider P, Moser C, Mori A, Hackl C, Gaumann A, Batt D, Schlitt HJ, Geissler EK and Stoeltzing O: Dual targeting of Raf and VEGF receptor 2 reduces growth and metastasis of pancreatic cancer through direct effects on tumor cells, endothelial cells, and pericytes. Mol Cancer Ther. 7:3509–3518. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Lang SA, Brecht I, Moser C, Obed A, Batt D, Schlitt HJ, Geissler EK and Stoeltzing O: Dual inhibition of Raf and VEGFR2 reduces growth and vascularization of hepatocellular carcinoma in an experimental model. Langenbecks Arch Surg. 393:333–341. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Cotrim CZ, Amado FL and Helguero LA: Estrogenic effect of the MEK1 inhibitor PD98059 on endogenous estrogen receptor alpha and beta. J Steroid Biochem Mol Biol. 124:25–30. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Han Y, Xu G, Zhang J, Yan M, Li X, Ma B, Jun L, Wang SJ and Tan J: Leptin induces osteocalcin expression in ATDC5 cells through activation of the MAPK-ERK1/2 signaling pathway. Oncotarget. Aug 24–2016.(Epub ahead of print).

61 

Davis JE, Xie X, Guo J, Huang W, Chu WM, Huang S, Teng Y and Wu G: ARF1 promotes prostate tumorigenesis via targeting oncogenic MAPK signaling. Oncotarget. 7:39834–39845. 2016.PubMed/NCBI

62 

Montagut C and Settleman J: Targeting the RAF-MEK-ERK pathway in cancer therapy. Cancer Lett. 283:125–134. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Barrett SD, Bridges AJ, Dudley DT, Saltiel AR, Fergus JH, Flamme CM, Delaney AM, Kaufman M, LePage S, Leopold WR, et al: The discovery of the benzhydroxamate MEK inhibitors CI-1040 and PD 0325901. Bioorg Med Chem Lett. 18:6501–6504. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Lorusso PM, Adjei AA, Varterasian M, Gadgeel S, Reid J, Mitchell DY, Hanson L, DeLuca P, Bruzek L, Piens J, et al: Phase I and pharmacodynamic study of the oral MEK inhibitor CI-1040 in patients with advanced malignancies. J Clin Oncol. 23:5281–5293. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Rinehart J, Adjei AA, Lorusso PM, Waterhouse D, Hecht JR, Natale RB, Hamid O, Varterasian M, Asbury P, Kaldjian EP, et al: Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol. 22:4456–4462. 2004. View Article : Google Scholar : PubMed/NCBI

66 

Ciuffreda L, Del Bufalo D, Desideri M, Di Sanza C, Stoppacciaro A, Ricciardi MR, Chiaretti S, Tavolaro S, Benassi B, Bellacosa A, et al: Growth-inhibitory and antiangiogenic activity of the MEK inhibitor PD0325901 in malignant melanoma with or without BRAF mutations. Neoplasia. 11:720–731. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Lorusso PM, Krishnamurthi SS, Rinehart JJ, Nabell LM, Malburg L, Chapman PB, DePrimo SE, Bentivegna S, Wilner KD, Tan W and Ricart AD: Phase I pharmacokinetic and pharmacodynamic study of the oral MAPK/ERK kinase inhibitor PD-0325901 in patients with advanced cancers. Clin Cancer Res. 16:1924–1937. 2010. View Article : Google Scholar : PubMed/NCBI

68 

Haura EB, Ricart AD, Larson TG, Stella PJ, Bazhenova L, Miller VA, Cohen RB, Eisenberg PD, Selaru P, Wilner KD and Gadgeel SM: A phase II study of PD-0325901, an oral MEK inhibitor, in previously treated patients with advanced non-small cell lung cancer. Clin Cancer Res. 16:2450–2457. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Davies BR, Logie A, McKay JS, Martin P, Steele S, Jenkins R, Cockerill M, Cartlidge S and Smith PD: AZD6244 (ARRY-142886), a potent inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 kinases: Mechanism of action in vivo, pharmacokinetic/pharmacodynamic relationship, and potential for combination in preclinical models. Mol Cancer Ther. 6:2209–2219. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Do K, Speranza G, Bishop R, Khin S, Rubinstein L, Kinders RJ, Datiles M, Eugeni M, Lam MH, Doyle LA, et al: Biomarker-driven phase 2 study of MK-2206 and selumetinib (AZD6244, ARRY-142886) in patients with colorectal cancer. Invest New Drugs. 33:720–728. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Grasso S, Pereira GJ, Palmeira-Dos-Santos C, Calgarotto AK, Martínez-Lacaci I, Ferragut JA, Smaili SS and Bincoletto C: Autophagy regulates Selumetinib (AZD6244) induced-apoptosis in colorectal cancer cells. Eur J Med Chem. 122:611–618. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Yang QJ, Huo Y, Han YL, Wan LL, Li J, Huang JL, Lu J, Chen PG, Gan R and Guo C. Cheng: Selumetinib attenuate skeletal muscle wasting in murine cachexia model through ERK inhibition and AKT activation. Mol Cancer Ther. Sep 6–2016.(Epub ahead of print).

73 

O'Neil BH, Goff LW, Kauh JS, Strosberg JR, Bekaii-Saab TS, Lee RM, Kazi A, Moore DT, Learoyd M, Lush RM, et al: Phase II study of the mitogen-activated protein kinase 1/2 inhibitor selumetinib in patients with advanced hepatocellular carcinoma. J Clin Oncol. 29:2350–2356. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Holkova B, Zingone A, Kmieciak M, Bose P, Badros AZ, Voorhees PM, Baz R, Korde N, Lin HY, Chen JQ, et al: Phase II trial of AZD6244 (Selumetinib, ARRY-142886), an oral MEK1/2 inhibitor, in relapsed/refractory multiple myeloma. Clin Cancer Res. 22:1067–1075. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Coleman RL, Sill MW, Thaker PH, Bender DP, Street D, McGuire WP, Johnston CM and Rotmensch J: A phase II evaluation of selumetinib (AZD6244, ARRY-142886), a selective MEK-1/2 inhibitor in the treatment of recurrent or persistent endometrial cancer: An NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol. 138:30–35. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Barrett SD, Bridges AJ, Dudley DT, Saltiel AR, Fergus JH, Flamme CM, Delaney AM, Kaufman M, LePage S, Leopold WR, et al: The discovery of the benzhydroxamate MEK inhibitors CI-1040 and PD 0325901. Bioorg Med Chem Lett. 18:6501–6504. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2017
Volume 13 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang S and Yang S: Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma (Review). Oncol Lett 13: 1041-1047, 2017
APA
Yang, S., & Yang, S. (2017). Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma (Review). Oncology Letters, 13, 1041-1047. https://doi.org/10.3892/ol.2017.5557
MLA
Yang, S., Liu, G."Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma (Review)". Oncology Letters 13.3 (2017): 1041-1047.
Chicago
Yang, S., Liu, G."Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma (Review)". Oncology Letters 13, no. 3 (2017): 1041-1047. https://doi.org/10.3892/ol.2017.5557