Cancer Letters

Cancer Letters

Volume 283, Issue 2, 8 October 2009, Pages 125-134
Cancer Letters

Mini-review
Targeting the RAF–MEK–ERK pathway in cancer therapy

https://doi.org/10.1016/j.canlet.2009.01.022Get rights and content

Abstract

The clinical success of selective kinase inhibitors, such as imatinib and erlotinib, as therapeutic agents for several human cancers has prompted substantial interest in the further development and clinical testing of such inhibitors for a wide variety of malignancies. While much of this effort has been focused on the receptor tyrosine kinases, including EGFR, HER2, PDGF receptor, c-KIT, and MET, inhibitors of serine/threonine kinases are also beginning to emerge within discovery pipelines. Among these kinases, the RAF and MEK kinases have received substantial attention, owing largely to the relatively high frequency of activating mutations of RAS (∼20% of all human cancers), an upstream activator of the well established RAF–MEK–ERK signaling cascade, as well as frequent activating mutations in the BRAF kinase (∼7% of all human cancers). Here, we summarize the current state of development of kinase inhibitors directed at this signaling pathway, a few of which have already demonstrating favorable toxicity profiles as well as promising activity in early phase clinical studies.

Introduction

Mitogen-activated protein kinases (MAPKs) mediate intracellular signals transduced by a wide variety of cell surface receptors. Mammalian cells employ four well-characterized MAPK cascades and numerous associated proteins that constitute a complex signaling network. The basic functional “architecture” of each MAPK cascade comprises a MAPK kinase kinase (MAP3K), a MAPK kinase (MAP2K) and a MAPK. The terminal MAPKs are the Erk, JNK (also called SAPK), p38 and ERK5 kinases. Although MAPKs cascades are usually simplified as linear unidirectional pathways, it is important to note the greater complexity of such networks in the context of signal cross-talk with a large number of additional associated proteins [1], [2]. Therefore, further efforts to model the MAPK pathway are of great value in understanding how the pathway potentially responds and adapts when challenged with small molecule inhibitors.

Among the MAPK-mediated pathways, the RAF–MAPK/extracellular signal-related kinase (ERK) kinase (MEK)–ERK signaling cascade has been the most extensively characterized thus far. This largely reflects its established role in a variety of human cancers, particularly those associated with activation of RAS proteins, which bind to and activate the RAF kinase, triggering engagement of this pathway. Upon RAS activation, RAF is recruited to the cell membrane where subsequent changes in RAF phosphorylation status result in stimulation of its serine-threonine kinase activity [3], [4]. Activated RAF triggers sequential phosphorylation and activation of the MEK1/MEK2 dual-specificity protein kinases and ERK, which translocates to the nucleus where they regulate the activity of several transcription factors that induce the expression of multiple genes required for survival and proliferation [5], [6].

Since the first reports of the oncogenic transforming activity of the viral oncogenic form of c-RAF-1 more than twenty years ago, there has been accumulating evidence of the importance of RAF–MEK–ERK pathway in oncogenesis [7]. Aberrant activation of the pathway is frequently seen in human cancers (Fig. 1), and is clearly contributing to oncogenic properties such as independence from growth factors and enhanced proliferation and survival [8], [9]. Constitutive activation of the RAF–MEK–ERK pathway can be driven either by ligand-induced stimulation of membrane associated receptor tyrosine kinases or by ligand-independent mechanisms such as increased expression or mutational activation of proteins that constitute the pathway. The most frequently observed mutations are those that arise in one of the three human RAS genes, HRAS, NRAS, and KRAS, which occur in approximately 20% of human neoplasms [3]. Moreover, recent sequencing efforts have led to the identification of activating mutations in the BRAF gene in 7% of all cancers (discussed below) [10]. Interestingly, NRAS and BRAF mutations appear to be mutually exclusive, suggesting that both proteins function in the same signaling pathway and that activation of either one is sufficient to promote engagement of the MEK–ERK cascade [11], [12].

The fact that the RAF–MEK–ERK cascade plays a central role in the regulation of cell proliferation and survival, together with the high frequency with which this pathway is deregulated in human cancer makes it an attractive target for drug development. The therapeutic approach of selectively targeting activated kinases has proven to be of great value for the treatment of a variety of malignancies, with the best examples thus far being the success of the ABL, KIT, PDGFR inhibitor imatinib in chronic myelogenous leukemias and gastrointestinal stromal tumors (GIST), and the EGFR kinase inhibitor erlotinib in a subset of non-small cell lung cancers [13], [14].

It is difficult to predict which of the various RAS–RAF–MEK–ERK pathway components will constitute the most useful therapeutic target. Moreover, considering the well documented widespread function of each of these proteins in a variety of normal physiological processes, it would not be surprising to find that inhibition of these proteins leads to unacceptable toxicity. Therefore, it is hoped that, as has been seen in other kinase-driven cancers, the mutational activation of this pathway in tumors leads to a therapeutic window that reflects enhanced dependency on tumor cells relative to normal cells [14]. While substantial efforts made over the years to target the activated RAS protein have been unsuccessful, leading many to conclude that RAS is a pharmacologically intractable target, the downstream kinases in the RAS cascade remain attractive as therapeutic targets [15]. Among these, BRAF has emerged as a particularly appealing target due to its high mutation prevalence in tumors. Consequently, intensive preclinical and clinical research has led to the development of small molecule kinase inhibitors of RAF, as well as its downstream effector MEK (Fig. 2). While clinical evaluation of these inhibitors is still in its early days, some promising findings have begun to emerge. In the following sections, we will focus on the various inhibitors of the RAF–MEK–ERK pathway that are currently being tested.

Section snippets

RAF kinase inhibitors

The RAF serine/threonine family is comprised of three isoforms: ARAF, BRAF and CRAF (also referred as c-RAF-1). While these three RAF isoforms share three common conserved regions, they are expressed in a tissue- and cell-specific manner and have been associated with distinct normal biological functions [16]. Recent data show that BRAF exhibits higher basal kinase activity than ARAF and CRAF which require a greater number of phosphorylations within the N-terminal region of the kinase domain to

MEK kinase inhibitors

The two MEK homologues – MEK1 and MEK2- are dual specificity kinases that share the two consensus kinase motifs, one involved in phosphorylation of serine-threonine residues and another in the phosphorylation of tyrosine residues [55]. The only known catalytic substrates for both MEK isoforms are the MAP kinases ERK1 and ERK2 [56]. Such signaling selectivity confers upon MEK a central role in transducing MAPK-mediated proliferation signals and makes it a particularly attractive target for

Future directions

Although clear progress has been made in the development of drugs that target RAF and its downstream effector MEK, new challenges and drug-related issues have emerged from the analysis thus far. Whereas RAF inhibitors are still in a relatively early phase of their clinical development, lessons learned from the evaluation from MEK inhibitors should serve to guide us in the future rational and successful clinical development of RAF/MEK inhibitors. As with other targeted therapies, a current

Conflicts of interest statement

The authors declare that they have no financial interest that could bias their presentation of findings in this manuscript.

Acknowledgements

We apologize to the authors of many excellent and relevant publications that we were unable to include in this review due to limitations of space.

C.M. has been supported by RTICC 06/0020/0109 and Fundació Cellex (Barcelona).

References (83)

  • L. Chang et al.

    Mammalian MAP kinase signalling cascades

    Nature

    (2001)
  • ...
  • M. Malumbres et al.

    RAS oncogenes: the first 30 years

    Nat. Rev. Cancer

    (2003)
  • J. Avruch et al.

    Ras activation of the Raf kinase: tyrosine kinase recruitment of the MAP kinase cascade

    Recent Prog. Horm. Res.

    (2001)
  • S. Yoon et al.

    The extracellular signal-regulated kinase: multiple substrates regulate diverse cellular functions

    Growth Factors

    (2006)
  • U.R. Rapp et al.

    Structure and biological activity of v-raf, a unique oncogene transduced by a retrovirus

    Proc. Natl. Acad. Sci. USA

    (1983)
  • H. Davies et al.

    Mutations of the BRAF gene in human cancer

    Nature

    (2002)
  • A. Gorden et al.

    Analysis of BRAF and N-RAS mutations in metastatic melanoma tissues

    Cancer Res.

    (2003)
  • M.S. Brose et al.

    BRAF and RAS mutations in human lung cancer and melanoma

    Cancer Res.

    (2002)
  • J. Baselga

    Targeting tyrosine kinases in cancer: the second wave

    Science

    (2006)
  • S.V. Sharma et al.

    Oncogene addiction: setting the stage for molecularly targeted cancer therapy

    Genes Dev.

    (2007)
  • J.S. Sebolt-Leopold

    Advances in the development of cancer therapeutics directed against the RAS-mitogen-activated protein kinase pathway

    Clin. Cancer Res.

    (2008)
  • C.S. Mason et al.

    Serine and tyrosine phosphorylations cooperate in Raf-1, but not B-Raf activation

    Embo. J.

    (1999)
  • N. Li et al.

    B-Raf kinase inhibitors for cancer treatment

    Curr. Opin. Invest. Drugs

    (2007)
  • M. Karasarides et al.

    B-RAF is a therapeutic target in melanoma

    Oncogene

    (2004)
  • V. Emuss et al.

    Mutations of C-RAF are rare in human cancer because C-RAF has a low basal kinase activity compared with B-RAF

    Cancer Res.

    (2005)
  • K. Fransen et al.

    Mutation analysis of the BRAF, ARAF and RAF-1 genes in human colorectal adenocarcinomas

    Carcinogenesis

    (2004)
  • G. Singer et al.

    Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma

    J. Natl. Cancer Inst.

    (2003)
  • M.N. Nikiforova et al.

    BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas

    J. Clin. Endocrinol. Metab.

    (2003)
  • L. Wang et al.

    BRAF mutations in colon cancer are not likely attributable to defective DNA mismatch repair

    Cancer Res.

    (2003)
  • J.D. Benson et al.

    Validating cancer drug targets

    Nature

    (2006)
  • U. McDermott et al.

    Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling

    Proc. Natl. Acad. Sci. USA

    (2007)
  • J. Tsai et al.

    Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity

    Proc. Natl. Acad. Sci. USA

    (2008)
  • J.F. Lyons et al.

    Discovery of a novel Raf kinase inhibitor

    Endocr. Relat. Cancer

    (2001)
  • S.M. Wilhelm 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)
  • D.B. Mendel 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)
  • J.W. Clark et al.

    Safety and pharmacokinetics of the dual action Raf kinase and vascular endothelial growth factor receptor inhibitor, BAY 43-9006, in patients with advanced, refractory solid tumors

    Clin. Cancer Res.

    (2005)
  • A. Awada et al.

    Phase I safety and pharmacokinetics of BAY 43-9006 administered for 21 days on/7 days off in patients with advanced, refractory solid tumours

    Brit. J. Cancer

    (2005)
  • D. Strumberg et al.

    Phase I clinical and pharmacokinetic study of the novel Raf kinase and vascular endothelial growth factor receptor inhibitor BAY 43-9006 in patients with advanced refractory solid tumors

    J. Clin. Oncol.

    (2005)
  • B. Escudier et al.

    Sorafenib in advanced clear-cell renal-cell carcinoma

    New Engl. J. Med.

    (2007)
  • M.J. Ratain et al.

    Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma

    J. Clin. Oncol.

    (2006)
  • Cited by (0)

    View full text