Elsevier

Seminars in Oncology

Volume 33, Issue 4, August 2006, Pages 392-406
Seminars in Oncology

Role of Raf Kinase in Cancer: Therapeutic Potential of Targeting the Raf/MEK/ERK Signal Transduction Pathway

https://doi.org/10.1053/j.seminoncol.2006.04.002Get rights and content

Improvements in our understanding of the molecular basis of cancer have led to the clinical development of protein kinase inhibitors, which target pivotal molecules involved in intracellular signaling pathways implicated in tumorigenesis and progression. These novel targeted agents have demonstrated activity against a wide range of solid tumors, are generally better tolerated than standard chemotherapeutics, and may revolutionize the management of advanced refractory cancer. The ubiquitous Raf serine/threonine kinases are pivotal molecules within the Raf/mitogen extracellular kinase (MEK)/extracellular signal-related kinase (ERK) signaling pathway, which regulates cellular proliferation and survival. Raf kinase isoforms (wild-type Raf-1 or the b-raf V600E oncogene) are overactivated in a variety of solid tumor types, including renal cell carcinoma (RCC), hepatocellular carcinoma (HCC), non-small cell lung cancer (NSCLC), melanoma, and papillary thyroid carcinoma. In this review, the role of Raf in normal cells and in cancer is discussed, and an overview is given of Raf inhibitors currently in development, focusing on sorafenib tosylate (BAY 43-9006 or sorafenib). Sorafenib is the first oral multi-kinase inhibitor to be developed that targets Raf kinases (Raf-1, wild-type B-Raf, and b-raf V600E), in addition to receptor tyrosine kinases associated with angiogenesis (vascular endothelial growth factor receptor [VEGFR]-2/-3, platelet-derived growth factor receptor [PDGFR]-β) or tumor progression (Flt-3, c-kit). Preclinical and clinical sorafenib data that led to its recent approval for the treatment of advanced RCC are summarized, along with current thinking on sorafenib’s mechanism of effect on the tumor and tumor vasculature in melanoma and RCC.

Section snippets

Raf Kinase Isoforms: Gatekeepers to the Raf/MEK/ERK Signaling Pathway

The ubiquitous Raf/MEK/ERK pathway is activated by the binding of growth factors, such as transforming growth factor-alpha (TGF-α), epidermal growth factor (EGF), VEGF, and PDGF-β to their cognate receptors (eg, EGFR, VEGFR-2/-3, and PDGFR-β).19, 20, 21, 22, 23 This results in autophosphorylation by RTKs and activation of a kinase cascade that relays growth factor signals from the cell surface to the nucleus.19, 20, 21, 22, 23 As pivotal members of the Raf/MEK/ERK signaling pathway, the Raf

Oncogenic raf-1 Mutations and Malignant Transformation In Vitro

Activated oncogenic forms of raf-1 (eg, v-raf), mutant Raf proteins, and Raf-1 fusion proteins all induce transformation of cell lines in vitro. Raf-1 has been shown to mediate transformation via the MEK/ERK mitogenic pathway, and also by a MEK/ERK-independent route.44 Despite the ease with which oncogenic raf-1 mutants transform cells in vitro, tumor-specific oncogenic raf-1 mutations have been observed only rarely in human tumor-derived cell lines.45, 46 Genetic rearrangements at the Raf-1

Raf Isoforms and Tumor Progression

Dysregulated overexpression of Raf isoforms is also associated with tumor progression. Raf isoforms are involved as downstream effectors of angiogenesis, which is essential for the establishment of the necessary vasculature to facilitate solid tumor growth beyond 2 to 3 mm3. Furthermore, Raf overexpression has been associated with a more aggressive and invasive tumor phenotype and has, therefore, been implicated in promoting tumor metastasis.

Raf: Role in Radio- and Chemoresistance

An essential strategy for any novel targeted anticancer agent involves the abrogation of tumor cell resistance to treatment-induced growth inhibition and cell death. Such resistance has hindered the clinical effectiveness of standard radiotherapy and cytotoxic chemotherapies. However, resistance has also been observed with targeted agents like imatinib, which act by blocking a single target within the tumor (bcr-abl in chronic myelogenous leukemia, c-kit in gastrointestinal stromal tumors

Raf as a Target for Anticancer Agents

The concept of Raf-targeted disruption of cell signaling for therapeutic purposes was first advanced in 1989 with the demonstration of tumor growth inhibition in athymic mice.116 Several agents have been designed that target Raf and are in various stages of clinical development.

Targeting Melanoma With Sorafenib

The high incidence of activating B-Raf mutations in melanoma makes sorafenib an attractive agent for the treatment of this disease, especially because preclinical data suggest that B-Raf inhibition can affect melanoma growth and survival. However, in a small phase II trial of sorafenib monotherapy, in which the drug was given to patients with metastatic melanoma at a dose of 400 mg orally twice per day in 12-week cycles, there was only one PR and three cases of SD among 20 patients.130 Although

Molecular Basis of RCC: Rationale for Targeting Raf With Sorafenib

Loss of function of the VHL tumor-suppressor gene, which resides on the short arm of chromosome 3, and consequent HIF-α upregulation have been observed in most cases of clear-cell RCC.133 The normal function of the VHL gene product (pVHL) is to downregulate expression of the HIF-α transcription factors under normoxic conditions.133, 134, 135, 136, 137, 138 This downregulation is achieved by pVHL forming a multi-protein complex that ubiquitinates the α-subunit of HIF, thereby targeting it for

Summary

Isoforms of the Raf kinase function both as gatekeepers to the ubiquitous Raf/MEK/ERK signaling pathway, and as direct mediators of MEK/ERK-independent cellular proliferation and survival. Dysregulated signaling through Raf-1 (NSCLC, RCC, and HCC) or through oncogenic b-raf V600E (malignant cutaneous melanoma and PTC) has been implicated in the pathogenesis and malignant phenotype of several human solid tumors that are notoriously resistant to conventional chemotherapy and radiation. Therefore,

References (151)

  • S. Jin et al.

    p21-activated kinase 1 (Pak1)-dependent phosphorylation of Raf-1 regulates its mitochondrial localization, phosphorylation of BAD, and Bcl-2 association

    J Biol Chem

    (2005)
  • G. Daum et al.

    The ins and outs of Raf kinases

    Trends Biochem Sci

    (1994)
  • R.T. Belly et al.

    Detection of mutated K12-ras in histologically negative lymph nodes as an indicator of poor prognosis in stage II colorectal cancer

    Clin Colorectal Cancer

    (2001)
  • L.A. Akslen et al.

    BRAF and NRAS mutations are frequent in nodular melanoma but are not associated with tumor cell proliferation or patient survival

    J Invest Dermatol

    (2005)
  • C. Alexia et al.

    An evaluation of the role of insulin-like growth factors (IGF) and of type-I IGF receptor signalling in hepatocarcinogenesis and in the resistance of hepatocarcinoma cells against drug-induced apoptosis

    Biochem Pharmacol

    (2004)
  • Y.H. Hwang et al.

    Over-expression of c-raf-1 proto-oncogene in liver cirrhosis and hepatocellular carcinoma

    Hepatol Res

    (2004)
  • G.G. McGill et al.

    Bcl2 regulation by the melanocyte master regulator Mitf modulates lineage survival and melanoma cell viability

    Cell

    (2002)
  • J. Du et al.

    Critical role of CDK2 for melanoma growth linked to its melanocyte-specific transcriptional regulation by MITF

    Cancer Cell

    (2004)
  • P. Baluk et al.

    Cellular abnormalities of blood vessels as targets in cancer

    Curr Opin Genet Dev

    (2005)
  • N. Ferrara

    Role of vascular endothelial growth factor in physiologic and pathologic angiogenesistherapeutic implications

    Semin Oncol

    (2002)
  • M.B. Sporn

    The war on cancer

    Lancet

    (1996)
  • A. Jemal et al.

    Cancer statistics, 2005

    CA Cancer J Clin

    (2005)
  • E.S. Kim et al.

    Prevention of lung cancer. The new millennium

    Chest Surg Clin North Am

    (2000)
  • P. Tai et al.

    Survival of patients with metastatic breast cancerTwenty-year data from two SEER registries

    BMC Cancer

    (2004)
  • S. Negrier et al.

    Long-term follow-up of patients with metastatic renal cell carcinoma treated with intravenous recombinant interleukin-2 in Europe

    Cancer J Sci Am

    (2000)
  • V. Bordin et al.

    Five-year survival results of subcutaneous low-dose immunotherapy with interleukin-2 alone in metastatic renal cell cancer patients

    Urol Int

    (2000)
  • H.G. Welch et al.

    Are increasing 5-year survival rates evidence of success against cancer?

    JAMA

    (2000)
  • J. Dancey et al.

    Issues and progress with protein kinase inhibitors for cancer treatment

    Nat Rev Drug Discov

    (2003)
  • W. Kolch et al.

    The role of Raf kinases in malignant transformation

    Expert Rev Mol Med

    (2002)
  • S. Chan

    Targeting the mammalian target of rapamycin (mTOR)A new approach to treating cancer

    Br J Cancer

    (2004)
  • K. Peggs

    Imatinib mesylate—Gold standards and silver linings

    Clin Exp Med

    (2004)
  • W. Kolch

    Meaningful relationshipsThe regulation of the Ras/Raf/MEK/ERK pathway by protein interactions

    Biochem J

    (2000)
  • S.M. Wilhelm et al.

    BAY 43-9006 exhibits broad spectrum oral anti-tumor activity and targets the Raf/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis

    Cancer Res

    (2004)
  • C. Yu et al.

    The role of Mcl-1 down-regulation in the pro-apoptotic activity of the multi-kinase inhibitor BAY 43-9006

    Oncogene

    (2005)
  • Y.S. Chang et al.

    BAY 43-9006 (Sorafenib) inhibits ectopic and orthotopic growth of a murine model of renal adenocarcinoma (Renca) predominantly through inhibition of tumor angiogenesis

    Clin Cancer Res

    (2005)
  • V.C. Gray-Schopfer et al.

    The role of B-RAF in melanoma

    Cancer Metastasis Rev

    (2005)
  • M.L. Edin et al.

    Raf-1 serine 338 phosphorylation plays a key role in adhesion-dependent activation of extracellular signal-regulated kinase by epidermal growth factor

    Mol Cell Biol

    (2005)
  • K. Ehrenreiter et al.

    Raf-1 regulates Rho signaling and cell migration

    J Cell Biol

    (2005)
  • K.E. Hamden et al.

    Raf and VEGFEmerging therapeutic targets in Kaposi’s sarcoma-associated herpesvirus infection and angiogenesis in hematopoietic and nonhematopoietic tumors

    Leukemia

    (2005)
  • A. Schulze et al.

    Analysis of the transcriptional program induced by Raf in epithelial cells

    Genes Dev

    (2001)
  • C.S. Mason et al.

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

    EMBO J

    (1999)
  • H. Chong et al.

    Positive and negative regulation of Raf kinase activity and function by phosphorylation

    EMBO J

    (2001)
  • U. Kasid et al.

    Activation of Raf by ionizing radiation

    Nature

    (1996)
  • M. Mikula et al.

    Embryonic lethality and fetal liver apoptosis in mice lacking the c-raf-1 gene

    EMBO J

    (2001)
  • M. Huser et al.

    MEK kinase activity is not necessary for Raf-1 function

    EMBO J

    (2001)
  • P. Bouillet et al.

    BH3-only proteins—Evolutionarily conserved proapoptotic Bcl-2 family members essential for initiating programmed cell death

    J Cell Sci

    (2002)
  • V. Le Mellay et al.

    Negative regulation of mitochondrial VDAC channels by C-Raf kinase

    BMC Cell Biol

    (2002)
  • J. Chen et al.

    Raf-1 promotes cell survival by antagonizing apoptosis signal-regulating kinase 1 through a MEK-ERK independent mechanism

    Proc Natl Acad Sci U S A

    (2001)
  • E.H. Cheng et al.

    VDAC2 inhibits BAK activation and mitochondrial apoptosis

    Science

    (2003)
  • J.H. Yoon et al.

    Bile acids inhibit Mcl-1 protein turnover via an epidermal growth factor receptor/Raf-1-dependent mechanism

    Cancer Res

    (2002)
  • Cited by (321)

    • Metabolic pathway-based target therapy to hepatocellular carcinoma: a computational approach

      2022, Theranostics and Precision Medicine for the Management of Hepatocellular Carcinoma, Volume 2: Diagnosis, Therapeutic Targets, and Molecular Mechanisms
    • Signaling pathways in hepatocellular carcinoma

      2021, Advances in Cancer Research
    View all citing articles on Scopus
    View full text