Elsevier

Methods in Enzymology

Volume 439, 2008, Pages 467-489
Methods in Enzymology

The Ras Inhibitor Farnesylthiosalicylic Acid (Salirasib) Disrupts The Spatiotemporal Localization Of Active Ras: A Potential Treatment For Cancer

https://doi.org/10.1016/S0076-6879(07)00432-6Get rights and content

Abstract

Chronic activation of Ras proteins by mutational activation or by growth factor stimulation is a common occurrence in many human cancers and was shown to induce and be required for tumor growth. Even if additional genetic defects are present, “correction” of the Ras defect has been shown to reverse Ras‐dependent tumorigenesis. One way to block Ras protein activity is by interfering with their spatiotemporal localization in cellular membranes or in membrane microdomains, a prerequisite for Ras signaling and biological activity. Detailed reports describe the use of this method in studies employing farnesylthiosalicylic acid (FTS, Salirasib), a Ras farnesylcysteine mimetic, which selectively disrupts the association of chronically active Ras proteins with the plasma membrane. FTS competes with Ras for binding to Ras‐escort proteins, which possess putative farnesyl‐binding domains and interact only with the activated form of Ras proteins, thereby promoting Ras nanoclusterization in the plasma membrane and robust signals. This chapter presents three‐dimensional time‐lapse images that track the FTS‐induced inhibition of membrane‐activated Ras in live cells on a real‐time scale. It also describes a mechanistic model that explains FTS selectivity toward activated Ras. Selective blocking of activated Ras proteins results in the inhibition of Ras transformation in vitro and in animal models, with no accompanying toxicity. Phase I clinical trials have demonstrated a safe profile for oral FTS, with minimal side effects and promising activity in hematological malignancies. Salirasib is currently undergoing trials in patients with pancreatic cancer and with nonsmall cell lung cancer, with or without identified K‐Ras mutations. The findings might indicate whether with the disruption of the spatiotemporal localization of oncogenic Ras proteins and the targeting of prenyl‐binding domains by anticancer drugs is worth developing as a means of cancer treatment.

Introduction

Mutations in ras genes occur in 30% of all human cancers, with the highest incidence of mutational activation of Ras being detected in pancreatic (90%) and colon (50%) cancers (Baines 2005, Chin 1999, Cox 2002, Downward 2003b, Hahn 1999, Hanahan 2000). K‐ras and N‐ras are the most frequently mutated genes (in most cases displaying a point mutation at codon 12), yet all three of the highly homologous Ras isoforms (H‐Ras, K‐Ras, and N‐Ras) promote malignant transformation (Baines 2005, Chin 1999, Cox 2002, Downward 2003b, Hahn 1999, Hanahan 2000). Oncogenic Ras isoforms (e.g., G12V mutants) are constitutively active (bound to GTP) and are critically required for both initiation and maintenance of the transformed phenotype of cancer cells that harbor the mutated Ras (Baines 2005, Chin 1999, Cox 2002, Hahn 1999, Hanahan 2000). Thus, for example, constitutively active Ras causes growth transformation of primary human cells, but other genetic alterations are required to facilitate the Ras transformation (Hahn et al., 1999). Ras activation induces and is required for tumor growth, and even if many genes are defective, “correction” of the Ras defect alone is sufficient to reverse the process (Chin et al., 1999). Blockage of capan‐1 (a human pancreatic cell line that harbors K‐RasG12V tumor growth in mice) by siRNA for K‐RasG12V provides additional strong support for the notion that blockage of Ras will be of clinical benefit (Baines et al., 2005). The aforementioned experimental findings highlight the importance of Ras proteins as a target for cancer drugs. One way to block the functions of Ras proteins is by interfering with their trafficking to and from cellular membranes, as well as with their proper localizations in different cellular localities and microdomains (Kloog 2000, Philips 2007)—all of which are required for Ras signaling and biological activities (Cox 2002, Dong 2003, Hancock 2005, Kloog 2000, Philips 2007, Silvius 2006). This chapter describes the methods employed to study the effects of farnesylthiosalicylic acid (FTS; Salirasib), a Ras farnesylcysteine mimetic shown to selectively disrupt the association of chronically active Ras proteins with the plasma membrane, on Ras membrane anchorage and function (Elad 1999, Goldberg 2006, Haklai 1998, Yaari 2005).

Section snippets

Preparation of FTS solutions for cell culture experiments

Farnesylthiosalicylic acid is a relatively stable compound (molecular weight 357) that appears as a whitish‐yellowish powder. It can be stored dry at room temperature, but we recommend keeping it in a foil‐covered closed tube at −70° (Marciano et al., 1995). We have found that under such conditions FTS remains stable for at least 3 years. The simplest way to verify that FTS has not been altered during storage is by subjecting it to thin‐layer chromatography (results are obtained within 10–15

Location is critical for Ras signaling and biological activity

Ras protein signaling depends on specific combinations of Ras activation at the plasma membrane, endomembranes, or both (Hancock and Parton, 2005), dynamic lateral segregation of Ras within the plasma membrane (Niv 1999, Niv 2002, Roy 2005), and translocation of Ras from the plasma membrane to intracellular compartments (Hancock 2003, Hancock 2005, Philips 2005). Ras proteins signal from the plasma membrane, Golgi, and perhaps also from mitochondrial membranes (Bivona 2006, Hancock 2003,

Acknowledgments

YK is the incumbent of The Jack H. Skirball Chair for Applied Neurobiology. We thank S.R. Smith for editorial assistance. This work was supported in part by grants to YK from the Israel Science Foundation (Grant 912/06) and the Wolfson Foundation.

References (91)

  • J. Halaschek‐Wiener et al.

    Ras inhibition leads to transcriptional activation of p53 and down‐regulation of Mdm2: Two mechanisms that cooperatively increase p53 function in colon cancer cells

    Cell Signal

    (2004)
  • D. Hanahan et al.

    The hallmarks of cancer

    Cell

    (2000)
  • M.F. Hanzal‐Bayer et al.

    Lipid rafts and membrane traffic

    FEBS Lett.

    (2007)
  • A. Harding et al.

    Subcellular localization determines MAP kinase signal output

    Curr. Biol.

    (2005)
  • J. Jindrich et al.

    Regioselectivity of alkylation of cyclomaltoheptaose (beta‐cyclodextrin) and synthesis of its mono‐2‐O‐methyl, ‐ethyl, ‐allyl, and ‐propyl derivatives

    Carbohydr. Res.

    (1995)
  • N. Jura et al.

    Differential modification of ras proteins by ubiquitination

    Mol. Cell

    (2006)
  • A. Khwaja et al.

    The inhibition of human mesangial cell proliferation by S‐trans, trans‐farnesylthiosalicylic acid

    Kidney Int.

    (2005)
  • Y. Kloog et al.

    RAS inhibitors: Potential for cancer therapeutics

    Mol. Med. Today

    (2000)
  • J. Liao et al.

    K‐ras regulates the steady‐state expression of matrix metalloproteinase 2 in fibroblasts

    J. Biol. Chem.

    (2003)
  • M. Marom et al.

    Selective inhibition of Ras‐dependent cell growth by farnesylthiosalisylic acid

    J. Biol. Chem.

    (1995)
  • H. Niv et al.

    Membrane interactions of a constitutively active GFP‐K‐Ras 4B and their role in signaling: Evidence from lateral mobility studies

    J. Biol. Chem.

    (1999)
  • R.G. Parton et al.

    Lipid rafts and plasma membrane microorganization: Insights from Ras

    Trends Cell Biol.

    (2004)
  • S. Reif et al.

    The Ras antagonist, farnesylthiosalicylic acid (FTS) inhibits experimentally induced liver cirrhosis in rats

    J. Hepatol.

    (1999)
  • M. Serrano et al.

    Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a

    Cell

    (1997)
  • J.M. Shields et al.

    Understanding Ras: ‘it ain’t over ‘til it's over’

    Trends Cell Biol.

    (2000)
  • U. Ashery et al.

    Spatiotemporal organization of ras signaling: Rasosomes and the galectin switch

    Cell. Mol.Neurobiol.

    (2006)
  • U. Ashery et al.

    Nonconventional trafficking of Ras associated with Ras signal organization

    Traffic

    (2006)
  • A.T. Baines et al.

    Use of retrovirus expression of interfering RNA to determine the contribution of activated k‐ras and ras effector expression to human tumor cell growth

    Methods Enzymol.

    (2005)
  • B. Barkan et al.

    The Ras inhibitor farnesylthiosalicylic acid as a potential therapy for neurofibromatosis type 1

    Clin. Cancer Res.

    (2006)
  • A.H. Bild et al.

    Linking oncogenic pathways with therapeutic opportunities

    Nat. Rev. Cancer

    (2006)
  • A.H. Bild et al.

    Oncogenic pathway signatures in human cancers as a guide to targeted therapies

    Nature

    (2006)
  • R. Blum et al.

    Gene expression signature of human cancer cell lines treated with the Ras inhibitor Salirasib (S‐farnesylthiosalicylic acid)

    Cancer Res.

    (2007)
  • R. Blum et al.

    Ras inhibition in glioblastoma down‐regulates hypoxia‐inducible factor‐1alpha, causing glycolysis shutdown and cell death

    Cancer Res.

    (2005)
  • R. Blum et al.

    Suppression of survivin expression in glioblastoma cells by the Ras inhibitor farnesylthiosalicylic acid promotes caspase‐dependent apoptosis

    Mol. Cancer Ther.

    (2006)
  • R. Blum et al.

    E2F1 identified by promoter and biochemical analysis as a central target of glioblastoma cell‐cycle arrest in response to Ras inhibition

    Int. J. Cancer

    (2006)
  • G. Borthakur et al.

    Phase I study of S‐trans, trans‐farnesylthiosalicylic acid (FTS), a noval oral Ras inhibitor, in patients with refractory hematologic malignancies

  • P.J. Casey et al.

    p21ras is modified by a farnesyl isoprenoid

    Proc. Natl. Acad. Sci. USA

    (1989)
  • L. Chin et al.

    Essential role for oncogenic Ras in tumour maintenance

    Nature

    (1999)
  • H.C. Clarke et al.

    Ras antagonist farnesylthiosalicylic acid (FTS) reduces glomerular cellular proliferation and macrophage number in rat thy‐1 nephritis

    J. Am. Soc. Nephrol.

    (2003)
  • A.D. Cox et al.

    Ras family signaling: Therapeutic targeting

    Cancer Biol. Ther.

    (2002)
  • X. Dong et al.

    Palmitoylation and plasma membrane localization of Ras2p by a nonclassical trafficking pathway in

    Saccharomyces cerevisiae. Mol. Cell Biol.

    (2003)
  • J. Downward

    Role of receptor tyrosine kinases in G‐protein‐coupled receptor regulation of Ras: Transactivation or parallel pathways?

    Biochem. J.

    (2003)
  • J. Downward

    Targeting RAS signalling pathways in cancer therapy

    Nat. Rev. Cancer

    (2003)
  • Y. Egozi et al.

    Growth inhibition of ras‐dependent tumors in nude mice by a potent ras‐dislodging antagonist

    Int. J. Cancer

    (1999)
  • M. Fivaz et al.

    Reversible intracellular translocation of KRas but not HRas in hippocampal neurons regulated by Ca2+/calmodulin

    J. Cell Biol.

    (2005)
  • Cited by (93)

    • Aquaporin water channels affect the response of conventional anticancer therapies of 3D grown breast cancer cells

      2023, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      As a model system, we used AQP overexpressing breast cancer cells grown as 3D spheroids, which recapitulate the tumor microenvironment and structural organization with cellular layered assembling, which affects hypoxia and nutrient gradients [36]. We used the conventional breast cancer anticancer chemotherapies Cisplatin, 5-FU and Doxorubicin, either alone or as Combination, and the Combination with the Ras inhibitor Salirasib [37]; Salirasib was added to test the effect of AQP5 mediated Ras signaling. AQP1-EGFP, AQP3-EGFP and AQP5-EGFP plasmids were from Prof. Peter Pohl (Johannes Kepler University, Austria (AQP1-EGFP)) and Prof. Anita Aperia (Karolinska Institutet, Sweden (AQP3-EGFP and AQP5-EGFP)), respectively and were previously characterized [23,38].

    • The interplay between Ras, Autophagy and cancer

      2021, Advances in Cancer Biology - Metastasis
    • Small molecule tyrosine kinase inhibitors and pancreatic cancer—Trials and troubles

      2019, Seminars in Cancer Biology
      Citation Excerpt :

      Salirasib is a Ras farnesylcysteine mimic that interrupts the association of the active forms of Ras (H-Ras, K-Ras and N-Ras) with the cellular membrane [186]. The mechanisms of action of salirasib have been elucidated and are highly specific [187]. For instance, the nanoclustering of the active H-Ras-GTP in the plasma membrane is modulated by the escort molecule galectin-1, which attaches to this Ras isoform in a farnesyl-dependent fashion.

    View all citing articles on Scopus
    View full text