Elsevier

European Journal of Cancer

Volume 38, Issue 13, September 2002, Pages 1685-1700
European Journal of Cancer

Review
Farnesyl transferase inhibitors as anticancer agents

https://doi.org/10.1016/S0959-8049(02)00166-1Get rights and content

Abstract

Protein farnesylation catalysed by the enzyme farnesyl protein transferase involves the addition of a 15-carbon farnesyl group to conserved amino acid residues at the carboxyl terminus of certain proteins. Protein substrates of farnesyl transferase include several G-proteins, which are critical intermediates of cell signalling and cytoskeletal organisation such as Ras, Rho, PxF and lamins A and B. Activated Ras proteins trigger a cascade of phosphorylation events through sequential activation of the PI3 kinase/AKT pathway, which is critical for cell survival, and the Raf/Mek/Erk kinase pathway that has been implicated in cell proliferation. Ras mutations which encode for constitutively activated proteins are found in 30% of human cancers. Because farnesylation of Ras is required for its transforming and proliferative activity, the farnesyl protein transferase inhibitors were designed as anticancer agents to abrogate Ras function. However, current evidence suggests that the anticancer activity of the farnesyl transferase inhibitors may not be simply due to Ras inhibition. This review will discuss available clinical data on three of these agents that are currently undergoing clinical trials.

Introduction

Advances in molecular biology over the past decade have identified a number of novel targets for cancer therapy. One such target is the enzyme farnesyl protein transferase (FT), which catalyses a key step in the addition of an aliphatic isoprenoid side chain to a number of proteins. A novel class of antineoplastic agents, the farnesyl transferase inhibitors (FTIs), have recently been developed to specifically inhibit FT. These inhibitors were designed to target Ras, a G-protein with 4 isoforms (H-, N- and K-RasA/K-RasB) mutated in a large number of cancers, which requires prenylation for function [1]. However, it has become clear in many instances that Ras may not be the critical target of FTIs. For instance, in human tumour cell lines, the antitumour activity of FTIs does not correlate with a mutated Ras status 2, 3. The findings that K- and N-Ras can be alternatively prenylated by geranylgeranyl protein transferase (GGT) also argues against Ras as the target, as preclinical models bearing these mutations are sensitive to FTI treatment 4, 5, 6, 7. To date, more than a hundred polypeptides possessing a ‘CAAX’ (tetrapeptide motif, where C is cysteine, A is any alphatic amino acid and X is serine, leucine, glutamine or methironine) sequence that can potentially be farnesylated have been identified [8]. Theoretically, the inhibition of farnesylation of any of these polypeptides could result in the antiproliferative effects of the FTIs in human tumours.

Despite uncertainty about the true target of FTIs, these agents demonstrate anticancer activity as single agents and in combination with standard cytotoxic chemotherapy 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18. In addition, FTIs synergise with gamma irradiation, and may have a role in chemoprevention 19, 20, 21. This review will focus on the FTIs that are currently undergoing clinical investigations. Published results of these clinical trials will be discussed. Recent investigations into the targets of FTIs will also be discussed.

Section snippets

Farnesyl protein transferase

FT is a heterodimeric metalloenzyme whose activity is dependent on magnesium and zinc, the latter of which is required for coordination of the farnesyl-accepting cysteine of the target protein and the active site of the enzyme [22]. FT catalyses the addition of a 15-carbon isoprenyl farnesyl moiety, to the carboxy terminus of proteins containing the peptide target motif ‘CAAX’. Similarly, the related protein geranylgeranyl transferase type I (GGT-1) catalyses the addition of a 20-carbon

Protein farnesylation

The addition of a farnesyl group to conserved amino acid residues at the carboxy terminus is necessary for the proper functioning of many farnesylated proteins. The farnesylation reaction is part of a series of modifications necessary for plasma membrane association of various proteins (Fig. 1). While it is generally believed that prenylation confers a hydrophobic core necessary to anchor proteins to cell membranes, more investigation is needed to understand the entire spectrum of function that

Mechanism of FTI cytotoxicity

More than 70% of cancer cell lines have shown sensitivity to FTI treatment. FTIs inhibit the anchorage-independent growth of this large variety of transformed cell lines 2, 39, 40, 41. In addition, while FTIs clearly abrogate FT activity both in cultured cell lines and in surrogate tissue 9, 15 or tumour cells 9, 15 from patients, it has become increasingly clear that FTIs do not target only Ras, but may have other protein targets as well. As mentioned earlier in the introduction, there is no

Preclinical/clinical studies

Three FTIs (Fig. 3) are undergoing clinical testing currently as monotherapy, and in combination with standard cytotoxic agents (Table 2). An additional agent, L-778,123, was introduced into the clinic, but its further development has been halted due to toxicity. At least three other compounds are in late preclinical testing and may be introduced into clinical trials shortly.

FTIs in combination with ionising radiation

Ras oncogenes have been reported to confer resistance to ionising radiation 93, 94. Since the FTIs were presumed to be inhibiting ras proteins, their role as radiosensitising agents, have been evaluated. In spite of the uncertainty surrounding their ras inhibitory effects, the FTIs have been shown to synergise with gamma radiation in preclinical models.

Because activated H-ras expression had been shown to markedly increase radiation resistance in some transformed cells and FTI-277 was shown to

Conclusions

FTIs are a promising class of novel antineoplastic agents. Although initially designed as anti-Ras agents, mounting evidence indicates that other farnesylated targets are involved in the cytotoxic effects of these agents. Identification of the critical protein target(s) of FTIs is currently an active area of research 2, 38, 60, 61, 62, 74, 76, 100, 101. The FTIs are generally well tolerated at clinically effective doses [102]. Some of their toxicities, such as nausea, vomiting and diarrhoea,

Acknowledgements

A.A.A. is a recipient of a Research Scholar Grant (RSG-01–155–01-CCE) from the American Cancer Society. The authors wish to thank Mrs Gail Prechel for expert secretarial assistance.

References (129)

  • A.D. Cox et al.

    The CAAX peptidomimetic compound B581 specifically blocks farnesylated, but not geranylgeranylated or myristylated, oncogenic ras signaling and transformation

    J. Biol. Chem.

    (1994)
  • S.A. Armstrong et al.

    CAAX geranylgeranyl transferase transfers farnesyl as efficiently as geranylgeranyl to RhoB

    J. Biol. Chem.

    (1995)
  • A. Vogt et al.

    The geranylgeranyltransferase-I inhibitor GGTI-298 arrests human tumor cells in G0/G1 and induces p21(WAF1/CIP1/SDI1) in a p53-independent manner

    J. Biol. Chem.

    (1997)
  • B.K. Law et al.

    Farnesyltransferase inhibitor induces rapid growth arrest and blocks p70s6k activation by multiple stimuli

    J. Biol. Chem.

    (2000)
  • J. Zha et al.

    BH3 domain of BAD is required for heterodimerization with BCL-XL and pro-apoptotic activity

    J. Biol. Chem.

    (1997)
  • S. Ottilie et al.

    Dimerization properties of human BAD. Identification of a BH-3 domain and analysis of its binding to mutant BCL-2 and BCL-XL proteins

    J. Biol. Chem.

    (1997)
  • H.R. Ashar et al.

    Farnesyl transferase inhibitors block the farnesylation of CENP-E and CENP-F and alter the association of CENP-E with the microtubules

    J. Biol. Chem.

    (2000)
  • N.C. Crespo et al.

    The farnesyltransferase inhibitor, FTI-2153, blocks bipolar spindle formation and chromosome alignment and causes prometaphase accumulation during mitosis of human lung cancer cells

    J. Biol. Chem.

    (2001)
  • H.R. Ashar et al.

    The farnesyl transferase inhibitor SCH 66336 induces a G(2)→M or G(1) pause in sensitive human tumor cell lines

    Exp. Cell Res.

    (2001)
  • W.R. Bishop et al.

    Novel tricyclic inhibitors of farnesyl protein transferase. Biochemical characterization and inhibition of Ras modification in transfected Cos cells

    J. Biol. Chem.

    (1995)
  • A. Reichert et al.

    Treatment of Bcr/Abl-positive acute lymphoblastic leukemia in P190 transgenic mice with the farnesyl transferase inhibitor SCH66336

    Blood

    (2001)
  • K. Kato et al.

    Isoprenoid addition to Ras protein is the critical modification for its membrane association and transforming activity

    Proc. Natl. Acad. Sci. USA

    (1992)
  • L. Sepp-Lorenzino et al.

    A peptidomimetic inhibitor of farnesyl:protein transferase blocks the anchorage-dependent and-independent growth of human tumor cell lines

    Cancer Res.

    (1995)
  • T. Nagasu et al.

    Inhibition of human tumor xenograft growth by treatment with the farnesyl transferase inhibitor B956

    Cancer Res.

    (1995)
  • J. Sun et al.

    Both farnesyltransferase and geranylgeranyltransferase I inhibitors are required for inhibition of oncogenic K-Ras prenylation but each alone is sufficient to suppress human tumor growth in nude mouse xenografts

    Oncogene

    (1998)
  • E.C. Lerner et al.

    Inhibition of the prenylation of K-Ras, but not H- or N-Ras, is highly resistant to CAAX peptidomimetics and requires both a farnesyltransferase and a geranylgeranyltransferase I inhibitor in human tumor cell lines

    Oncogene

    (1997)
  • S.R. Johnston et al.

    A phase II study of the farnesyl transferase inhibitor R115777 in patients with advanced breast cancer

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2000)
  • M.S. Kies et al.

    Induction therapy with SCH 66336, a farnesyltransferase inhibitor, in squamous cell carcinoma (SCC) of the head and neck

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2001)
  • M.J. Piccart-Gebhart et al.

    A phase I, clinical and pharmacokinetic (PK) trial of the farnesyl transferase inhibitor (FTI) R115777+docetaxela promising combination in patients (PTS) with solid tumors

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2001)
  • J.H. Schellens et al.

    Phase I and pharmacologic study with the novel farnesyl transferase inhibitor (FTI) R115777

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2000)
  • J. Lancet et al.

    Use of farnesyl transferase inhibitor R115777 in relapsed and refractory acute leukemias: preliminary results of a phase I trial

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2000)
  • S.N. Holden et al.

    A phase I pharmacokinetic (PK) and biological study of the farnesyl transferase inhibitor (FTI) R115777 and capecitabine in patients (PTS) with advanced solid malignancies

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2001)
  • A.A. Adjei et al.

    A Phase I trial of the farnesyl transferase inhibitor SCH66336evidence for biological and clinical activity

    Cancer Res.

    (2000)
  • H.I. Hurwitz et al.

    Phase I pharmacokinetic trial of the farnesyl transferase inhibitor SCH66336 plus gemcitabine in advanced cancers

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2000)
  • F.R. Khuri et al.

    Phase I study of farnesyl transferase inhibitor (FTI) SCH66336 with paclitaxel in solid tumors: dose finding, pharmacokinetics, efficacy/safety

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2000)
  • E.S. Kim et al.

    A phase I/II study of the farnesyl transferase inhibitor (FTI) SCH66336 with paclitaxel in patients with solid tumors

    Proc. Annu. Meet. Am. Assoc. Cancer Res.

    (2001)
  • E. Cohen-Jonathan et al.

    The farnesyltransferase inhibitor FTI-277 suppresses the 24-kDa FGF2-induced radioresistance in HeLa cells expressing wild-type RAS

    Radiat. Res.

    (1999)
  • E.J. Bernhard et al.

    Inhibiting Ras prenylation increases the radiosensitivity of human tumor cell lines with activating mutations of ras oncogenes

    Cancer Res.

    (1998)
  • E.J. Bernhard et al.

    The farnesyltransferase inhibitor FTI-277 radiosensitizes H-ras- transformed rat embryo fibroblasts

    Cancer Res.

    (1996)
  • P.J. Casey et al.

    Enzymatic modification of proteins with a geranylgeranyl isoprenoid

    Proc. Natl. Acad. Sci. USA

    (1991)
  • P.J. Casey et al.

    p21ras is modified by a farnesyl isoprenoid

    Proc. Natl. Acad. Sci. USA

    (1989)
  • Y. Reiss et al.

    Sequence requirement for peptide recognition by rat brain p21ras protein farnesyltransferase

    Proc. Natl. Acad. Sci. USA

    (1991)
  • M. Sinensky et al.

    The prenylation of proteins

    Bioessays

    (1992)
  • C.L. Strickland et al.

    Crystal structure of farnesyl protein transferase complexed with a CaaX peptide and farnesyl diphosphate analogue

    Biochemistry

    (1998)
  • C.L. Strickland et al.

    Tricyclic farnesyl protein transferase inhibitorscrystallographic and calorimetric studies of structure-activity relationships

    J. Med. Chem.

    (1999)
  • W.A. Maltese

    Posttranslational modification of proteins by isoprenoids in mammalian cells

    Faseb. J.

    (1990)
  • J.F. Hancock et al.

    A CAAX or a CAAL motif and a second signal are sufficient for plasma membrane targeting of ras proteins

    Embo. J.

    (1991)
  • J.E. Buss et al.

    Direct identification of palmitic acid as the lipid attached to p21ras

    Mol. Cell Biol.

    (1986)
  • A.A. Adjei

    Blocking oncogenic Ras signaling for cancer therapy

    J. Natl. Cancer Inst.

    (2001)
  • M. Crul et al.

    Ras biochemistry and farnesyl transferase inhibitorsa literature survey

    Anticancer Drugs

    (2001)
  • Cited by (149)

    View all citing articles on Scopus
    View full text