Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Oncogene ; 26(5): 633-40, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-16909123

ABSTRACT

Recently, we have shown that RhoB suppresses EGFR-, ErbB2-, Ras- and Akt-mediated malignant transformation and metastasis. In this paper, we demonstrate that the novel antitumor agents farnesyltransferase inhibitors (FTIs) and geranylgeranyltransferase I inhibitors (GGTIs) upregulate RhoB expression in a wide spectrum of human cancer cells including those from pancreatic, breast, lung, colon, bladder and brain cancers. RhoB induction by FTI-277 and GGTI-298 occurs at the transcriptional level and is blocked by actinomycin D. Reverse transcription-PCR experiments documented that the increase in RhoB protein levels is due to an increase in RhoB transcription. Furthermore, treatment with FTIs and GGTIs of cancer cells results in HDAC1 dissociation, HAT association and histone acetylation of the RhoB promoter. Thus, promoter acetylation is a novel mechanism by which RhoB expression levels are regulated following treatment with the anticancer agents FTIs and GGTIs.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Farnesyltranstransferase/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Promoter Regions, Genetic , rhoB GTP-Binding Protein/genetics , Acetylation , Alkyl and Aryl Transferases/metabolism , Antineoplastic Agents , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/metabolism , Histone Deacetylase 1 , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Up-Regulation , rhoB GTP-Binding Protein/metabolism
3.
Mol Cell Biol ; 18(12): 6962-70, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9819384

ABSTRACT

We have recently reported that the geranylgeranyltransferase I inhibitor GGTI-298 arrests human tumor cells at the G1 phase of the cell cycle and increases the protein and RNA levels of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1). Here, we show that GGTI-298 acts at the transcriptional level to induce p21(WAF1/CIP1) in a human pancreatic carcinoma cell line, Panc-1. This upregulation of p21(WAF1/CIP1) promoter was selective, since GGTI-298 inhibited serum responsive element- and E2F-mediated transcription. A functional analysis of the p21(WAF1/CIP1) promoter showed that a GC-rich region located between positions -83 and -74, which contains a transforming growth factor beta-responsive element and one Sp1-binding site, is sufficient for the upregulation of p21(WAF1/CIP1) promoter by GGTI-298. Electrophoretic mobility shift assays showed a small increase in the amount of DNA-bound Sp1-Sp3 complexes. Furthermore, the analysis of Sp1 transcriptional activity in GGTI-298-treated cells by using GAL4-Sp1 chimera or Sp1-chloramphenicol acetyltransferase reporter revealed a significant increase in Sp1-mediated transcription. Moreover, GGTI-298 treatment also resulted in increased Sp1 and Sp3 phosphorylation. These results suggest that GGTI-298-mediated upregulation of p21(WAF1/CIP1) involves both an increase in the amount of DNA-bound Sp1-Sp3 and enhancement of Sp1 transcriptional activity. To identify the geranylgeranylated protein(s) involved in p21(WAF1/CIP1) transcriptional activation, we analyzed the effects of the small GTPases Rac1 and RhoA on p21(WAF1/CIP1) promoter activity. The dominant negative mutant of RhoA, but not Rac1, was able to activate p21(WAF1/CIP1). In contrast, constitutively active RhoA repressed p21(WAF1/CIP1). Accordingly, the ADP-ribosyl transferase C3, which specifically inhibits Rho proteins, enhanced the activity of p21(WAF1/CIP1). Taken together, these results suggest that one mechanism by which GGTI-298 upregulates p21(WAF1/CIP1) transcription is by preventing the small GTPase RhoA from repressing p21(WAF1/CIP1) induction.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Benzamides/pharmacology , Cyclins/genetics , GTP-Binding Proteins/genetics , Genes, Regulator/genetics , Sp1 Transcription Factor/genetics , Transforming Growth Factor beta/genetics , Up-Regulation/genetics , Binding Sites/genetics , Cyclin-Dependent Kinase Inhibitor p21 , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , G1 Phase/genetics , Humans , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Phosphorylation , Promoter Regions, Genetic/genetics , Sp3 Transcription Factor , Transcription Factors/genetics , Transfection/genetics , Tumor Cells, Cultured , rhoA GTP-Binding Protein
4.
Mol Cell Biol ; 20(1): 139-48, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10594016

ABSTRACT

Farnesyltransferase inhibitors (FTIs) represent a novel class of anticancer drugs that exhibit a remarkable ability to inhibit malignant transformation without toxicity to normal cells. However, the mechanism by which FTIs inhibit tumor growth is not well understood. Here, we demonstrate that FTI-277 inhibits phosphatidylinositol 3-OH kinase (PI 3-kinase)/AKT2-mediated growth factor- and adhesion-dependent survival pathways and induces apoptosis in human cancer cells that overexpress AKT2. Furthermore, overexpression of AKT2, but not oncogenic H-Ras, sensitizes NIH 3T3 cells to FTI-277, and a high serum level prevents FTI-277-induced apoptosis in H-Ras- but not AKT2-transformed NIH 3T3 cells. A constitutively active form of AKT2 rescues human cancer cells from FTI-277-induced apoptosis. FTI-277 inhibits insulin-like growth factor 1-induced PI 3-kinase and AKT2 activation and subsequent phosphorylation of the proapoptotic protein BAD. Integrin-dependent activation of AKT2 is also blocked by FTI-277. Thus, a mechanism for FTI inhibition of human tumor growth is by inducing apoptosis through inhibition of PI 3-kinase/AKT2-mediated cell survival and adhesion pathway.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Apoptosis , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Signal Transduction , Apoptosis/drug effects , Cell Line , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Female , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects
5.
Nat Biotechnol ; 18(10): 1065-70, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017044

ABSTRACT

We have designed a molecule, GFB-111, that binds to platelet-derived growth factor (PDGF), prevents it from binding to its receptor tyrosine kinase, and blocks PDGF-induced receptor autophosphorylation, activation of Erk1 and Erk2 kinases, and DNA synthesis. GFB-111 is highly potent (IC50 = 250 nM) and selective for PDGF over EGF, IGF-1, aFGF, bFGF, and HRGbeta (IC50 values > 100 microM), but inhibits VEGF-induced Flk-1 tyrosine phosphorylation and Erk1/Erk2 activation with an IC50 of 10 microM. GFB-111 treatment of nude mice bearing human tumors resulted in significant inhibition of tumor growth and angiogenesis. The results demonstrate the feasibility of designing novel growth factor-binding molecules with potent anticancer and antiangiogenic activity.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Design , Glioblastoma/drug therapy , Peptides, Cyclic/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line , DNA/biosynthesis , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/pharmacology , Enzyme Activation/drug effects , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Inhibitory Concentration 50 , Lymphokines/antagonists & inhibitors , Lymphokines/pharmacology , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/therapeutic use , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Substrate Specificity , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Cancer Res ; 55(19): 4243-7, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7671229

ABSTRACT

Farnesylation of the oncoprotein Ras is required for its cancer-causing activity. We have designed farnesyltransferase inhibitor (FTI)-276, a tetrapeptide mimetic of the carboxyl terminus of K-Ras4B, as a highly potent and selective inhibitor of Ras farnesylation in vitro and in vivo. FTI-276 blocked the growth in nude mice of a human lung carcinoma that expresses the two most prevalent genetic alterations in human cancers (K-Ras oncogenic mutation and deletion in the tumor suppressor gene p53). In contrast, FTI-276 did not inhibit tumor growth of a human lung carcinoma that harbors no Ras mutations. Furthermore, FTI-276 inhibited oncogenic signaling and tumor growth of NIH 3T3 cells transformed with the ras but not the raf oncogene. Inhibition of tumor growth in vivo was dose dependent and correlated with inhibition of Ras processing in tumors in vivo. The work described here identifies FTI-276 as a highly selective suppressor of Ras-dependent oncogenicity and suggests that a broad spectrum of human cancers with aberrant Ras function could benefit from farnesyltransferase inhibitor treatment.


Subject(s)
Alkyl and Aryl Transferases , Antineoplastic Agents/pharmacology , Genes, p53 , Genes, ras , Lung Neoplasms/prevention & control , Oligopeptides/pharmacology , Transferases/antagonists & inhibitors , 3T3 Cells , Animals , Farnesyltranstransferase , Female , Gene Deletion , Humans , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Transplantation, Heterologous
7.
Cancer Res ; 57(10): 1846-50, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9157972

ABSTRACT

The mechanism by which the geranylgeranyltransferase I inhibitor GGTI-298 and the farnesyltransferase inhibitor FTI-277 inhibit human tumor growth is not known. Herein, we demonstrate that in the human lung adenocarcinoma A549 cells, GGTI-298 induced a G1-G0 block whereas FTI-277 induced an enrichment in the G2-M phase of the cell cycle. Although FTI-277, GGTI-298, and compactin inhibited A549 cell growth, only GGTI-298 and compactin induced apoptosis as demonstrated by four criteria: 4',6-diamidine-2-phenylindoledihydrochloride staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, DNA fragmentation assay, and flow cytometry. Furthermore, the involvement of geranylgeranylated proteins in apoptotic pathways was confirmed by demonstrating that geranylgeraniol was able to block the ability of compactin to induce apoptosis. These results suggest that protein geranylgeranylation is critical for the control of programmed cell death and that, in A549 cells, farnesylated and geranylgeranylated proteins are involved in G2-M and G0-G1, respectively.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Alkyl and Aryl Transferases , Apoptosis/drug effects , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Methionine/analogs & derivatives , Adenocarcinoma/enzymology , Apoptosis/physiology , Cell Division/drug effects , G1 Phase/drug effects , G2 Phase/drug effects , Humans , Lung Neoplasms/enzymology , Methionine/pharmacology , Mitosis/drug effects , Resting Phase, Cell Cycle/drug effects , Transferases/antagonists & inhibitors , Tumor Cells, Cultured
8.
Cancer Res ; 61(4): 1280-4, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245420

ABSTRACT

The ubiquitin proteasome system is responsible for the proteolysis of important cell cycle and apoptosis-regulatory proteins. In this paper we report that the dipeptidyl proteasome inhibitor, phthalimide-(CH2)8CH-(cyclopentyl) CO-Arg(NO2)-Leu-H (CEP1612), induces apoptosis and inhibits tumor growth of the human lung cancer cell line A-549 in an in vivo model. In cultured A-549 cells, CEP1612 treatment results in accumulation of two proteasome natural substrates, the cyclin-dependent kinase inhibitors p21WAF1 and p27KIP1, indicating its ability to inhibit proteasome activity in intact cells. Furthermore, CEP1612 induces apoptosis as evident by caspase-3 activation and poly(ADP-ribose) polymerase cleavage. Treatment of A-549 tumor-bearing nude mice with CEP1612 (10 mg/kg/day, i.p. for 31 days) resulted in massive induction of apoptosis and significant (68%; P < 0.05) tumor growth inhibition, as shown by terminal deoxynucleotidyltransferase-mediated UTP end labeling. Furthermore, immunostaining of tumor specimens demonstrated in vivo accumulation of p21WAF1 and p27KIP1 after CEP1612 treatment. The results suggest that CEP1612 is a promising candidate for further development as an anticancer drug and demonstrate the feasibility of using proteasome inhibitors as novel antitumor agents.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/drug effects , Cell Cycle Proteins , Cyclins/biosynthesis , Dipeptides/pharmacology , Lung Neoplasms/pathology , Microtubule-Associated Proteins/biosynthesis , Phthalimides/pharmacology , Protease Inhibitors/pharmacology , Tumor Suppressor Proteins , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Female , Growth Inhibitors/pharmacology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cancer Res ; 56(8): 1727-30, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8620483

ABSTRACT

Many tumor cells have a greater resistance to ionizing radiation than their normal counterparts, suggesting that the development of drugs that can reduce that radioresistance would potentiate the efficacy of radiation therapy. Because activated H-ras expression has been shown to markedly increase radiation resistance in some transformed cells, the inactivation of H-ras would then be predicted to radiosensitize these tumor cells, while leaving normal cells unaffected. H-ras depends for activity upon farnesylation, which can be blocked by farnesylation inhibitors, including the compound FTI-277. In keeping with this prediction, inhibition of H-ras processing using FTI-277 resulted in higher levels of apoptosis after irradiation and increased radiosensitivity in H-ras-transformed rat embryo cells but did not affect control cells. These experiments suggest that farnesylation inhibitors may prove clinically useful as radiosensitizers of tumors that depend on ras function.


Subject(s)
Alkyl and Aryl Transferases , Apoptosis/drug effects , Cell Transformation, Neoplastic/drug effects , Enzyme Inhibitors/pharmacology , Genes, ras , Methionine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Transferases/antagonists & inhibitors , Animals , Apoptosis/radiation effects , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Embryo, Mammalian , Farnesyltranstransferase , Fibroblasts , Genes, myc , Genes, ras/drug effects , Kinetics , Methionine/pharmacology , Rats , Transfection , Urinary Bladder Neoplasms/genetics
10.
Cancer Res ; 59(19): 4919-26, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10519405

ABSTRACT

Ras malignant transformation requires posttranslational modification by farnesyltransferase (FTase). Here we report on the design and antitumor activity, in monotherapy as well as in combination therapy with cytotoxic agents, of a novel class of non-thiol-containing peptidomimetic inhibitors of FTase and the closely related family member geranylgeranyltransferase I (GGTase I). The non-thiol-containing FTI-2148 is highly selective for FTase (IC50, 1.4 nM) over GGTase I (IC50, 1700 nM), whereas GGTI-2154 is highly selective for GGTase I (21 nM) over FTase (IC50, 5600 nM). In whole cells, the corresponding methylester prodrug FTI-2153 is >3000-fold more potent at inhibiting H-Ras (IC50, 10 nM) than Rap1A processing, whereas GGTI-2166 is over 100-fold more selective at inhibiting Rap1A (IC50, 300 nM) over H-Ras processing. Furthermore, FTI-2153 was highly effective at suppressing oncogenic H-Ras constitutive activation of mitogen-activated protein kinase and human tumor growth in soft agar. FTI-2148 suppressed the growth of the human lung adenocarcinoma A-549 cells in nude mice by 33, 67, and 91% in a dose-dependent manner. Combination therapy of FTI-2148 with either cisplatin, gemcitabine, or Taxol resulted in a greater antitumor efficacy than monotherapy. GGTI-2154 in similar antitumor efficacy experiments is less potent than FTI-2148 and inhibits tumor growth by 9, 27, and 46%. Combination therapy of GGTI-2154 with cisplatin, gemcitabine, or Taxol is also more effective. Finally, FTI-2148 and GGTI-2154 are 30- and 33-fold more selective and 30- and 16-fold more potent in whole cells than our previously reported thiol-containing FTI-276 and GGTI-297, respectively. Thus, our results demonstrate that this highly potent and selective novel class of non-thiol-containing peptidomimetics inhibits human tumor growth in whole animals and that combination therapy with cytotoxic agents is more beneficial than monotherapy.


Subject(s)
Adenocarcinoma/drug therapy , Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/chemistry , Benzamides/therapeutic use , Enzyme Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Oligopeptides/chemistry , 3T3 Cells , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Benzamides/toxicity , Cell Division/drug effects , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Cisplatin/toxicity , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Farnesyltranstransferase , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/drug effects , Molecular Structure , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Paclitaxel/toxicity , Transplantation, Heterologous , Tumor Cells, Cultured , rap1 GTP-Binding Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Gemcitabine
11.
Cancer Res ; 58(8): 1754-61, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9563495

ABSTRACT

The influence of activated ras oncogenes on the sensitivity of human tumor cells to killing by radiation has been an unresolved question in radiobiology. We have examined this question by measuring the radiation sensitivity of human tumor cell lines with oncogenic mutations in their H- or K-ras genes after treatment with prenyltransferase inhibitors that prevent the posttranslational modification of ras required for its activity. Using two measures of clonogenic survival, we have demonstrated radiosensitization in cell lines with oncogenic H-ras mutations or with oncogenic K-ras mutations when ras processing was inhibited by prenyltransferase inhibitor treatment. In contrast, the inhibition of ras processing in cell lines expressing wild-type ras had no effect on radiation-induced cell death. The prenyltransferase inhibitors themselves inhibited clonogenic survival in some cases, but this inhibition did not correlate with ras mutational status. Although treatment with prenyltransferase inhibitors and radiation resulted in a greater reduction of clonogenicity than either treatment alone in cells with wild-type ras, treatment with both agents had a synergistic effect on cell killing in tumor cells with ras mutations. Our results demonstrate that the inhibition of oncogenic ras activity in human tumor cells can reduce the radiation survival of these cells, suggesting that oncogenic ras can contribute to radiation resistance in human tumors. These results further demonstrate the potential of using prenyltransferase inhibitors in combination with radiotherapy in the treatment of human malignancies.


Subject(s)
Genes, ras/genetics , Protein Prenylation/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Radiation Tolerance/drug effects , Blotting, Western , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Mutation , Proto-Oncogene Proteins p21(ras)/metabolism , Radiation-Sensitizing Agents/pharmacology , Time Factors , Tumor Cells, Cultured
12.
Oncogene ; 19(56): 6566-73, 2000 Dec 27.
Article in English | MEDLINE | ID: mdl-11426641

ABSTRACT

This review describes our recent efforts in the development of novel therapies for cancer. Our primary approach is to design synthetic agents that antagonize the function of growth factors that are critically involved in oncogenesis and angiogenesis. We achieve this by designing synthetic molecules that can recognize the exterior surface of the growth factor and so block the interaction with its receptor tyrosine kinase. A key step is the construction of synthetic agents that contain a large (> 400A2) and functionalized surface area to recognize a complementary surface on the target growth factor. In the course of this work we have discovered a molecule, GFB-111, that binds to PDGF, prevents it from binding to its receptor tyrosine kinase, blocks PDGF-induced receptor autophosphorylation, activation of Erk1 and Erk2 kinases and DNA synthesis. The binding affinity for PDGF is high (IC50=250 nM) and selective over EGF, IGF-1, aFGF, bFGF and HRGbeta. In nude mouse models GFB-111 also shows significant inhibition of tumor growth and angiogenesis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Design , Peptides, Cyclic/therapeutic use , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Angiogenesis Inhibitors/metabolism , Animals , Antineoplastic Agents/metabolism , Cell Division/drug effects , Mice , Mice, Nude , Models, Molecular , Peptides, Cyclic/metabolism , Protein Binding , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction
13.
Oncogene ; 19(56): 6584-93, 2000 Dec 27.
Article in English | MEDLINE | ID: mdl-11426643

ABSTRACT

In 1990, more than 10 years after the discovery that the low molecular weight GTPase Ras is a major contributor to human cancer, farnesylation, a lipid posttranslational modification required for the cancer-causing activity of Ras, emerged as a major target for the development of novel anticancer agents. However, it took only 5 years from 1993, when the first farnesyltransferase inhibitors (FTIs) were reported, to 1998 when results from the first phase I clinical trials were described. This rapid progress was due to the demonstration of outstanding antitumor activity and lack of toxicity of FTIs in preclinical models. Although, many FTIs are currently in phase H and at least one is in phase III clinical trial, the mechanism of FTI antitumor activity is not known. In this review a brief summary of the development of FTIs as antitumor agents will be given. The focus of the review will be on important mechanistic and bench-to-bedside translational issues. Among the issues that will be addressed are: evidence for and against inhibition of the prenylation of Ras and RhoB proteins in the mechanism of action of FTIs; implications of the alternative prenylation of K-Ras by geranylgeranyl-transferase I (when FTase is inhibited) in cancer therapy; GGTase I inhibitors (GGTIs) as antitumor agents; effects of FTIs and GGTIs on cell cycle machinery and progression and potential mechanisms by which FTIs and GGTIs induce apoptosis in human cancer cells. A thorough discussion about bench-to-bedside issues relating to hypothesis-driven clinical trials with proof-of-principle in man will also be included. This section will cover issues relating to whether the biochemical target (FTase) is inhibited and the level of inhibition of FTase required for clinical response; are signaling pathways such as H-Ras/PI3K/Akt and/or K-Ras/Raf/MEK/Erk relevant biological readouts?; is Ras (particularly N-Ras and H-Ras) mutation status a good predictor of clinical response?; in phase I trials should effective biological dose, not maximally tolerated dose, be used to determine phase II dose?; and finally, in phase II/III trials what are the most appropriate clinical end points for anti-signaling molecules such as FTIs? Parts of this topic have been recently reviewed (Sebti and Hamilton, 2000c).


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Animals , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Clinical Trials as Topic , Combined Modality Therapy , Drug Delivery Systems , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Humans , Mice , Proto-Oncogene Proteins p21(ras)/metabolism , Substrate Specificity , rhoB GTP-Binding Protein/metabolism
14.
Oncogene ; 16(11): 1467-73, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9525745

ABSTRACT

The ability of Ras oncoproteins to cause malignant transformation requires their post-translational modifications by prenyl groups. Because K-Ras can be both farnesylated and geranylgeranylated it is not known whether both farnesyltransferase and geranylgeranyltransferase I inhibitors are required for suppressing human tumor growth in whole animals. In this paper we report that oncogenic Ras processing, MAP kinase activation and growth in nude mice are inhibited by the farnesyltransferase inhibitor FTI-276 in H- and N-Ras transformed NIH3T3 cells; whereas in KB-Ras transformed NIH3T3 cells both FTI-276 and the geranylgeranyltransferase I inhibitor GGTI-297 are required for inhibition. Furthermore, human lung A-549 and Calu-1 carcinoma cell lines were found to co-express H-, N- and K-Ras. In Calu-1 cells, the processing of H- and N-Ras is inhibited greatly by FTI-276 but only partially by GGTI-297 whereas K-Ras processing inhibition requires both FTI-276 and GGTI-297. In contrast, in A-549 cells the processing of H- and N-Ras is inhibited only by FTI-276 and K-Ras processing is resistant to co-treatment with FTI-276 and GGTI-297. Yet, the growth in nude mice of A-549 and Calu-1 xenografts, both of which express K-Ras mutations, is inhibited by FTI-276 (80% inhibition) and GGTI-297 (60%). Furthermore, FTI-276 inhibits tumor growth of NIH3T3 cells transformed by a form of oncogenic H-Ras that is exclusively geranylgeranylated and whose processing is resistant to this inhibitor. Taken together, the results demonstrate that both FTase and GGTase I inhibitors are required for inhibition of K-Ras processing but that each alone is sufficient to suppress human tumor growth in nude mice.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Benzamides/pharmacology , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Methionine/analogs & derivatives , Oncogene Protein p21(ras)/metabolism , 3T3 Cells , Animals , Farnesyltranstransferase , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Methionine/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Protein Prenylation , Signal Transduction
15.
Oncogene ; 15(11): 1283-8, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9315095

ABSTRACT

The farnesyltransferase (FTase) inhibitor FTI-277 is highly effective at blocking oncogenic H-Ras but not K-Ras4B processing and signaling. While inhibition of processing and signaling of oncogenic K-Ras4B is more sensitive to the geranylgeranyltransferase I (GGTase I) inhibitor GGTI-286 than it is to FTI-277 in K-Ras4B-transformed NIH3T3 cells, the sensitivity of K-Ras as well as H- and N-Ras to the CAAX peptidomimetics in human tumor cell lines is not known. Here, we report that a panel of five human carcinoma cell lines from pancreatic, pulmonary, and bladder origins all express H-, N-, and K-Ras, and their respective prenylation sensitivities to the FTase and GGTase I inhibitors is variable. In all of the cell lines investigated, the prenylation of N-Ras was highly sensitive to FTI-277, and in two of the cell lines, N-Ras showed slight sensitivity to GGTI-298, an analog of GGTI-286. Although the prenylation of H-Ras was also sensitive to FTI-277, complete inhibition of H-Ras processing even at high concentrations of FTI-277 and/or GGTI-298 was never achieved. The prenylation of K-Ras, on the other hand, was highly resistant to FTI-277 and GGTI-298. Most significantly, treatment of human tumor cell lines with both inhibitors was required for inhibition of K-Ras prenylation. In one cell line, the human lung adenocarcinoma A-549, prenylation of K-Ras was highly resistant even when co-treated with both inhibitors. Furthermore, soft agar experiments demonstrated that in all the human tumor cell lines tested inhibition of K-Ras prenylation was not necessary for inhibition of anchorage-independent growth. In addition, although GGTI-298 had very little effect on soft agar growth, the combination of FTI-277 and GGTI-298 resulted in significant growth inhibition. Therefore, the results demonstrate that while FTI-277 inhibits N-Ras and H-Ras processing in the human tumor cell lines evaluated, inhibition of K-Ras processing requires both an FTase inhibitor as well as a GGTase I inhibitor, and that inhibition of human tumor growth in soft agar does not require inhibition of oncogenic K-Ras processing.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Genes, ras , ras Proteins/metabolism , Benzamides/pharmacology , Cell Division/drug effects , Cell Division/genetics , Farnesyltranstransferase , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/immunology , GTP-Binding Proteins/metabolism , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Peptides/pharmacology , Protein Prenylation/drug effects , Tumor Cells, Cultured , rap GTP-Binding Proteins , ras Proteins/genetics , ras Proteins/immunology
16.
Oncogene ; 13(9): 1991-9, 1996 Nov 07.
Article in English | MEDLINE | ID: mdl-8934546

ABSTRACT

In order to assess the relative contributions of farnesylated and/or geranylgeranylated proteins on cell cycle progression from G1 to S phase we designed potent and selective farnesyltransferase (FTI-277) and geranylgeranyltransferase-I (GGTI-298) inhibitors. Flow cytometry studies showed that treatment of NIH3T3 cells with GGTI-298 or lovastatin, which inhibits both protein farnesylation and geranylgeranylation, arrested cells in G0/G1 whereas cells treated with FTI-277 progressed normally through the cell cycle. [3H]thymidine incorporation studies showed that mevalonate and geranylgeraniol, but not farnesol, released the lovastatin G1 block. Furthermore, mevalonate release of the lovastatin G1 block was inhibited by GGTI-298 but not by FTI-277. These results demonstrate that geranylgeranylated proteins are required for cells to proceed from G1 to S phase, and that farnesylated proteins do not play an essential role in the G1 to S phase transition


Subject(s)
Alkyl and Aryl Transferases , G1 Phase/physiology , Protein Prenylation/physiology , S Phase/physiology , Actins/drug effects , Actins/ultrastructure , Animals , Benzamides/pharmacology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , DNA/biosynthesis , DNA/drug effects , Diterpenes/metabolism , Enzyme Inhibitors/pharmacology , Farnesol/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , G1 Phase/drug effects , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Genes, ras , Lovastatin/pharmacology , Methionine/analogs & derivatives , Methionine/pharmacology , Mevalonic Acid/pharmacology , Mice , Protein Prenylation/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/physiology , S Phase/drug effects , Transferases/antagonists & inhibitors , rap GTP-Binding Proteins , ras Proteins/drug effects , ras Proteins/metabolism
17.
Oncogene ; 19(48): 5525-33, 2000 Nov 16.
Article in English | MEDLINE | ID: mdl-11114730

ABSTRACT

Several small GTPases of the Ras superfamily have been shown to antagonize TGFbeta signaling in human tumor cell lines. Some of these GTPases are post-translationally modified by farnesylation, a lipid modification catalyzed by farnesyltransferase and required for the proteins to attach to membranes and to function. In this study, we investigated the effect of the farnesyltransferase inhibitor FTI-277 on TGFbeta-regulated cell growth and transcription. Treatment of the human pancreatic tumor cell line, Panc-1, with FTI-277 enhanced the ability of TGFbeta to inhibit both anchorage-dependent and -independent tumor cell growth. FTI-277 also enhanced the ability of TGFbeta to induce transcription, as measured by p3TP-lux reporter activity and collagen synthesis. The enhancement of TGFbeta responses by FTI-277 correlated with the stimulation of transcription and protein expression of type II TGFbeta receptor (TbetaRII). Consequently, FTI-277-treated cells exhibited a higher level of TGFbeta binding to its receptor. Thus, inhibition of protein farnesylation stimulates TbetaRII expression, which leads to increased TGFbeta receptor binding and signaling as well as inhibition of tumor cell growth and transformation.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Methionine/analogs & derivatives , Methionine/pharmacology , Receptors, Transforming Growth Factor beta/biosynthesis , Signal Transduction/physiology , Transforming Growth Factor beta/pharmacology , 3T3 Cells/drug effects , 3T3 Cells/metabolism , Animals , Cell Division/drug effects , Cell Transformation, Neoplastic/drug effects , Drug Synergism , Farnesyltranstransferase , Humans , Mice , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Tumor Cells, Cultured/drug effects
18.
Cell Death Differ ; 9(7): 702-9, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12058275

ABSTRACT

Recently, we have shown that the farnesyltransferase inhibitor FTI-2153 induces accumulation of two human lung cancer cell lines in mitosis by inhibiting bipolar spindle formation during prometaphase. Here we investigate whether this mitotic arrest depends on transformation, Ras and/or p53 mutation status. Using DAPI staining (DNA) and immunocytochemistry (microtubules), we demonstrate that in normal primary foreskin fibroblasts (HFF), as well as in several cancer cell lines of different origins including human ovarian (OVCAR3), lung (A-549 and Calu-1) and fibrosarcoma (HT1080), FTI-2153 inhibits bipolar spindle formation and induces a rosette morphology with a monopolar spindle surrounded by chromosomes. In both malignant cancer cell lines and normal primary fibroblasts, the percentage of prometaphase cells with bipolar spindles decreases from 67-92% in control cells to 2-28% in FTI-2153 treated cells. This inhibition of bipolar spindle formation correlates with an accumulation of cells in prometaphase. The ability of FTI-2153 to inhibit bipolar spindle formation is not dependent on p53 mutation status since both wild-type (HFF, HT1080 and A-549) and mutant (Calu-1 and OVCAR3) p53 cells were equally affected. Similarly, both wild-type (HFF and OVCAR3) and mutant (HT1080, Calu-1 and A-549) Ras cells accumulate monopolar spindles following treatment with FTI-2153. However, two cell lines, NIH3T3 (WT Ras and WT p53) and the human bladder cancer cell line, T-24 (mutant H-Ras and mutant p53) are highly resistant to FTI-2153 inhibition of bipolar spindle formation. Finally, the ability of FTI-2153 to inhibit tumor cell proliferation does not correlate with inhibition of bipolar spindle formation. Taken together these results demonstrate that the ability of FTI-2153 to inhibit bipolar spindle formation and accumulate cells in mitosis is not dependent on transformation, Ras or p53 mutation status. Furthermore, in some cell lines, FTIs inhibit growth by mechanisms other than interfering with the prophase/metaphase traverse.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Mitosis/drug effects , Spindle Apparatus/drug effects , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism , 3T3 Cells , Animals , Cell Division/drug effects , Farnesyltranstransferase , Humans , Metaphase , Mice , Mitosis/physiology , Mutagenesis , Spindle Apparatus/physiology , Transformation, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics
19.
Pharmacol Ther ; 74(1): 103-14, 1997.
Article in English | MEDLINE | ID: mdl-9336018

ABSTRACT

The demonstration that Ras requires prenylation for its cancer-causing activity led several groups of investigators to an intense search for farnesyltransferase and geranylgeranyltransferase inhibitors as potential anticancer drugs. Rational design of small organic molecules that mimic the carboxyl terminal tetrapeptide prenylation site on Ras resulted in pharmacological agents capable of inhibiting Ras processing and selectively antagonizing oncogenic signaling, and suppressing human tumor growth in mouse models without side effects. These agents presently are undergoing advanced preclinical studies. This review describes the efforts of several groups to design, synthesize and evaluate the biological activities of several classes of prenyltransferase inhibitors. Several important issues, such as mechanism of action of prenyltransferase inhibitors and potential mechanisms of resistance to inhibition of K-Ras farnesylation, are also discussed.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , ras Proteins/genetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/standards , Drug Design , Farnesyltranstransferase , Humans , Neoplasms/drug therapy , Protein Prenylation/drug effects
20.
Curr Opin Chem Biol ; 3(3): 307-12, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10359712

ABSTRACT

A combinatorial approach to receptor design provides an expedient method to discover the most effective host-guest complexes from within a library. Recent advances focus on generation of larger libraries, facile detection, combinatorial catalysis and the formation of dynamic receptor libraries.


Subject(s)
Receptors, Drug/chemistry , Chemistry, Organic , Ligands , Organic Chemistry Phenomena , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL