Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Toxicology ; 258(2-3): 116-30, 2009 Apr 28.
Article in English | MEDLINE | ID: mdl-19428931

ABSTRACT

Ezetimibe blocks intestinal absorption of sterols via interaction with the Neimann-Pick C1-Like 1 (NPC1L1) transporter and is approved for use in the treatment of primary hyperlipidemia (heterozygous familial and non-familial), homozygous familial hypercholesterolemia, and homozygous sitosterolemia. A recently completed randomized clinical trial [simvastatin and ezetimibe in aortic stenosis (SEAS)] testing the effectiveness of Vytorin (a combination of simvastatin and ezetimibe) in patients with aortic stenosis reported an unexpected safety finding: an increase in overall cancer incidence and cancer-associated mortality (all types) in the treated groups relative to the placebo control. A subsequent meta-analysis utilizing a much larger database from two ongoing clinical trials indicated that the observed findings in the SEAS trial were likely due to chance and not a true drug-induced effect. Nonetheless, it has been suggested by various commentators on the SEAS trial that ezetimibe may be carcinogenic. The extensive nonclinical database for ezetimibe was used to test the hypothesis that ezetimibe may be a direct or indirect carcinogen. Using two different in silico approaches, ezetimibe showed no structural alerts for genetic toxicity or carcinogenicity. Ezetimibe was not genotoxic in two reverse mutation assays, one in vitro clastogenicity assay, and two mouse micronucleus assays. No evidence of proliferative lesions was observed in three species in studies of 1-12 months in duration. Ezetimibe was not carcinogenic in standard 2-year bioassays in mice and rats. Additionally, in these 2-year bioassays, no drug-related non-neoplastic lesions were noted. The absence of drug-induced non-neoplastic or proliferative lesions in these studies indicates that ezetimibe treatment was not associated with findings characteristic of carcinogens (i.e., DNA reactivity or cell proliferation) Administration of pharmacologic doses of ezetimibe to mice did not alter hepatic expression patterns of genes associated with apoptosis, cell proliferation, or epithelial-mesenchymal transition. No evidence of drug-induced tumors was observed in mice in which the molecular target of ezetimibe (NPC 1L1) was knocked out over the life span of the animal. In conclusion, the nonclinical data do not support the proposed hypothesis based on the single observation from the SEAS trial and, rather, support the conclusion that ezetimibe does not represent a carcinogenic hazard to humans using this drug in a therapeutic setting.


Subject(s)
Anticholesteremic Agents/toxicity , Azetidines/toxicity , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/chemistry , Azetidines/administration & dosage , Azetidines/chemistry , Carcinogenicity Tests , Carcinogens/toxicity , Dogs , Ezetimibe , Female , Gene Expression/drug effects , Male , Membrane Transport Proteins/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Mutagenicity Tests , Rats , Rats, Sprague-Dawley
2.
Leukemia ; 22(9): 1707-11, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18548095

ABSTRACT

Lonafarnib is an orally bio-available farnesyltransferase inhibitor that prevents farnesylation of specific target proteins including Ras. In a multicenter study, 67 patients with advanced myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) were treated with a continuous oral dose of 200-300 mg of lonafarnib and were evaluated for hematologic, pathologic and pharmacodynamic response. The median age of patients was 70 years (range 44-86). There were 32 patients with MDS (RAEB-20 and RAEB-t-12) and 35 with CMML. Overall 16 (24%) of the patients responded with two patients achieving a complete remission and one a partial response. Responses were seen in 6/32 and 10/35 patients with MDS and CMML, respectively. Of the 19 patients who were platelet transfusion-dependent prior to treatment, 5 (26%) became transfusion-free for a median duration of 185 days. A decrease in the farnesylation of the HDJ-2 protein measured in patient-derived cells was observed in the majority of patients during treatment with lonafarnib, but no clear correlation between changes in farnesylation and clinical effect could be made. Gastrointestinal toxicity was significant with 19% of patients discontinuing therapy due to diarrhea, nausea and/or anorexia. Lonafarnib has demonstrable activity in patients with advanced MDS and CMML.


Subject(s)
Leukemia, Myelomonocytic, Chronic/drug therapy , Myelodysplastic Syndromes/drug therapy , Piperidines/administration & dosage , Pyridines/administration & dosage , Adult , Aged , Aged, 80 and over , Drug Monitoring , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Gastrointestinal Diseases/chemically induced , Humans , Leukemia, Myelomonocytic, Chronic/complications , Maximum Tolerated Dose , Middle Aged , Myelodysplastic Syndromes/complications , Piperidines/toxicity , Pyridines/toxicity , Remission Induction , Treatment Outcome
3.
J Biol Chem ; 276(47): 43604-10, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11571296

ABSTRACT

The completion of the human genome sequence (International Human Genome Sequence Consortium (2001) Nature 409, 860-921; Venter, J. C., et al. (2001) Science 291, 1304-1351) allows for new ways to analyze global cellular regulatory mechanisms. Here we present a strategy to identify genes regulated by specific transcription factors in the human genome, and apply it to p53. We first collected promoters or introns of all genes available using two methods: GenBank(TM) annotation and a computationally derived transcript map. 4,852 genes analyzed in this way contained at least one p53 consensus binding sequence. Of 13 genes randomly selected for mRNA analysis, 11 were shown to respond to p53 expression. Five promoters were analyzed by chromatin immunoprecipitation, which revealed that all were bound by p53 in vivo. We then analyzed 33,615 unique human genes on cDNA microarrays, identifying 1,501 genes that respond to p53 expression. A parameter was derived that demonstrates that in silico prediction greatly enriches for genes that are activated and repressed by p53 and assists us to suggest other signaling pathways that may be connected to p53. The methods shown here illustrate a novel approach to analysis of global gene regulatory network through the integration of human genomic sequence information and genome-wide gene expression analysis.


Subject(s)
Computational Biology , Genes, p53 , Genome, Human , Oligonucleotide Array Sequence Analysis , Gene Expression Profiling , Humans , RNA, Messenger/genetics , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
4.
Methods Enzymol ; 333: 306-18, 2001.
Article in English | MEDLINE | ID: mdl-11400347

ABSTRACT

The in vivo evaluation process described here was instrumental in the identification of SCH 66336 as a clinical candidate. Our lead FTI, SCH 66336, and several other FTIs are being evaluated in early-phase clinical trials to establish proof-of-principle for farnesyl transferase inhibition in human patients. The preclinical studies described here suggest that FTIs may have utility against a wide array of human cancers as a single agent and may, at least in some cases, lead to tumor regression. In addition, the results to date in combination with cytotoxic chemotherapeutic agents in animal models indicate that these combinations may enhance the clinical efficacy of FPT inhibitors. Further preclinical studies should help to guide the clinical development of this class of novel antitumor agents.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Antineoplastic Agents/metabolism , Enzyme Inhibitors/metabolism , 3T3 Cells , Administration, Oral , Alkyl and Aryl Transferases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Apoptosis , Biological Availability , Cell Division , Cell Transformation, Neoplastic , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Farnesyltranstransferase , Fibroblasts/metabolism , Genes, ras , Humans , Mice , Mice, Transgenic , Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Blood ; 97(5): 1404-12, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222387

ABSTRACT

BCR/ABL, the oncoprotein responsible for chronic myeloid leukemia (CML), transforms hematopoietic cells through both Ras-dependent and -independent mechanisms. Farnesyl protein transferase inhibitors (FTIs) were designed to block mutant Ras signaling, but they also inhibit the growth of transformed cells with wild-type Ras, implying that other farnesylated targets contribute to FTI action. In the current study, the clinical candidate FTI SCH66336 was characterized for its ability to inhibit BCR/ABL transformation. When tested against BCR/ABL-BaF3 cells, a murine cell line that is leukemogenic in mice, SCH66336 potently inhibited soft agar colony formation, slowed proliferation, and sensitized cells to apoptotic stimuli. Quantification of activated guanosine triphosphate (GTP)-bound Ras protein and electrophoretic mobility shift assays for AP-1 DNA binding showed that Ras effector pathways are inhibited by SCH66336. However, SCH66336 was more inhibitory than dominant-negative Ras in assays of soft agar colony formation and cell proliferation, suggesting activity against targets other than Ras. Cell cycle analysis of BCR/ABL-BaF3 cells treated with SCH66336 revealed G2/M blockade, consistent with recent reports that centromeric proteins that regulate the G2/M checkpoint are critical farnesylated targets of FTI action. Mice injected intravenously with BCR/ABL-BaF3 cells developed acute leukemia and died within 4 weeks with massive splenomegaly, elevated white blood cell counts, and anemia. In contrast, nearly all mice treated with SCH66336 survived and have remained disease-free for more than a year. Furthermore, SCH66336 selectively inhibited the hematopoietic colony formation of primary human CML cells. As an oral, nontoxic compound with a mechanism of action distinct from that of ABL tyrosine kinase inhibition, FTI SCH66336 shows promise for the treatment of BCR/ABL-induced leukemia.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Farnesyltranstransferase , Genes, abl/physiology , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Spleen/drug effects , Spleen/pathology , Survival Rate , Transformation, Genetic , Tumor Cells, Cultured
7.
Exp Cell Res ; 262(1): 17-27, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11120601

ABSTRACT

SCH 66336 is a potent farnesyl transferase inhibitor (FTI) in clinical development. It efficiently prevents the membrane association of H-ras, but not K- or N-ras. Yet, in soft agar, it reverts the anchorage-independent growth of human tumor cell lines (hTCLs) harboring H-ras, K-ras, and N-ras mutations, implying that blocking farnesylation of proteins besides ras may be responsible for this effect. Experiments show that SCH 66336 altered the cell cycle distribution of sensitive human tumor cells in two distinct ways. Most sensitive hTCLs accumulated in the G(2)-->M phase after the FTI treatment, but those with an activated H-ras accumulated in G(1) phase, suggesting that the biological effects induced by FTIs in cells with an activated H-ras are distinct from other sensitive cells. A careful genotypic comparison of the hTCLs revealed that those cells with wild-type p53 are especially sensitive to the FTIs. In these cells p53 and its downstream target gene p21(Cip1) are induced after treatment with SCH 66336 for 24 h. These data suggest that cell cycle effects, either G(1) or G(2)-->M accumulation, and p53 status are important for mediating the effects of FTIs on tumor cells.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Cell Cycle/drug effects , 3T3 Cells , Animals , Cell Division/drug effects , Cell Membrane/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , G1 Phase , G2 Phase , Humans , K562 Cells , Kinetics , Mice , Mitosis , Molecular Structure , Oncogene Protein p21(ras)/metabolism , Piperidines/chemistry , Piperidines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
9.
J Biol Chem ; 275(39): 30451-7, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-10852915

ABSTRACT

Human tumor cell lines that are sensitive to the effects of farnesyl transferase inhibitors accumulate in G(2) --> M (except for cells with an activated Ha-ras that accumulate in G(1)). A search for CAAX box proteins from Swiss-Prot revealed more than 300 peptides. Of these, the centromeric proteins CENP-E and CENP-F are preferentially expressed during mitosis and are implicated as mediators of the G(2) --> M checkpoint. Experiments performed here show that peptides from the COOH-terminal CAAX box of CENP-E and CENP-F are substrates for farnesyl transferase but not geranylgeranyl transferase-I. Although both proteins are prenylated in the human tumor cell line DLD-1, their prenylation is completely inhibited by the farnesyl transferase inhibitor, SCH 66336. Immunohistochemical data with the lung carcinoma cell line, A549, showed that preventing the farnesylation of CENP-E and CENP-F by treatment with the farnesyl transferase inhibitor SCH 66336 does not affect their localization to the kinetochores. However, the presence of farnesyl transferase inhibitors alters the association between CENP-E and the microtubules. Our results imply that the inhibition of CENP-E farnesylation results in the alteration of the microtubule-centromere interaction during mitosis and results in the accumulation of cells prior to metaphase.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Prenylation , Cell Cycle/drug effects , Enzyme Inhibitors/pharmacology , Humans , Mevalonic Acid/metabolism , Microfilament Proteins , Piperidines/pharmacology , Pyridines/pharmacology , Substrate Specificity , Tumor Cells, Cultured
10.
Cancer Res ; 60(7): 1871-7, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10766174

ABSTRACT

Farnesyl protein transferase (FT), an enzyme that catalyzes the first step in the posttranslational modification of ras and a number of other polypeptides, has emerged as an important target for the development of anticancer agents. SCH66336 is one of the first FT inhibitors to undergo clinical testing. We report a Phase I trial to assess the maximum tolerated dose, toxicities, and biological effectiveness of SCH66336 in inhibiting FT in vivo. Twenty patients with solid tumors received 92 courses of escalating SCH66336 doses given orally twice a day (b.i.d.) for 7 days out of every 3 weeks. Gastrointestinal toxicity (nausea, vomiting, and diarrhea) and fatigue were dose-limiting at 400 mg of SCH66336 b.i.d. Moderate reversible renal insufficiency, secondary to dehydration from gastrointestinal toxicity, was also seen. Inhibition of prelamin A farnesylation in buccal mucosa cells of patients treated with SCH66336 was demonstrated, confirming that SCH66336 inhibits protein farnesylation in vivo. One partial response was observed in a patient with previously treated metastatic non-small cell lung cancer, who remained on study for 14 months. This study not only establishes the dose for future testing on this schedule (350 mg b.i.d.) but also provides the first evidence of successful inhibition of FT in the clinical setting and the first hint of clinical activity for this class of agents.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Piperidines/adverse effects , Pyridines/adverse effects , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Dose-Response Relationship, Drug , Enzyme Inhibitors/adverse effects , Farnesyltranstransferase , Female , Humans , Lamin Type A , Lamins , Male , Middle Aged , Mouth Mucosa/pathology , Nuclear Proteins/analysis , Piperidines/administration & dosage , Protein Precursors/analysis , Pyridines/administration & dosage
11.
J Med Chem ; 42(14): 2651-61, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10411485

ABSTRACT

Farnesyl protein transferase (FPT) is a promising target for the development of cancer chemotherapeutics because it is responsible for the farnesylation of oncogenic p21 Ras proteins which are found in nearly 30% of all human cancers and necessary for cellular development and growth. The recent discovery and progression to phase II clinical trials of trihalobenzocycloheptapyridine Sch-66336 as a potent inhibitor of FPT with oral, in vivo efficacy in mice have spawned extensive structure-activity relationship studies (SAR) of this class of compounds. Of the many trihalobenzocycloheptapyridine analogues prepared, we have identified several which inhibit FPT and cellular proliferation at single-digit nanomolar concentrations and which have good pharmacokinetic properties in mice.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Piperidines/chemical synthesis , Pyridines/chemical synthesis , Sulfonamides/chemical synthesis , Sulfonylurea Compounds/chemical synthesis , Administration, Oral , Animals , Biological Availability , COS Cells , Cell Division/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Haplorhini , Mice , Mice, Nude , Piperidines/chemistry , Piperidines/pharmacokinetics , Protein Prenylation , Proto-Oncogene Proteins p21(ras)/metabolism , Pyridines/chemistry , Pyridines/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/pharmacokinetics
12.
Bioorg Med Chem ; 7(5): 861-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10400339

ABSTRACT

Introduction of bromine at the 10-position of 3-bromo-8-chloro-benzocycloheptapyridine analogues of type 3 results in formation of atropisomeric compounds of type (+/-)-1 and (+/-)-2 that are easily separable at room temperature on a ChiralPak AD column providing pure atropisomers, (+)-1, (-)-1, and (+)-2, (-)-2, respectively. Evaluation of the FPT activity of these atropisomers revealed that compounds (+)-1 and (+)-2 were more potent in the FPT enzyme and cellular assay than their (-)-isomer counterparts. Compounds (+)-1 and (+)-2 were found to inhibit FPT processing in COS cells at low micro molar range. They were also found to have excellent cellular antitumor activity. Evaluation of compound (+)-1 and (+)-2 in DLD-tumor model in nude mice revealed that they were efficacious, inhibiting tumor growth by 55 and 63% at 50 mpk, respectively.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Pyridines/chemical synthesis , Pyridines/pharmacology , Pyridines/pharmacokinetics , Animals , COS Cells , Dose-Response Relationship, Drug , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Nude , Models, Chemical , Time Factors , Tumor Cells, Cultured
13.
Cancer Chemother Pharmacol ; 43(1): 50-8, 1999.
Article in English | MEDLINE | ID: mdl-9923541

ABSTRACT

The products of the Ha-, Ki-, and N-ras proto-oncogenes comprise a family of 21 kDa guanine nucleotide-binding proteins which play a crucial role in growth factor signal transduction and in the control of cellular proliferation and differentiation. Activating mutations in the ras oncogenes occur in a wide variety of human tumors. Ras proteins undergo a series of posttranslational processing events. The first modification is addition of the 15-carbon isoprene, farnesyl, to a Cys residue near the carboxy-terminus of Ras. Prenylation allows the Ras oncoprotein to localize to the plasma membrane where it can initiate downstream signalling events leading to cellular transformation. Inhibitors of the enzyme which catalyzes this step, farnesyl protein transferase (FPT), are a potential class of novel anticancer drugs which interfere with Ras function. SCH 59228 is a tricyclic FPT inhibitor which inhibits the farnesylation of purified Ha-Ras with an IC50 of 95 nM and blocks the processing of Ha-Ras in Cos cells with an IC50 of 0.6 microM. SCH 59228 has favorable pharmacokinetic properties upon oral dosing in nude mice. The in vivo efficacy of SCH 59228 was evaluated using a panel of tumor models grown in nude mice. These included several rodent fibroblast lines expressing mutationally-activated (val12) forms of the Ha-Ras oncogene. In some cases, these proteins contain their native C-terminal sequence (CVLS) which directs farnesylation. In one model, the C-terminal sequence was altered to CVLL, making the expressed protein a substrate for a distinct prenyl transferase, geranylgeranyl protein transferase-1. When dosed orally at 10 and 50 mg/kg (four times a day, 7 days a week) SCH 59228 significantly inhibited tumor growth of cells expressing farnesylated Ha-Ras in a dose-dependent manner; over 90% growth inhibition was observed at the 50 mg/kg dose. Tumor growth of cells expressing the geranylgeranylated form of Ha-Ras was less potently inhibited. Growth of tumors derived from a rodent fibroblast line expressing activated Ki-Ras containing its native C-terminal sequence (CVIM), which preferentially directs farnesylation, was also inhibited by SCH 59228. Inhibition in the Ki-Ras model was less than that observed in the Ha-Ras model. In contrast, tumors derived from cells transformed with the mos oncogene were not significantly inhibited even at the highest dose level. SCH 59228 also significantly and dose-dependently inhibited the growth of human colon adenocarcinoma DLD-1 xenografts (which express activated Ki-ras). These results indicate that SCH 59228 possesses in vivo antitumor activity upon oral dosing in tumor models expressing activated ras oncogenes. This is the first report of oral antitumor activity with an FPT inhibitor. These results are discussed in light of recent observations on alternative prenylation of some Ras isoforms.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Colonic Neoplasms/pathology , Cyclic N-Oxides/pharmacology , Enzyme Inhibitors/pharmacology , Genes, ras , Piperazines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Cell Division/drug effects , Cell Line, Transformed , Colonic Neoplasms/drug therapy , Cyclic N-Oxides/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Fibroblasts , Genes, mos , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Piperazines/pharmacokinetics , Transfection
14.
Biochemistry ; 37(45): 15631-7, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9843367

ABSTRACT

Mutated, tumorigenic Ras is present in a variety of human tumors. Compounds that inhibit tumorigenic Ras function may be useful in the treatment of Ras-related tumors. The interaction of a novel GDP exchange inhibitor (SCH-54292) with the Ras-GDP protein was studied by NMR spectroscopy. The binding of the inhibitor to the Ras protein was enhanced at low Mg2+ concentrations, which enabled the preparation of a stable complex for NMR study. To understand the enhanced inhibitor binding and the increased GDP dissociation rates of the Ras protein, the conformational changes of the Ras protein at low Mg2+ concentrations was investigated using two-dimensional 1H-15N HSQC experiments. The Ras protein existed in two conformations in slow exchange on the NMR time scale under such conditions. The conformational changes mainly occurred in the GDP binding pocket, in the switch I and the switch II regions, and were reversible. The Ras protein resumed its regular conformation after an excess amount of Mg2+ was added. A model of the inhibitor in complex with the Ras-GDP protein was derived from intra- and intermolecular NOE distance constraints, and revealed that the inhibitor bound to the critical switch II region of the Ras protein.


Subject(s)
Glucosides/metabolism , Guanosine Diphosphate/metabolism , Proteins/antagonists & inhibitors , Sulfonamides/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Binding Sites , Computer Simulation , Glucosides/chemistry , Guanine Nucleotide Exchange Factors , Humans , Macromolecular Substances , Magnesium/metabolism , Models, Molecular , Molecular Sequence Data , Monte Carlo Method , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Proteins/chemistry , Sulfonamides/chemistry , ras Guanine Nucleotide Exchange Factors
15.
J Med Chem ; 41(24): 4890-902, 1998 Nov 19.
Article in English | MEDLINE | ID: mdl-9822558

ABSTRACT

We have previously shown that appropriate modification of the benzocycloheptapyridine tricyclic ring system can provide potent farnesyl protein transferase (FPT) inhibitors with good cellular activity. Our laboratories have also established that incorporation of either pyridinylacetyl N-oxide or 4-N-carboxamidopiperidinylacetyl moieties results in pharmacokinetically stable inhibitors that are orally efficacious in nude mice. We now demonstrate that further elaboration of the tricyclic ring system by introducing a bromine atom at the 7- or the 10-position of the 3-bromo-8-chlorotricyclic ring system provides compounds that have superior potency and selectivity in FPT inhibition. These compounds have good serum levels and half-lives when given orally to rodents and primates. In vitro and in vivo evaluation of a panel of these inhibitors has led to identification of 15 (SCH 66336) as a highly potent (IC50 = 1.9 nM) antitumor agent that is currently undergoing human clinical trials.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Piperidines/chemical synthesis , Protein Prenylation/drug effects , Pyridines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , COS Cells , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Macaca fascicularis , Mice , Mice, Nude , Piperidines/chemistry , Piperidines/pharmacokinetics , Piperidines/pharmacology , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
16.
J Med Chem ; 41(6): 877-93, 1998 Mar 12.
Article in English | MEDLINE | ID: mdl-9526562

ABSTRACT

The synthesis of a variety of novel 4-amido, 4-carbamoyl and 4-carboxamido derivatives of 1-(8-chloro-6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-yl) piperazine to explore the SAR of this series of FPT inhibitors is described. This resulted in the synthesis of the 4- and 3-pyridylacetyl analogues 45a and 50a, respectively, both of which were orally active but were found to be rapidly metabolized in vivo. Identification of the principal metabolites led to the synthesis of a variety of new compounds that would be less readily metabolized, the most interesting of which were the 3- and 4-pyridylacetyl N-oxides 80a and 83a. Novel replacements for the pyridylacetyl moiety were also sought, and this resulted in the discovery of the 4-N-methyl and 4-N-carboxamidopiperidinylacetyl derivatives 135a and 160a, respectively. All of these derivatives exhibited greatly improved pharmacokinetics. The synthesis of the corresponding 3-bromo analogues resulted in the discovery of the 4-pyridylacetyl N-oxides 83b (+/-) and 85b [11S(-)] and the 4-carboxamidopiperidinylacetamido derivative 160b (+/-), all of which exhibited potent FPT inhibition in vitro. All three showed excellent oral bioavailability in vivo in nude mice and cynomolgus monkeys and exhibited excellent antitumor efficacy against a series of tumor cell lines when dosed orally in nude mice.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Cyclic N-Oxides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Piperazines/chemical synthesis , Piperidines/chemical synthesis , 3T3 Cells , Administration, Oral , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , COS Cells , Cell Line, Transformed , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/metabolism , Cyclic N-Oxides/pharmacokinetics , Drug Screening Assays, Antitumor , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Genes, ras , Macaca fascicularis , Mice , Mice, Nude , Neoplasm Transplantation , Piperazines/chemistry , Piperazines/metabolism , Piperazines/pharmacokinetics , Piperidines/chemistry , Piperidines/metabolism , Piperidines/pharmacokinetics , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 8(18): 2521-6, 1998 Sep 22.
Article in English | MEDLINE | ID: mdl-9873573

ABSTRACT

Bioisosteric replacement of the C-6 carbon atom in piperidine I and piperazine II with S, O, and N heteroatoms is described. Amide and cyanoguanidine derivatives of these compounds were evaluated in vitro and found to be good inhibitors of farnesyl-protein transferase. An improved method of preparing the 5,11-dihydro-[1]-benzthiepin nucleus 6 was accomplished in high yield and with excellent regioselectivity using an AlCl3 melt protocol.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Amides/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Guanidines/chemistry , Protein Prenylation/drug effects , Benzazepines/chemistry , Benzoxepins/chemistry , Models, Chemical , Piperazines/chemistry , Piperidines/chemistry , Pyridines/chemistry , Structure-Activity Relationship
18.
Bioorg Med Chem ; 5(5): 817-20, 1997 May.
Article in English | MEDLINE | ID: mdl-9208093

ABSTRACT

MS based methodology employing electrospray ionization (ESI) is described for the detection of ternary complexes in which SCH 54292 or SCH 54341 and GDP are noncovalently bound to oncogenic ras protein. The observed molecular weights of 19,816 and 19,570 Da confirmed the presence of noncovalent complexes of ras-GDP-SCH 54292 and ras-GDP-SCH 54341, respectively. We have also performed selective chemical modification of lysine residues of the ras protein complex followed by enzymatic digestion and on-line LC-ESI MS peptide mapping to determine protein-drug binding topography. There was a good correlation between nucleotide exchange inhibition as determined by the enzyme assay and evidence of complex formation as determined by MS.


Subject(s)
ras Proteins/antagonists & inhibitors , ras Proteins/chemistry , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Drug Evaluation, Preclinical , Glucosides/chemistry , Glucosides/pharmacology , Guanine Nucleotide Exchange Factors , In Vitro Techniques , Macromolecular Substances , Mass Spectrometry/methods , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Proteins/antagonists & inhibitors , Proteins/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology , ras Guanine Nucleotide Exchange Factors , ras Proteins/genetics
19.
J Biol Chem ; 272(22): 14459-64, 1997 May 30.
Article in English | MEDLINE | ID: mdl-9162087

ABSTRACT

The association of mutant forms of Ras protein with a variety of human cancers has stimulated intense interest in therapies based on inhibiting oncogenic Ras signaling. Attachment of Ras proteins to the plasma membrane is required for effective Ras signaling and is initiated by the enzyme farnesyl protein transferase. We found that in the presence of potent farnesyl protein transferase inhibitors, Ras proteins in the human colon carcinoma cell line DLD-1 were alternatively prenylated by geranylgeranyl transferase-1. When H-Ras, N-Ras, K-Ras4A, and K-Ras4B were expressed individually in COS cells, H-Ras prenylation and membrane association were found to be uniquely sensitive to farnesyl transferase inhibitors; N- and K-Ras proteins incorporated the geranylgeranyl isoprene group and remained associated with the membrane fraction. The alternative prenylation of N- and K-Ras has significant implications for our understanding of the mechanism of action of farnesyl protein transferase inhibitors as anti-cancer chemotherapeutics.


Subject(s)
Alkyl and Aryl Transferases , Enzyme Inhibitors/pharmacology , Protein Prenylation/drug effects , Transferases/antagonists & inhibitors , ras Proteins/metabolism , Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Humans , Tumor Cells, Cultured
20.
J Biol Chem ; 272(15): 10232-9, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9092572

ABSTRACT

Ras proteins are small GTP-binding proteins which are critical for cell signaling and proliferation. Four Ras isoforms exist: Ha-Ras, N-Ras, Ki-Ras4A, and Ki-Ras4B. The carboxyl termini of all four isoforms are post-translationally modified by farnesyl protein transferase (FPT). Prenylation is required for oncogenic Ras to transform cells. Recently, it was reported that Ki-Ras4B is also an in vitro substrate for the related enzyme geranylgeranyl protein transferase-1 (GGPT-1) (James, G. L., Goldstein, J. L., and Brown, M. S. (1995) J. Biol. Chem. 270, 6221-6226). In the current studies, we compared the four isoforms of Ras as substrates for FPT and GGPT-1. The affinity of FPT for Ki-Ras4B (Km = 30 nM) is 10-20-fold higher than that for the other Ras isoforms. Consistent with this, when the different Ras isoforms are tested at equimolar concentrations, it requires 10-20-fold higher levels of CAAX-competitive compounds to inhibit Ki-Ras4B farnesylation. Additionally, we found that, as reported for Ki-Ras4B, N-Ras and Ki-Ras4A are also in vitro substrates for GGPT-1. Of the Ras isoforms, N-Ras is the highest affinity substrate for GGPT-1 and is similar in affinity to a standard GGPT-1 substrate terminating in leucine. However, the catalytic efficiencies of these geranylgeranylation reactions are between 15- and 140-fold lower than the corresponding farnesylation reactions, largely reflecting differences in affinity. Carboxyl-terminal peptides account for many of the properties of the Ras proteins. One interesting exception is that, unlike the full-length N-Ras protein, a carboxyl-terminal N-Ras peptide is not a GGPT-1 substrate, raising the possibility that upstream sequences in this protein may play a role in its recognition by GGPT-1. Studies with various carboxyl-terminal peptides from Ki-Ras4B suggest that both the carboxyl-terminal methionine and the upstream polylysine region are important determinants for geranylgeranylation. Furthermore, it was found that full-length Ki-Ras4B, but not other Ras isoforms, can be geranylgeranylated in vitro by FPT. These findings suggest that the different distribution of Ras isoforms and the ability of cells to alternatively process these proteins may explain in part the resistance of some cell lines to FPT inhibitors.


Subject(s)
Alkyl and Aryl Transferases , Transferases/metabolism , ras Proteins/metabolism , Benzazepines/pharmacology , Binding Sites , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Methionine/metabolism , Piperidines/pharmacology , Polylysine/metabolism , Protein Prenylation
SELECTION OF CITATIONS
SEARCH DETAIL
...