Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 111(5): 894-902, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-24983364

ABSTRACT

BACKGROUND: The binding of STAT3 and STAT5 to growth factor and cytokine receptors such as EGFR and IL-6 receptor gp130 is critical to their activation and ability to contribute to malignant transformation. Therefore, interfering with these biochemical processes could lead to the discovery of novel anticancer agents. METHODS: Co-immunoprecipitation, western blotting, microscopy, DNA binding, invasion, and soft agar assays as well as a mouse model were used to investigate the mechanism by which the natural product Withacnistin (Wit) inhibits STAT 3/5 tyrosine phosphoryaltion and activation. RESULTS: Wit blocks EGF- and IL-6-stimulated binding of STAT3 and STAT5 to EGFR and gp130. Wit inhibits EGF-, PDGF-, IL-6-, IFNß-, and GM-CSF-stimulation of tyrosine phosphorylation of STAT3 and STAT5 but not of EGFR or PDGFR. The inhibition of P-STAT3 and P-STAT5 occurred rapidly, within minutes of Wit treatment and growth factor stimulation. Wit also inhibits STAT3 nuclear translocation, DNA binding, promoter transcriptional activation, and it suppresses the expression levels of STAT3 target genes such as Bcl-xL and Mcl-1. Finally, Wit induces apoptosis, inhibits anchorage-dependent and -independent growth and invasion, and causes breast tumour regression in an ErbB2-driven transgenic mouse model. CONCLUSIONS: These data warrant further development of Wit as a novel anticancer drug for targeting tumours that harbour hyperactivated STAT3 and STAT5.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Ergosterol/analogs & derivatives , Intercellular Signaling Peptides and Proteins/metabolism , Lactones/pharmacology , Receptors, Cytokine/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytokine Receptor gp130/metabolism , DNA-Binding Proteins , Epidermal Growth Factor/metabolism , Ergosterol/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interferon-beta/metabolism , Interleukin-6/metabolism , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , NIH 3T3 Cells , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , bcl-X Protein/metabolism
2.
Oncogene ; 33(5): 550-5, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-23396364

ABSTRACT

ROCK1 and ROCK2 mediate important processes such as cell migration, invasion and metastasis, making them good targets for the development of antitumor agents. Recently, using a fragment-based approach and X-ray crystallography, we reported on the design and synthesis of novel Rho-kinase inhibitors (RKIs). Here, we selected a pair of RKIs, the closely related structural analogs RKI-18 (potent; IC50 values of 397 nM (ROCK1) and 349 nM (ROCK2)) and RKI-11 (weak/inactive; IC50 values of 38 µM (ROCK1) and 45 µM (ROCK2)), as chemical probes and determined their effects on cytoskeleton organization, signaling, apoptosis, anchorage-dependent and independent growth, migration and invasion. RKI-18 but not RKI-11 suppresses potently the phosphorylation of the ROCK substrate myosin light chain 2 (MLC2) in intact human breast, lung, colon and prostate cancer cells. Furthermore, RKI-18 is highly selective at decreasing the levels of P-MLC2 over those of P-Akt, P-S6 and P-Erk ½. RKI-18 suppresses ROCK-mediated actin fiber formation, following stimulation with LPA as well as p21-activated kinase (PAK)-mediated lamellipodia and filopodia formation following bradykinin or PDGF stimulation. Furthermore, RKI-18 but not RKI-11 inhibits migration, invasion and anchorage-independent growth of human breast cancer cells. The fact that the active ROCK inhibitor RKI-18, but not the inactive closely related structural analog RKI-11 is effective at suppressing malignant transformation suggests that inhibition of ROCK with RKI-18 results in preventing migration, invasion and anchorage-independent growth. The potential of this class of RKIs as anti-tumor agents warrants further advanced preclinical studies.


Subject(s)
Antineoplastic Agents/isolation & purification , Cell Movement/drug effects , Protein Kinase Inhibitors/isolation & purification , rho-Associated Kinases/antagonists & inhibitors , 3T3 Cells , Actins/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cardiac Myosins/metabolism , Cell Line, Tumor , Cytoskeleton/drug effects , Humans , Mice , Myosin Light Chains/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/enzymology , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , p21-Activated Kinases/metabolism , rho-Associated Kinases/metabolism
3.
Cell Death Differ ; 17(11): 1795-804, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20489726

ABSTRACT

Persistently hyperphosphorylated Akt contributes to human oncogenesis and resistance to therapy. Triciribine (TCN) phosphate (TCN-P), the active metabolite of the Akt phosphorylation inhibitor TCN, is in clinical trials, but the mechanism by which TCN-P inhibits Akt phosphorylation is unknown. Here we show that in vitro, TCN-P inhibits neither Akt activity nor the phosphorylation of Akt S473 and T308 by mammalian target of rapamycin or phosphoinositide-dependent kinase 1. However, in intact cells, TCN inhibits EGF-stimulated Akt recruitment to the plasma membrane and phosphorylation of Akt. Surface plasmon resonance shows that TCN, but not TCN, binds Akt-derived pleckstrin homology (PH) domain (K(D): 690 nM). Furthermore, nuclear magnetic resonance spectroscopy shows that TCN-P, but not TCN, binds to the PH domain in the vicinity of the PIP3-binding pocket. Finally, constitutively active Akt mutants, Akt1-T308D/S473D and myr-Akt1, but not the transforming mutant Akt1-E17K, are resistant to TCN and rescue from its inhibition of proliferation and induction of apoptosis. Thus, the results of our studies indicate that TCN-P binds to the PH domain of Akt and blocks its recruitment to the membrane, and that the subsequent inhibition of Akt phosphorylation contributes to TCN-P antiproliferative and proapoptotic activities, suggesting that this drug may be beneficial to patients whose tumors express persistently phosphorylated Akt.


Subject(s)
Acenaphthenes/metabolism , Acenaphthenes/pharmacology , Cell Membrane/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribonucleotides/metabolism , Ribonucleotides/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Apoptosis , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Fluorescent Antibody Technique , Gene Amplification , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Magnetic Resonance Spectroscopy , Membrane Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation/drug effects , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction , Surface Plasmon Resonance , TOR Serine-Threonine Kinases/metabolism
4.
Oncogene ; 26(26): 3777-88, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17260025

ABSTRACT

A chemical biology approach identifies a beta 2 adrenergic receptor (beta2AR) agonist ARA-211 (Pirbuterol), which causes apoptosis and human tumor regression in animal models. beta2AR stimulation of cAMP formation and protein kinase A (PKA) activation leads to Raf-1 (but not B-Raf) kinase inactivation, inhibition of Mek-1 kinase and decreased phospho-extracellular signal-regulated kinase (Erk)1/2 levels. ARA-211 inhibition of the Raf/Mek/Erk1/2 pathway is mediated by PKA and not exchange protein activated by cAMP (EPAC). ARA-211 is selective and suppresses P-Erk1/2 but not P-JNK, P-p38, P-Akt or P-STAT3 levels. beta2AR stimulation results in inhibition of anchorage-dependent and -independent growth, induction of apoptosis in vitro and tumor regression in vivo. beta2AR antagonists and constitutively active Mek-1 rescue from the effects of ARA-211, demonstrating that beta2AR stimulation and Mek kinase inhibition are required for ARA-211 antitumor activity. Furthermore, suppression of growth occurs only in human tumors where ARA-211 induces cAMP formation and decreases P-Erk1/2 levels. Thus, beta2AR stimulation results in significant suppression of malignant transformation in cancers where it blocks the Raf-1/Mek-1/Erk1/2 pathway by a cAMP-dependent activation of PKA but not EPAC.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Ethanolamines/pharmacology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Signal Transduction/drug effects , Adrenergic beta-2 Receptor Agonists , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Female , Guanine Nucleotide Exchange Factors/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Humans , Immunohistochemistry , In Situ Nick-End Labeling , MAP Kinase Kinase 1/drug effects , Mice , Mice, Nude , Proto-Oncogene Proteins c-raf/drug effects , Receptors, Adrenergic, beta-2/drug effects
5.
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
6.
Leukemia ; 17(2): 451-7, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12592346

ABSTRACT

Mutations of the ras gene are among the most commonly identified transforming events in human cancers, including multiple myeloma. Farnesyltransferase inhibitors (FTI) were developed to prevent Ras processing and induce cancer cell death. Several FTIs are in phase II and one is in phase III clinical trials. Preclinically, most of the focus has been on solid tumors, and the effects of FTIs in multiple myeloma have not been investigated. In this study we examined the cytotoxic activity and inhibition of Ras processing in three myeloma cell lines with differing Ras mutation status. H929 cells with activated N-Ras were more sensitive to FTI-277 treatment than 8226 and U266 cells with activated K-Ras or wild-type Ras, respectively. A combination of FTI-277 and a geranylgeranyltransferase I inhibitor (GGTI)-2166 inhibited K-Ras processing and enhanced cell death in 8226 cells. U266 cells and Bcl-x(L) transfectants were equally sensitive to FTI-277 treatment. Similarly, 8226 cells selected for resistance to various chemotherapeutic agents, which resulted in either P-glycoprotein overexpression, altered topoisomerase II activity, or elevated glutathione levels, were equally sensitive to FTI-277. These preclinical studies suggest that prenylation inhibitors may represent new therapeutic agents for the treatment of refractory or drug-resistant multiple myeloma.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Apoptosis/drug effects , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Methionine/analogs & derivatives , Methionine/pharmacology , Cell Survival/drug effects , Drug Resistance, Neoplasm , Farnesyltranstransferase , Genes, ras/drug effects , Humans , Multiple Myeloma/genetics , Tumor Cells, Cultured , Tumor Stem Cell Assay
7.
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
8.
Arch Biochem Biophys ; 395(1): 103-12, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11673871

ABSTRACT

It is possible that many of the fibrogenic effects of transforming growth factor-beta (TGF-beta) are mediated by connective tissue growth factor (CTGF). In the present work, we show that TGF-beta1 produces a 5- to 6-fold increase in CTGF expression by cultured human lung fibroblasts that is due mainly to increased transcription. The half-life of CTGF mRNA is 1.96 h, consistent with its role as a cytokine. In addition to requiring Smad activity, based upon the effects of specific inhibitors, the TGF-beta intracellular signaling pathway requires the activity of a phosphatidylcholine-specific phospholipase C, a protein kinase C, and one or more tyrosine kinases. It is also likely that the pathway requires a member of the Ras superfamily of small GTPases, but not trimeric G proteins. Pharmacologic inhibition of TGF-beta stimulation of CTGF expression may be an effective therapeutic approach to a variety of undesirable fibrotic reactions.


Subject(s)
Fibroblasts/metabolism , Growth Substances/biosynthesis , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins , Lung/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/pharmacology , Cells, Cultured , Cholera Toxin/pharmacology , Connective Tissue Growth Factor , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Growth Substances/genetics , Humans , Immediate-Early Proteins/genetics , Lung/cytology , Lung/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Prenylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , Signal Transduction/drug effects , Trans-Activators/metabolism , Transforming Growth Factor beta1 , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Virulence Factors, Bordetella/pharmacology , ras Proteins/metabolism
9.
Oncogene ; 20(45): 6531-7, 2001 Oct 04.
Article in English | MEDLINE | ID: mdl-11641777

ABSTRACT

While both nitric oxide synthase-2 (NOS-2) and low molecular weight GTPases, such as Ras and Rho, have been implicated in malignant transformation, the cross talk between these important proteins is ill understood. In this study we examined the ability of H-Ras, RhoA, RhoB and Rac1 to modulate cytokine-induced NOS2. In the normal human liver AKN-1 cell line and in the human non-small cell lung carcinoma cell line, A-549, the ability of the cytokines (INF-gamma, IL-1beta and TNF-alpha) to activate NOS-2 was blocked by activated L61-H-Ras whereas dominant negative N17-H-Ras enhanced NOS-2 activation. Consistent with this dominant negative Erk2 as well as a MEK inhibitor also enhanced cytokine activation of NOS-2. Furthermore, activated L63-RhoA blocked whereas activated V14-RhoB enhanced cytokine NOS-2 activation. Activated I115-Racl did not affect NOS-2 activation. These results demonstrate that the Ras/Erk and the Ras/RhoA pathways negatively regulate whereas RhoB enhances cytokine-induced NOS-2. This is the first demonstration that genes that promote malignant transformation such as Ras and RhoA inhibit, whereas genes with tumor suppressor activity such as RhoB enhance NOS2 induction.


Subject(s)
Cytokines/pharmacology , Liver/metabolism , Lung Neoplasms/genetics , Nitric Oxide Synthase/genetics , Proto-Oncogene Proteins p21(ras)/physiology , rho GTP-Binding Proteins/physiology , Cell Line , Genes, Reporter , Humans , Liver/drug effects , Lung Neoplasms/enzymology , Mitogen-Activated Protein Kinase Kinases/physiology , Mitogen-Activated Protein Kinases/physiology , Mutation , Nitric Oxide Synthase Type II , Proto-Oncogene Proteins p21(ras)/genetics , Transcriptional Activation , Transfection , Tumor Cells, Cultured , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/physiology , rhoB GTP-Binding Protein/physiology
10.
Am J Physiol Lung Cell Mol Physiol ; 281(4): L816-23, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11557585

ABSTRACT

Interleukin (IL)-1beta is an important early mediator of inflammation in pulmonary artery smooth muscle cells. We previously reported that a geranylgeranyltransferase inhibitor elevated basal levels of inducible nitric oxide synthase (iNOS) and enhanced IL-1beta-mediated induction, suggesting that Rac or Rho small G proteins are candidates for antagonism of such induction. In this study, overexpression of constitutively active Rac1 or its dominant negative mutant did not affect IL-1beta induction of iNOS. Alternatively, treatment with Clostridium botulinum C3 exoenzyme, which ADP-ribosylates Rho, was associated with superinduction of iNOS, suggesting an inhibitory role for Rho. IL-1beta activated the three mitogen-activated protein kinase (extracellular signal-regulated kinases 1 and 2, c-Jun NH2-terminal kinase/stress-activated protein kinase, and p38) and the Janus kinase (JAK)-signal transducer and activator of transcription pathways. The former two pathways were not associated with IL-1beta-mediated iNOS induction, whereas the latter two appeared to have inhibitory roles in iNOS expression. These data suggest that a broad intracellular signaling response to IL-1beta in rat pulmonary artery smooth muscle cells results in elevated levels of iNOS that is opposed by the geranylgeranylated small G protein Rho as well as the p38 and JAK2 pathways.


Subject(s)
Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System/physiology , Muscle, Smooth, Vascular/enzymology , Nitric Oxide Synthase/metabolism , Pulmonary Artery/cytology , Animals , Botulinum Toxins/pharmacology , Cells, Cultured , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , MAP Kinase Kinase 4 , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Nitric Oxide Synthase Type II , Phosphotransferases (Phosphate Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor , Trans-Activators/metabolism , p38 Mitogen-Activated Protein Kinases , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
11.
Bioorg Med Chem Lett ; 11(6): 761-4, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11277514

ABSTRACT

Malaria continues to represent a very serious health problem in the tropics. The current methods of clinical treatment are showing deficiencies due to the increased incidence of resistance in the parasite. In the present paper we report the design, synthesis, and evaluation of potential antimalarial agents against a novel target, protein farnesyltransferase. We show that the most potent compounds are active against Plasmodium falciparum in vitro at submicromolar concentrations.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Plasmodium falciparum/drug effects , Alkyl and Aryl Transferases/metabolism , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Drug Design , Drug Resistance , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Imidazoles/chemical synthesis , Imidazoles/chemistry , Inhibitory Concentration 50 , Parasitic Sensitivity Tests , Structure-Activity Relationship
12.
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
13.
J Biol Chem ; 276(19): 16161-7, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11154688

ABSTRACT

Even though farnesyltransferase inhibitors (FTIs), a novel class of therapeutic agents presently in clinical trials, have preclinically outstanding anticancer activity and impressive lack of toxicity, their mechanism of action is not well understood. To enhance our understanding of how FTIs inhibit the growth of tumors, we have investigated their effects on cell cycle progression of two human lung cancer cell lines, A-549 and Calu-1. In this report, we show in synchronized A-549 and Calu-1 cells that FTI-2153 treatment resulted in a large accumulation of cells in the mitosis phase of the cell division cycle, with some cells in the G(0)/G(1) phase. Furthermore, microtubule immunostaining and 4,6-diamidino-2-phenylindole DNA staining demonstrated that the FTI-2153-induced accumulation in mitosis is due to the inability of these cells to progress from prophase to metaphase. FTI-2153 inhibited the ability of A-549 and Calu-1 cells to form bipolar spindles and caused formation of monoasteral spindles. Furthermore, FTI-2153 induced a ring-shaped chromosome morphology and inhibited chromosome alignment. Time-lapse videomicroscopy confirmed this result by showing that FTI-2153-treated cells are unable to align their chromosomes at the metaphase plate. FTI-2153 did not affect the localization to the kinetochores of two farnesylated centromeric proteins, CENP-E and CENP-F. Thus, a mechanism by which FTIs inhibit progression through mitosis and tumor growth is by blocking bipolar spindle formation and chromosome alignment.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Cell Cycle/drug effects , Chromosomes, Human/drug effects , Enzyme Inhibitors/pharmacology , Spindle Apparatus/drug effects , Cell Division/drug effects , Chromosomes, Human/physiology , Chromosomes, Human/ultrastructure , Farnesyltranstransferase , Humans , Lung Neoplasms , Metaphase/drug effects , Mitosis/drug effects , Spindle Apparatus/ultrastructure , Tumor Cells, Cultured
14.
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
15.
Oncogene ; 19(47): 5338-47, 2000 Nov 09.
Article in English | MEDLINE | ID: mdl-11103935

ABSTRACT

Upregulation of the cyclin-dependent kinase inhibitor p21WAF1/CIP1 and subsequent cell growth arrest or senescence is one mechanism by which normal cells are believed to respond to stress induced by the constitutively activated GTPase Ras. We hypothesize that in the absence of p21, the onset of Ras-dependent oncogenesis is accelerated. To test this hypothesis, we crossed MMTV/v-Ha-ras transgenic mice into a p21-deficient background. By 63 days of age, all 8 ras/p21-/- mice developed either malignant (mammary and/or salivary adenocarcinomas) or benign (Harderian hyperplasia) tumors. In contrast, by the same age, only one out of nine of the ras/p21+/+ mice developed a tumor. Furthermore, by 94 days of age, half of the ras/p21-/- mice, but none of the ras/p21+/+ mice, developed mammary tumors. p21-deficiency also accelerated the development of salivary (T50=66 days for ras/p21-/- vs T50=136 days for ras/p21+/+) and Harderian (T50=52 days for ras/p21-/- vs T50>221 days for ras/p21+/+) tumors. Furthermore, two out of the eight ras/p21-/- mice had metastatic lesions, one in its lungs, the other in its abdomen. None of the nine ras/p21+/+ mice had metastatic lesions. By 4 months of age, the mammary tumor multiplicity was 10-fold greater in ras/p21-/- (average 3.40 tumors/mouse) than in ras/p21+/+ (average 0.33 tumor/mouse) mice. However, once the tumors appeared, their growth rate, apoptosis level, and mitotic index were not affected by the loss of p21, suggesting that loss of p21 is critical in early but not late events of Ras oncogenesis. Altogether, the results show that tumor onset in MMTV/v-Ha-ras mice is p21-dependent with loss of p21 associated with earlier tumor appearance and increased tumor multiplicity and aggressiveness.


Subject(s)
Carcinoma, Ductal, Breast/physiopathology , Cyclins/physiology , Genes, ras/physiology , Mammary Neoplasms, Animal/physiopathology , Oncogene Protein p21(ras)/physiology , Adenocarcinoma/etiology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Carcinoma, Ductal, Breast/etiology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Disease Models, Animal , Female , Gene Expression , Male , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Mice, Transgenic , Oncogene Protein p21(ras)/genetics , Salivary Gland Neoplasms/etiology , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/physiopathology
16.
Mol Biol Cell ; 11(11): 3835-48, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11071910

ABSTRACT

Colony-stimulating factor-1 (CSF-1) induces expression of immediate early gene, such as c-myc and c-fos and delayed early genes such as D-type cyclins (D1 and D2), whose products play essential roles in the G1 to S phase transition of the cell cycle. Little is known, however, about the cytoplasmic signal transduction pathways that connect the surface CSF-1 receptor to these genes in the nucleus. We have investigated the signaling mechanism of CSF-1-induced D2 expression. Analyses of CSF-1 receptor autophosphorylation mutants show that, although certain individual mutation has a partial inhibitory effect, only multiple combined mutations completely block induction of D2 in response to CSF-1. We report that at least three parallel pathways, the Src pathway, the MAPK/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway, and the c-myc pathway, are involved. Induction of D2 is partially inhibited in Src(-/-) bone marrow-derived macrophages and by Src inhibitor PP1 and is enhanced in v-Src-overexpressing cells. Activation of myc's transactivating activity selectively induces D2 but not D1. Blockade of c-myc expression partially blocks CSF-1-induced D2 expression. Complete inhibition of the MEK/ERK pathway causes 50% decrease of D2 expression. Finally, simultaneous inhibition of Src, MEK activation, and c-myc expression additively blocks CSF-1-induced D2 expression. This study indicates that multiple signaling pathways are involved in full induction of a single gene, and this finding may also apply broadly to other growth factor-inducible genes.


Subject(s)
Cyclins/genetics , MAP Kinase Kinase Kinase 1 , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Animals , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin D2 , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Genes, myc , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
17.
Expert Opin Investig Drugs ; 9(12): 2767-82, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11093352

ABSTRACT

The fact that proteins such as Ras, Rac and RhoA require farnesylation or geranylgeranylation to induce malignant transformation prompted many investigators to develop farnesyltransferase (FTase) and geranylgeranyltransferase I (GGTase I) inhibitors (FTIs and GGTIs, respectively) as novel anticancer drugs. Although FTIs have been shown to antagonise oncogenic signalling, reverse malignant transformation, inhibit human tumour growth in nude mice and induce tumour regression in transgenic mice without any signs of toxicity, their mechanism of action is not known. This review will focus on important mechanistic issues as well as bench to bedside translational issues. These will include the relevance to cancer therapy of the alternative geranylgeranylation of K-Ras when FTase is inhibited; a thorough discussion about evidence for and against the involvement of inhibition of prenylation of Ras and RhoB in the mechanism of FTIs' antitumour activity as well as effects of FTIs and GGTIs on the cell cycle machinery and the dynamics of bipolar spindle formation and chromosome alignment during mitosis. Bench to bedside issues relating to the design of hypothesis-driven clinical trials with biochemical correlates for proof-of-concept in man will also be discussed. This will include Phase I issues such as determining maximally tolerated dose (MTD) versus effective biological dose (EBD), as well as whether Phase II trials are still needed for clinical evaluations of anti-signalling agents. Other questions that will be addressed include: what levels of inhibition of FTase activity are required for tumour response in Phase II clinical evaluations? What FTase substrates are most relevant as biochemical correlates? Are signalling pathways such as H-Ras/PI3K/Akt and K-Ras/Raf/MEK/Erk significant biological readouts? Does Ras mutation status predict response? What are appropriate clinical end-points for FTI Phase II trials? For this latter important question, time to tumour progression, median survival, percentage of patients that progress, clinical benefits and improvement in quality of life will all be discussed.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Animals , Drug Design , Farnesyltranstransferase , Humans , Neoplasms/enzymology
18.
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
20.
Radiat Res ; 154(2): 125-32, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10931682

ABSTRACT

Successful radiosensitization requires that tumor cells become more radiosensitive without causing an equivalent reduction in the survival of cells of the surrounding normal tissues. Since tumor cell radiosensitivity can be influenced by RAS oncogene activation, we have hypothesized that inhibition of oncogenic RAS activity would lead to radiosensitization of tumors with activated RAS. We previously showed in tissue culture that prenyltransferase treatment of cells with activated RAS resulted in radiosensitization, whereas treatment of cells with wild-type RAS had no effect on radiation survival. Here we ask whether the findings obtained in vitro have applicability in vivo. We found that treatment of nude mice bearing T24 tumor cell xenografts with farnesyltransferase inhibitors resulted in a significant and synergistic reduction in tumor cell survival after irradiation. The regrowth of T24 tumors expressing activated RAS was also significantly prolonged by the addition of treatment with farnesyltransferase inhibitors compared to the regrowth after irradiation alone. In contrast, there was no effect on the radiosensitivity of HT-29 tumors expressing wild-type RAS. These results demonstrate that specific radiosensitization of tumors expressing activated RAS oncogenes can be obtained in vivo.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Colonic Neoplasms/radiotherapy , Enzyme Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology , Urinary Bladder Neoplasms/radiotherapy , Animals , Colonic Neoplasms/genetics , Farnesyltranstransferase , Gene Expression Regulation, Neoplastic/drug effects , Genes, ras/drug effects , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Nude , Neoplasm Recurrence, Local/pathology , Neoplasm Transplantation , Tumor Cells, Cultured , Tumor Stem Cell Assay , Urinary Bladder Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...