Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Int J Cancer ; 139(7): 1648-57, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27194579

ABSTRACT

The eukaryotic translation initiation factor 4E (eIF4E) is a potent oncogene that is found to be dysregulated in 30% of human cancer, including colorectal carcinogenesis (CRC). ISIS 183750 is a second-generation antisense oligonucleotide (ASO) designed to inhibit the production of the eIF4E protein. In preclinical studies we found that EIF4e ASOs reduced expression of EIF4e mRNA and inhibited proliferation of colorectal carcinoma cells. An additive antiproliferative effect was observed in combination with irinotecan. We then performed a clinical trial evaluating this combination in patients with refractory cancer. No dose-limiting toxicities were seen but based on pharmacokinetic data and tolerability the dose of irinotecan was reduced to 160 mg/m(2) biweekly. Efficacy was evaluated in 15 patients with irinotecan-refractory colorectal cancer. The median time of disease control was 22.1 weeks. After ISIS 183750 treatment, peripheral blood levels of eIF4E mRNA were decreased in 13 of 19 patients. Matched pre- and posttreatment tumor biopsies showed decreased eIF4E mRNA levels in five of nine patients. In tumor tissue, the intracellular and stromal presence of ISIS 183750 was detected by IHC in all biopsied patients. Although there were no objective responses stable disease was seen in seven of 15 (47%) patients who were progressing before study entry, six of whom were stable at the time of the week 16 CT scan. We were also able to confirm through mandatory pre- and posttherapy tumor biopsies penetration of the ASO into the site of metastasis.


Subject(s)
Camptothecin/analogs & derivatives , Colorectal Neoplasms/therapy , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Oligoribonucleotides/therapeutic use , Adult , Aged , Camptothecin/adverse effects , Camptothecin/blood , Camptothecin/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Combined Modality Therapy , Eukaryotic Initiation Factor-4E/genetics , Female , HCT116 Cells , Humans , Irinotecan , Male , Middle Aged , Oligonucleotides , Oligonucleotides, Antisense/genetics , Oligoribonucleotides/genetics , RNA, Messenger/blood , RNA, Messenger/genetics
2.
Ann Oncol ; 27(4): 693-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26802155

ABSTRACT

BACKGROUND: KRAS mutations in NSCLC are associated with a lack of response to epidermal growth factor receptor inhibitors. Selumetinib (AZD6244; ARRY-142886) is an oral selective MEK kinase inhibitor of the Ras/Raf/MEK/ERK pathway. PATIENTS AND METHODS: Advanced nonsmall-cell lung cancer (NSCLC) patients failing one to two prior regimens underwent KRAS profiling. KRAS wild-type patients were randomized to erlotinib (150 mg daily) or a combination of selumetinib (150 mg daily) with erlotinib (100 mg daily). KRAS mutant patients were randomized to selumetinib (75 mg b.i.d.) or the combination. The primary end points were progression-free survival (PFS) for the KRAS wild-type cohort and objective response rate (ORR) for the KRAS mutant cohort. Biomarker studies of ERK phosphorylation and immune subsets were carried out. RESULTS: From March 2010 to May 2013, 89 patients were screened; 41 KRAS mutant and 38 KRAS wild-type patients were enrolled. Median PFS in the KRAS wild-type arm was 2.4 months [95% confidence interval (CI) 1.3-3.7] for erlotinib alone and 2.1 months (95% CI 1.8-5.1) for the combination. The ORR in the KRAS mutant group was 0% (95% CI 0.0% to 33.6%) for selumetinib alone and 10% (95% CI 2.1% to 26.3%) for the combination. Combination therapy resulted in increased toxicities, requiring dose reductions (56%) and discontinuation (8%). Programmed cell death-1 expression on regulatory T cells (Tregs), Tim-3 on CD8+ T cells and Th17 levels were associated with PFS and overall survival in patients receiving selumetinib. CONCLUSIONS: This study failed to show improvement in ORR or PFS with combination therapy of selumetinib and erlotinib over monotherapy in KRAS mutant and KRAS wild-type advanced NSCLC. The association of immune subsets and immune checkpoint receptor expression with selumetinib may warrant further studies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Erlotinib Hydrochloride/administration & dosage , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Aged , Aged, 80 and over , Benzimidazoles/administration & dosage , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Female , Humans , MAP Kinase Kinase Kinase 1/genetics , Male , Middle Aged , Mutation , Protein Kinase Inhibitors/administration & dosage
3.
J Clin Invest ; 73(2): 587-92, 1984 Feb.
Article in English | MEDLINE | ID: mdl-6607934

ABSTRACT

B cell chronic lymphocytic leukemia (CLL) cells appear to be arrested in their differentiation so that little immunoglobulin is secreted in most cases. To determine their capacity for further differentiation we stimulated cells from a series of 10 cases of CLL with a phorbol ester and assayed for production of immunoglobulin protein, accumulation of immunoglobulin mRNA, and alterations in cell surface markers. We found that cells from all cases were induced to secret monoclonal immunoglobulin of the same heavy and light chain type as the surface membrane immunoglobulin type. Immunoglobulin secretion was preceded by a rapid increase in the levels of mRNA coding for IgM, predominantly the secretory form, mu s-mRNA, rather than the membrane form, mu m-mRNA. A similar selection of mu s- over mu m-mRNA is known to occur in plasma cells by a mechanism of differential processing of mRNA from a single mu-chain gene. Except for a decline in the expression of surface IgD, cell surface determinants remained unaffected both in terms of the percentage of positive cells and the relative number of sites per cell. In contrast to previous studies, these results indicate that CLL cells consistently retain the capacity to further differentiate toward plasma cells and secrete immunoglobulin. The immunoglobulin secretion is mediated, at least in part, by a developmentally regulated increment in mu s-mRNA.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulins/genetics , Leukemia, Lymphoid/immunology , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Immunoglobulin M/genetics , Immunoglobulins/metabolism , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/pharmacology
4.
J Clin Invest ; 89(1): 191-6, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1309535

ABSTRACT

To develop a new approach to the treatment of advanced, hormone-refractory prostate cancer, the signal transductions regulating the growth of human androgen-independent prostate carcinoma cell lines were studied. Agonist-stimulated Ca2+ mobilization, a critical regulatory event in other secretory cell types, was studied as a means of identifying previously undescribed plasma membrane receptors that may transduce a growth inhibitory signal. In all of the cell lines tested, P2-purinergic receptor agonists, including ATP and certain hydrolysis-resistant adenine nucleotides, induced a rapid, transient increase in cytoplasmic free Ca2+ that was detectable at 50 to 100 nM ATP, was maximal at 100 microM ATP, and was inhibited approximately 50% by chelation of extracellular Ca2+. Within 8 s after addition, ATP stimulated accumulation of the polyphosphatidylinositol products inositol (1, 4, 5) trisphosphate, inositol (1, 3, 4) trisphosphate, and inositol tetrakisphosphate. In addition to stimulating phosphatidylinositol turnover and Ca2+ mobilization, ATP and hydrolysis-resistant ATP analogues induced greater than 90% inhibition of the growth of all lines tested. These data demonstrate that human androgen-independent prostate carcinoma cells express functional P2-purinergic receptors linked to phospholipase C, and that agonists of this receptor are markedly growth inhibitory, suggesting a novel therapeutic approach to this common adult neoplasm.


Subject(s)
Androgens/metabolism , Carcinoma/metabolism , Prostatic Neoplasms/metabolism , Receptors, Purinergic/metabolism , Adenine Nucleotides/pharmacology , Calcium/metabolism , Calcium Channels/metabolism , Evaluation Studies as Topic , Humans , Inositol Phosphates/metabolism , Male , Phosphatidylinositols/metabolism , Signal Transduction/physiology , Tumor Cells, Cultured , Type C Phospholipases/metabolism
5.
Mol Cell Biol ; 6(5): 1847-50, 1986 May.
Article in English | MEDLINE | ID: mdl-3466024

ABSTRACT

The phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) induced megakaryoblastic differentiation and c-sis expression in the human hematopoietic stem cell line K-562. This induction occurred at the transcriptional level, as determined by a nuclear runoff transcriptional assay, and was not a generalized effect of TPA, since the treatment of other hematopoietic cell lines and normal peripheral blood lymphocytes with TPA did not result in the appearance of c-sis mRNA.


Subject(s)
Oncogenes/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Cell Differentiation/drug effects , Cell Line , Hematopoietic Stem Cells/cytology , Humans , Leukemia, Myeloid , RNA, Messenger/genetics
6.
Mol Cell Biol ; 6(12): 4244-50, 1986 Dec.
Article in English | MEDLINE | ID: mdl-2432398

ABSTRACT

Transferrin receptor expression is essential for the proliferation of both normal and malignant T cells. While transferrin receptor expression in normal T cells is tightly coupled to interleukin-2 receptor expression, transferrin receptor expression in malignant cells is usually constitutive and is released from this constraint. Temporally, the appearance of these membrane receptors is preceded by changes in the expression of the proto-oncogenes c-myc and c-myb. In addition, although an increase in the level of intracellular free calcium occurs early in the sequence of T-cell activation, the activation events dependent on this calcium flux have not been resolved. In the present study we report that diltiazem, an ion channel-blocking agent that inhibits calcium influx, arrested the growth in vitro of both normal and malignant human T cells in the G1 phase of the cell cycle. However, diltiazem did not inhibit the expression of c-myc or interleukin-2 receptor mRNA and protein in normal mitogen-activated T cells or the constitutive expression of c-myc and c-myb mRNA in malignant T cells (T acute lymphoblastic leukemia cells). In contrast, diltiazem prevented the induction of transferrin receptor (mRNA and protein) in normal T cells and caused a progressive loss of transferrin receptor (mRNA and protein) in malignant T cells. These data demonstrate that diltiazem can dissociate several growth-related processes normally occurring in G1 and thereby disrupt the biochemical cascade leading to cell proliferation.


Subject(s)
Cell Transformation, Neoplastic , Diltiazem/pharmacology , Receptors, Transferrin/genetics , T-Lymphocytes/metabolism , Cell Cycle/drug effects , Cell Line , DNA Replication/drug effects , Humans , Interleukin-2/pharmacology , RNA/isolation & purification , Receptors, Transferrin/biosynthesis , Receptors, Transferrin/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
7.
Mol Cell Biol ; 7(7): 2644-8, 1987 Jul.
Article in English | MEDLINE | ID: mdl-3039354

ABSTRACT

Treatment of HL-60 cells with dibutyryl cyclic AMP induced rapid transcriptional inactivation of c-myc and the transferrin receptor. Transcriptional inactivation was followed by loss of c-myc and transferrin receptor mRNA and protein. Treated cells completed one round of proliferation, followed by growth arrest, G1 synchronization, and monocytic differentiation. These data suggest that cyclic AMP-mediated control of growth and differentiation may be achieved, at least in part, by transcriptional regulation of certain growth-associated proto-oncogenes and growth factor receptor genes.


Subject(s)
Bucladesine/pharmacology , Proto-Oncogenes/drug effects , Receptors, Transferrin/genetics , Transcription, Genetic/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc , Receptors, Transferrin/biosynthesis , Receptors, Transferrin/drug effects
8.
J Natl Cancer Inst ; 84(9): 711-6, 1992 May 06.
Article in English | MEDLINE | ID: mdl-1349044

ABSTRACT

BACKGROUND: Multidrug resistance (MDR) mediated by high levels of mdr-1 (also known as PGY1)/P-glycoprotein (Pgp) has been studied in tissue culture systems; however, most tumor samples which express mdr-1/Pgp have much lower levels. PURPOSE: We wanted to determine if levels seen clinically could be detected by commonly used methods and to determine if these levels conferred MDR reversible by Pgp antagonists. METHODS: We studied multi-drug-resistant cell lines and sublines with levels of mdr-1/Pgp expression comparable to those seen clinically. We evaluated the expression of mdr-1 RNA by Northern blot analysis, slot blot analysis, polymerase chain reaction (PCR) analysis, and in situ hybridization. We evaluated protein expression by immunofluorescence, immunohistochemistry, fluorescence-activated cell sorting, and immunoblotting analyses. Drug resistance and reversibility were determined by cell growth during continuous drug exposure. RESULTS: In most cases, the low level of mdr-1/Pgp present in these cell lines could be detected by each method, but the assays were at the limit of sensitivity for all methods except the PCR method. These low levels of mdr-1/Pgp are capable of conferring MDR, which can be antagonized by verapamil. CONCLUSIONS: Levels of mdr-1/Pgp similar to those found in clinical samples can be detected by each of these methods, but the PCR method was the most sensitive and most reliably quantitative. IMPLICATIONS: In vitro sensitization by the addition of verapamil in cell lines with these low levels of mdr-1/Pgp suggests that clinically detected levels may confer drug resistance in vivo.


Subject(s)
Colonic Neoplasms/physiopathology , Drug Resistance , Membrane Glycoproteins/analysis , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Flow Cytometry , Gene Expression , Humans , Immunohistochemistry , In Vitro Techniques , Membrane Glycoproteins/genetics , Nucleic Acid Hybridization , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Tumor Cells, Cultured
9.
Cancer Res ; 50(17): 5257-62, 1990 Sep 01.
Article in English | MEDLINE | ID: mdl-2167151

ABSTRACT

In [3H]inositol-labeled membranes prepared from Swiss mouse 3T3 and human small cell lung carcinoma cells, [Tyr4]-bombesin stimulated production of water-soluble inositol phosphates. The reaction was stimulated by guanosine 5'-O-[3-thiotriphosphate] and was specifically inhibited by both [Leu13-psi-CH2NHLeu14]-bombesin and the antibombesin antibody 2A11. [Tyr4]-bombesin-induced activation of phospholipase C is most apparent in Ca2(+)-depleted conditions (less than 1 microM[Ca2+]free). The kinetics of activation by ligand also demonstrate that [Tyr4]-bombesin-dependent phospholipase C activation is most apparent at [Mg2+]free of approximately 0.2 microM. At millimolar concentrations of [Mg2+]free, there is considerably less dependence on [Tyr4]-bombesin for activation of phospholipase C. ATP is not necessary for initial activation of phospholipase C, and beta, gamma-imidoadenosine-5'-triphosphate does not inhibit the reaction. These results demonstrate that in these cell types [Tyr4]-bombesin activates phospholipase C in conjunction with guanine nucleotides. Phospholipase C-coupled guanine nucleotide regulatory proteins would be appropriately considered as novel targets for the development of therapeutic strategies in small cell lung carcinoma.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Bombesin/pharmacology , Guanine Nucleotides/pharmacology , Guanosine Diphosphate/pharmacology , Guanosine Triphosphate/analogs & derivatives , Thionucleotides/pharmacology , Tumor Cells, Cultured/enzymology , Type C Phospholipases/metabolism , Animals , Antibodies , Bombesin/immunology , Calcium/pharmacology , Carcinoma, Small Cell , Cell Line , Cell Membrane/enzymology , Guanosine 5'-O-(3-Thiotriphosphate) , Guanosine Diphosphate/analogs & derivatives , Guanosine Triphosphate/pharmacology , Humans , Kinetics , Lung Neoplasms , Magnesium/pharmacology , Mice , Tumor Cells, Cultured/drug effects
10.
Cancer Res ; 55(10): 2122-8, 1995 May 15.
Article in English | MEDLINE | ID: mdl-7743512

ABSTRACT

The propensity of a cell to undergo apoptosis has been proposed to be a determinant for chemotherapy sensitivity that is not directly dependent on specific drug-target interactions. Androgen-independent prostate cancer is typically refractory to cytotoxic drugs, and we tested whether this is due to a loss of the ability to undergo apoptosis. Exposure of the hormone-insensitive and p53-negative human prostate carcinoma cell line PC-3 to 22 microM cisplatin, 1 microM camptothecin, 10 microM tenoposide, 135 nM vincristine, or 10 microM lovastatin for 72 h caused cell death, internucleosomal DNA fragmentation, and morphological changes typical for apoptosis. One microM cycloheximide prevented anticancer drug-induced apoptosis, whereas high concentration (1 mM) of cycloheximide alone induced apoptosis, indicating that protein synthesis was not needed for these cells to undergo apoptosis. Since cycloheximide affected DNA synthesis and proliferation of PC-3 cells, we tested whether the DNA polymerase inhibitor aphidicolin could also suppress drug-induced apoptosis. In contrast to cycloheximide, aphidicolin inhibited only vincristine-induced apoptosis. Cycloheximide prevented drug-induced changes in cell cycle distribution except for vincristine, while aphidicolin led to an accumulation of cells at the G1-S border independent of the drug used. These data indicate that macromolecular synthesis, active cell cycling, and p53 expression are not required for apoptosis to proceed in this system.


Subject(s)
Antineoplastic Agents/pharmacology , Aphidicolin/pharmacology , Apoptosis/drug effects , Cycloheximide/pharmacology , DNA, Neoplasm/drug effects , Prostatic Neoplasms , Cell Cycle/drug effects , Cell Division/drug effects , DNA, Neoplasm/biosynthesis , Humans , Lovastatin/pharmacology , Male , Microscopy, Electron , Nucleosomes , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/ultrastructure , Tumor Cells, Cultured
11.
Cancer Res ; 61(3): 931-4, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11221885

ABSTRACT

Transcriptional repression of the transforming growth factor (TGF)-1P type II receptor (TPRII) gene appears to be a major mechanism to inactivate TGF-beta responsiveness in many human cancers. Because histone acetylation/deacetylation plays a role in transcriptional regulation, we have examined the effect of MS-275, a synthetic inhibitor of histone deacetylase, in human breast cancer cell lines. MS-275 showed antiproliferative activity against all human breast cancer cell lines examined and induced TbetaRII mRNA, but not TGF-beta type I receptor mRNA. MS-275 caused an accumulation of acetylated histones H3 and H4 in total cellular chromatin. An increase in the accumulation of acetylated histones H3 and H4 was detected in the TbetaRII promoter after treatment with MS-275. However, the level of histone acetylation did not change in chromatin associated with the TGF-beta type I receptor gene. MS-275 treatment enhanced TGF-beta1-induced plasminogen activator inhibitor 1 expression. Thus, antitumor activity of MS-275 may be mediated in part through the induction of TbetaRII expression and consequent potentiation of TGF-beta signaling.


Subject(s)
Benzamides/pharmacology , Breast Neoplasms/metabolism , Pyridines/pharmacology , Receptors, Transforming Growth Factor beta/biosynthesis , Acetylation/drug effects , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Division/drug effects , Chromatin/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Growth Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/drug effects , Histones/metabolism , Humans , Plasminogen Activator Inhibitor 1/biosynthesis , Protein Serine-Threonine Kinases , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/drug effects , Transforming Growth Factor beta/physiology , Tumor Cells, Cultured
12.
Cancer Res ; 50(9): 2773-80, 1990 May 01.
Article in English | MEDLINE | ID: mdl-2158397

ABSTRACT

We report the establishment and characterization of four continuous cell lines derived from human primary and metastatic gastric carcinomas, and we compare their properties with a panel of colorectal carcinoma cell lines previously established and reported by us. Our success rate in culturing gastric carcinomas was relatively low, especially from primary tumors, compared to colorectal carcinoma. These observations may reflect the relatively modest number of gastric carcinoma cell lines established (mainly from Japan), compared to the abundance of colorectal carcinoma lines established worldwide. All four gastric lines expressed the surface glycoproteins carcinoembryonic antigen and TAG-72 and three lines expressed CA 19-9. Two of the lines expressed aromatic amino acid decarboxylase but lacked other markers for neuroendocrine differentiation. All four lines were positive for vasoactive intestinal peptide receptors but lacked gastrin receptors. In addition, two lines expressed receptors for muscarinic/cholinergic receptors but not beta-adrenergic receptors. Cytogenetic evidence for gene amplification was present in the cell lines. All four lines contained varying numbers of double-minute chromosomes. One line, SNU-16, was amplified for the c-myc proto-oncogene and contained four homogeneously staining regions. While c-myc and c-erb-B-2 RNA were expressed by all lines, there was no evidence of amplification or overexpression of several other proto-oncogenes and growth factors. The multiple properties we have described in our gastric carcinoma cell lines are remarkably similar to those found in the panel of colorectal carcinoma cell lines. These properties include morphology, growth characteristics, expression of surface glycoproteins, partial expression of neuroendocrine cell markers, frequent chromosomal evidence of gene amplification, and occasional amplification of the c-myc proto-oncogene. Our four well characterized cell lines should provide useful additions to the modest number currently available for in vitro studies of gastric carcinoma.


Subject(s)
Stomach Neoplasms/pathology , Antigens, Neoplasm/analysis , Cell Line , Chromosome Aberrations , Gene Amplification , Glycoproteins/analysis , Humans , Proto-Oncogene Mas , Proto-Oncogenes , Receptors, Gastrointestinal Hormone/analysis , Receptors, Vasoactive Intestinal Peptide , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Tumor Cells, Cultured
13.
Oncogene ; 18(47): 6460-8, 1999 Nov 11.
Article in English | MEDLINE | ID: mdl-10597248

ABSTRACT

BRCA1, a tumor suppressor protein implicated in hereditary forms of breast and ovarian cancer, is transcriptionally regulated in a proliferation-dependent manner. In this study, we demonstrate a substantial role for proteolysis in regulating the BRCA1 steady-state protein level in several cell lines. N-acetyl-leu-leu-norleucinal (ALLN), an inhibitor of the proteasome, calpain, and cathepsins, caused BRCA1 protein to accumulate in the nucleus of several human breast, prostate, and melanoma cell lines which express low or undetectable basal levels of BRCA1 protein, but not in cells with high basal expression of BRCA1. Protease inhibition did not increase BRCA1 synthesis, nor change its mRNA level, but it dramatically prolonged the protein's half-life. In contrast to ALLN, lactacystin and PS341, two specific proteasome inhibitors, as well as calpastatin peptide and PD150606, two selective calpain inhibitors, had no effect on BRCA1 stability, whereas ALLM, an effective calpain and cathepsin inhibitor but weak proteasome inhibitor, did stimulate accumulation of BRCA1. Moreover, three inhibitors of acidic cysteine proteases, chloroquine, ammonium chloride and bafilomycin, were as effective as ALLN. These results demonstrate that degradation by a cathepsin-like protease in fine balance with BRCA1 transcription is responsible for maintaining the low steady-state level of BRCA1 protein seen in many cancer cells.


Subject(s)
BRCA1 Protein/genetics , Gene Expression Regulation , Karyopherins , Receptors, Cytoplasmic and Nuclear , BRCA1 Protein/biosynthesis , BRCA1 Protein/metabolism , Calpain/metabolism , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cysteine Endopeptidases/metabolism , Dactinomycin/antagonists & inhibitors , Down-Regulation/drug effects , Endopeptidases/metabolism , Half-Life , Humans , Hydrolysis , Leupeptins/pharmacology , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Exportin 1 Protein
14.
Oncogene ; 20(46): 6718-23, 2001 Oct 11.
Article in English | MEDLINE | ID: mdl-11709706

ABSTRACT

Androgen deprivation therapy for advanced prostate cancer is often effective, but not curative. Molecular pathways mediating the therapeutic response and those contributing to the subsequent hormone-refractory cell growth remain poorly understood. Here, cDNA microarray analysis of human CWR22 prostate cancer xenografts during the course of androgen deprivation therapy revealed distinct global gene expression profiles in primary, regressing and recurrent tumors. Elucidation of the genes involved in the transition between these states implicated specific molecular mechanisms in therapy failure and tumor progression. First, we identified a set of androgen-responsive genes whose expression decreased during the therapy response, but was then systematically restored in the recurrent tumors. In addition, altered expression of genes that encode known targets of rapamycin or that converge on the PI3K/AKT/FRAP pathway was observed in the recurrent tumors. Further suggestion for the involvement of these genes in hormone-refractory prostate cancer came from the observation that cells established from the recurrent xenografts were strongly inhibited in vitro by rapamycin. The results of this functional genomic analysis suggest that the combined effect of re-expression of androgen-responsive genes as well as the activation of rapamycin-sensitive signaling may drive prostate cancer progression, and contribute to the failure of androgen-deprivation therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Drug Resistance, Neoplasm , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Sirolimus/pharmacology , Algorithms , Animals , Cell Survival/drug effects , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Nude , Models, Biological , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Signal Transduction , Software , Time Factors , Tumor Cells, Cultured
15.
Oncogene ; 19(52): 6082-90, 2000 Dec 07.
Article in English | MEDLINE | ID: mdl-11146561

ABSTRACT

To develop a new approach to the treatment of primitive neuroectodermal tumors we evaluated the effect of the HMG-CoA reductase inhibitor lovastatin on the Ewing's sarcoma cell line CHP-100. Lovastatin induced neural morphology and markers including neuron-specific enolase and neurofilament protein. The acquisition of neural morphology required new mRNA synthesis, and cDNA microarray analysis confirmed that lovastatin altered the program of gene expression. After morphologic differentiation the cells underwent rapidly progressive apoptosis. In normal development of neuronal progenitors, differentiation signals trigger p21WAF1 accumulation, RB hypophosphorylation, enhanced RB-E2F-1 association, and G1 arrest, and these events have been shown to protect from apoptosis. In contrast, in the Ewing's sarcoma cells lovastatin triggered differentiation without causing cell cycle arrest: p21WAF1 was not induced, RB remained hyperphosphorylated, and RB protein expression and RB-E2F-1 association were markedly downregulated, suggesting that loss of an RB-regulated G1 checkpoint promoted apoptosis. Consistent with this hypothesis, adenoviral p21WAF1 decreased DNA synthesis and partially protected from lovastatin-induced cytotoxicity. The data demonstrate a new model for examining the genetic regulation of cell fate in a neural progenitor tumor and suggest a new approach to the treatment of this neoplasm.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins , Ectoderm/drug effects , G1 Phase/drug effects , Lovastatin/pharmacology , Neurons/drug effects , Retinoblastoma Protein/metabolism , Tumor Suppressor Proteins , Biomarkers/analysis , Cell Size/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , DNA/biosynthesis , Down-Regulation/drug effects , Ectoderm/pathology , Ectoderm/ultrastructure , Flow Cytometry , Genes, cdc , Humans , Immunohistochemistry , Microscopy, Electron, Scanning , Microtubule-Associated Proteins/metabolism , Mitotic Index , Neurons/pathology , Neurons/ultrastructure , Sarcoma, Ewing/pathology , Sarcoma, Ewing/ultrastructure , Tumor Cells, Cultured
16.
Oncogene ; 18(10): 1789-96, 1999 Mar 11.
Article in English | MEDLINE | ID: mdl-10086333

ABSTRACT

The retinoblastoma protein plays a critical role in regulating the G1/S transition. Less is known about the function and regulation of the homologous pocket protein p107. Here we present evidence for the posttranslational regulation of p107 by the Ca2+-activated protease calpain. Three negative growth regulators, the HMG-CoA reductase inhibitor lovastatin, the antimetabolite 5-fluorouracil, and the cyclic nucleotide dibutyryl cAMP were found to induce cell type-specific loss of p107 protein which was reversible by the calpain inhibitor leucyl-leucyl-norleucinal but not by the serine protease inhibitor phenylmethylsulfonylfluoride, caspase inhibitors, or lactacystin, a specific inhibitor of the 26S proteasome. Purified calpain induced Ca2+-dependent p107 degradation in cell lysates. Transient expression of the specific calpain inhibitor calpastatin blocked the loss of p107 protein in lovastatin-treated cells, and the half-life of p107 was markedly lengthened in lovastatian-treated cells stably transfected with a calpastatin expression vector versus cells transfected with vector alone. The data presented here demonstrate down-regulation of p107 protein in response to various antiproliferative signals, and implicate calpain in p107 posttranslational regulation.


Subject(s)
Calpain/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Retinoblastoma Protein/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Bucladesine/pharmacology , Cyclin B/metabolism , Cyclin B1 , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Fluorouracil/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Ketones/pharmacology , Leupeptins/pharmacology , Lovastatin/pharmacology , Retinoblastoma-Like Protein p107 , Tumor Cells, Cultured
17.
Biochim Biophys Acta ; 1449(3): 261-8, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10209305

ABSTRACT

Classical cytotoxic therapy has been minimally useful in the treatment of hepatocellular carcinoma. In an effort to develop a new approach to the treatment of this neoplasm, we have investigated the signal transduction pathways regulating the growth of human hepatoma cells. In the data reported here, cyclic AMP (cAMP), a negative growth regulator for many cells of epithelial origin, induced G1 synchronization and apoptosis in the HepG2 human hepatoma cell line. The effects of cAMP on the components of the G1/S transition were analyzed. There was no detectable effect of two different cAMP analogs, 8-bromo cAMP or dibutyryl cAMP on the level of the D-type cyclins, cyclin E, cyclin-dependent kinase 2, cyclin-dependent kinase 4, p53, or the cyclin-dependent kinase inhibitors p21 or p27. In contrast, the cAMP analogs induced a dramatic downregulation of cyclin A protein, cyclin A messenger RNA, and cyclin A-dependent kinase activity. Cyclin A-dependent kinase has been shown to be required for the G1-S transition. Furthermore, cyclin A deregulation has been implicated in the pathogenesis of hepatocellular carcinoma. The data reported here suggest a novel signal transduction-based approach to hepatoma therapy.


Subject(s)
CDC2-CDC28 Kinases , Cyclic AMP/pharmacology , Cyclin A/antagonists & inhibitors , Proto-Oncogene Proteins , Apoptosis/drug effects , Carcinoma, Hepatocellular , Cell Cycle/drug effects , Cyclin A/genetics , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/antagonists & inhibitors , Down-Regulation , Gene Expression , Humans , Liver Neoplasms , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Messenger/metabolism , Time Factors , Tumor Cells, Cultured
18.
Leukemia ; 14(7): 1276-83, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914553

ABSTRACT

Inhibitors of proteases are currently emerging as a potential anti-cancer modality. Nonselective protease inhibitors are cytotoxic to leukemia and cancer cell lines and we found that this cytotoxicity is correlated with their potency as inhibitors of the proteasome but not as inhibitors of calpain and cathepsin. Highly selective inhibitors of the proteasome were more cytotoxic and fast-acting than less selective inhibitors (PS341>>ALLN>>ALLM). Induction of wt p53 correlated with inhibition of the proteasome and antiproliferative effect in MCF7, a breast cancer cell line, which was resistant to apoptosis caused by proteasome inhibitors. In contrast, inhibitors of the proteasome induced apoptosis in four leukemia cell lines lacking wt p53. The order of sensitivity of leukemia cells was: Jurkat>HL60> or =U937>>K562. The highly selective proteasome inhibitor PS-341 induced cell death with an IC50 as low as 5 nM in apoptosis-prone leukemia cells. Cell death was preceded by p21WAF1/CIP1 accumulation, an alternative marker of proteasome inhibition, and by cleavage of PARP and Rb proteins and nuclear fragmentation. Inhibition of caspases abrogated PARP cleavage and nuclear fragmentation and delayed, but did not completely prevent cell death caused by PS-341. Reintroduction of wt p53 into p53-null PC3 prostate carcinoma cells did not increase their sensitivity to proteasome inhibitors. Likewise, comparison of parental and p21-deficient cells demonstrated that p21WAF1/CIP1 was dispensable for proteasome inhibitor-induced cytotoxicity. We conclude that accumulation of wt p53 and induction of apoptosis are independent markers of proteasome inhibition.


Subject(s)
Apoptosis/drug effects , Cyclins/metabolism , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Neoplasm Proteins/metabolism , Protease Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Acetylcysteine/toxicity , Acrylates/pharmacology , Acrylates/toxicity , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Chloromethyl Ketones/toxicity , Boronic Acids/pharmacology , Boronic Acids/toxicity , Bortezomib , Calpain/antagonists & inhibitors , Cathepsins/antagonists & inhibitors , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Drug Synergism , Genes, p53 , Humans , Jurkat Cells/cytology , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Leupeptins/pharmacology , Leupeptins/toxicity , Neoplasm Proteins/genetics , Neoplasms/pathology , Oligopeptides/pharmacology , Oligopeptides/toxicity , Protease Inhibitors/toxicity , Proteasome Endopeptidase Complex , Pyrazines/pharmacology , Pyrazines/toxicity , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , U937 Cells/cytology , U937 Cells/drug effects , U937 Cells/metabolism
19.
Leukemia ; 15(10): 1537-43, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11587211

ABSTRACT

The Bcr-Abl fusion protein drives leukemogenesis and can render leukemia cells resistant to conventional chemotherapy. Geldanamycin (GA), a drug which destabilizes Hsp90-associated proteins, depletes cells of Bcr-Abl, an Hsp90 client, but not of Abl. Both HL60 cells transfected with Bcr-Abl and naturally Ph1-positive K562 leukemia cells are resistant to most cytotoxic drugs, but were found to be sensitive to GA. Furthermore, GA sensitized Bcr-Abl-expressing cells to doxorubicin (DOX) and paclitaxel (PTX). In contrast, in parental HL60 cells, 90 nM GA inhibited PARP cleavage, nuclear fragmentation, and cell death caused by 500 ng/ml DOX. Like GA, STI 571 (an inhibitor of the Abl kinase) sensitized Bcr-Abl-expressing cells to DOX. Unlike GA, STI 571 did not antagonize the cytotoxic effects of DOX in parental HL60 cells. These results indicate that sensitization of Bcr-Abl-expressing cells, but not desensitization of HL60 cells, depends on inhibition of Bcr-Abl. Thus, GA differentially affects leukemia cells depending on their Bcr-Abl expression and selectively increases apoptosis in Bcr-Abl-expressing cells.


Subject(s)
Fusion Proteins, bcr-abl/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Leukemia/pathology , Quinones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzoquinones , Doxorubicin/pharmacology , Drug Interactions , Drug Resistance , Fusion Proteins, bcr-abl/biosynthesis , Humans , Lactams, Macrocyclic , Leukemia/metabolism , Paclitaxel/pharmacology , Transfection , Tumor Cells, Cultured/drug effects
20.
FEBS Lett ; 353(2): 129-32, 1994 Oct 17.
Article in English | MEDLINE | ID: mdl-7926036

ABSTRACT

The detergent Triton X-100 (TX100) was used with the intention to establish a model for necrotic cell death. However, TX100 was found to induce apoptotic and necrotic death in prostate and colon cancer cell lines. Apoptosis was characterized by the typical morphological features and internucleosomal DNA fragmentation. The rapid onset within 60 min and the lack of inhibition by cycloheximide indicated that apoptosis induced by TX100 was not dependent on protein synthesis. Removal of extracellular calcium blocked internucleosomal DNA fragmentation. This pattern of cell death shows a striking similarity to the effect of cytotoxic lymphocytes on their target cells.


Subject(s)
Apoptosis/drug effects , Cell Death/drug effects , Octoxynol/pharmacology , T-Lymphocytes, Cytotoxic/physiology , Calcium/metabolism , Colonic Neoplasms/pathology , Cycloheximide/pharmacology , DNA, Neoplasm/drug effects , Humans , Kinetics , Male , Prostatic Neoplasms/pathology , Protein Biosynthesis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL