Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Prostate ; 68(4): 360-72, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18189232

ABSTRACT

BACKGROUND: Docetaxel-based chemotherapy is the only treatment that demonstrated an overall survival benefit in men with hormone refractory prostate cancer. 2-CADO inhibits the growth of PC3 cells by inducing apoptosis and cell cycle arrest through a mechanism that involves cellular uptake. METHODS: Androgen-independent and -sensitive (PC3 and LNCaP) prostate cancer cells and non-neoplastic HECV cells were used in the study. Proliferation and cell cycle progression were analyzed in the presence of 2-CADO and Docetaxel. Invasive potential was assessed by soft agar assay and metastatic ability by adhesion assay. IL-23 and PAR-1 expression were determined by real time PCR. RESULTS: 2-CADO pre-treatment followed by Docetaxel at subclinical dosage reduced the viability of either PC3 or LNCaP while it did not enhance Docetaxel-induced cytotoxicity in adherent non-neoplastic HECV. The drugs reduced the invasive potential of PC3 cells by inducing apoptosis and blocking cell cycle progression in the S-phase. Down-regulation of PAR-1 gene expression resulted in a slightly lower metastatic potential, whereas up-regulation of IL-23 induced the activation of the immune system. CONCLUSIONS: Pretreatment of PC3 cells with 2-CADO decreased the effective concentration of Docetaxel, lowered the metastatic potential, and induced the production of cytokines known to stimulate the immune response against cancer. The treatment was effective for prostate cancer cells independently on their androgen sensitiveness.


Subject(s)
2-Chloroadenosine/toxicity , Antineoplastic Agents/toxicity , Interleukin-23 Subunit p19/genetics , Prostatic Neoplasms/drug therapy , Receptor, PAR-1/genetics , Taxoids/toxicity , Agar , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Docetaxel , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-23 Subunit p19/immunology , Interleukin-23 Subunit p19/metabolism , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Receptor, PAR-1/metabolism , S Phase/drug effects , Stem Cells/drug effects
2.
Int J Mol Med ; 14(1): 113-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15202025

ABSTRACT

Pre-treatment with bryostatin 1 (bryo) has been shown to potentiate the efficacy of (2-chloro-2-deoxyadenosine, cladribine, 2-CdA) in B-cell chronic lymphocytic leukemia (B-CLL) by increasing the ratio of deoxycytidine kinase (dCK) to 5'-nucleotidase (5'-NT) activity. The bryo-induced increase in dCK/5'-NT activity alone has not been a conclusive indication of final clinical outcome. Therefore, we used an ex vivo assay to investigate factors which may affect the bryo-induced enhancement of 2-CdA efficacy in B-CLL patient-derived samples. Bryo-induced increase in dCK/5'-NT was inversely associated with Rai stage CLL (r=-0.86). Increased dCK/5'-NT activity was not correlated with increased efficacy (cell death) or percentage of cellular [8-3H]-2-CdA converted to [8-3H]-2-CdATP ex vivo. Bryo pre-treatment increased the cellular uptake of [8-3H]-2-CdA and incorporation of [8-3H]-2-CdA metabolites into the DNA fraction. Cell death from 2-CdA was inversely correlated with bryo-induced activity of the DNA repair enzyme, DNA-PKcs, (r=-0.77). Thus, the ability of B-CLL to repair damaged DNA may be a more important predictor of the response to bryo/2-CdA and eventual clinical outcome than dCK/5'-NT activity. Additional CLL patients under bryo-2-CdA therapy are needed to verify these important observations.


Subject(s)
2-Chloroadenosine/analogs & derivatives , 2-Chloroadenosine/toxicity , Antimetabolites, Antineoplastic/toxicity , Deoxyadenosines/toxicity , Lactones/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , 2-Chloroadenosine/pharmacokinetics , 5'-Nucleotidase/metabolism , Bryostatins , Cell Death , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Deoxyadenosines/pharmacokinetics , Deoxycytidine Kinase/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Macrolides , Nuclear Proteins , Protein Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , ras Proteins/genetics , ras Proteins/metabolism
3.
Blood ; 92(8): 2893-8, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9763575

ABSTRACT

We have examined the cytotoxic effects of cyclic adenosine-3', 5'-monophosphate (cAMP) derivatives on multiple myeloma cells lines and determined that the 8-Chloro substituted derivative (8Cl-cAMP) is one of the most potent. We report here that 8Cl-cAMP is cytotoxic to both steroid sensitive and insensitive myeloma cells with a half maximal concentration of approximately 3 micromol/L. 8Cl-cAMP toxicity in myeloma cells is dependent on phosphodiesterase activity in the serum of cell culture medium. A metabolite of 8Cl-cAMP, 8-Chloro-adenosine (8Cl-AD), kills myeloma cells as effectively as 8Cl-cAMP. Adenosine deaminase (ADA) converts 8Cl-AD into 8Cl-inosine and abrogates the cytotoxic effects of 8Cl-cAMP, 8Cl-AMP, and 8Cl-AD, as does 5-(p-Nitrobenzyl)-6-Thio-Inosine (NBTI), an inhibitor of nucleoside uptake. These data suggest that 8Cl-cAMP must be converted to 8Cl-AD and that 8Cl-AD is the compound that enters the cell. Contrary to glucocorticoid-mediated cell death in myeloma cells, the pathway of 8Cl-AD-mediated cell death appears to be independent of interleukin-6 (IL-6) actions. Although the exact mode of action for this agent is currently unknown, its ability to kill steroid sensitive and insensitive multiple myeloma cells in an IL-6 independent fashion may offer exciting new therapeutic options.


Subject(s)
2-Chloroadenosine/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , Antimetabolites, Antineoplastic/pharmacology , Multiple Myeloma/pathology , Prodrugs/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , 2-Chloroadenosine/toxicity , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenosine Deaminase/metabolism , Animals , Antimetabolites, Antineoplastic/metabolism , Apoptosis/drug effects , Biological Transport , Biotransformation , Cattle , Culture Media , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Interleukin-6/pharmacology , Phosphoric Diester Hydrolases/blood , Prodrugs/metabolism , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Tumor Cells, Cultured
4.
Zhonghua Zhong Liu Za Zhi ; 17(1): 5-8, 1995 Jan.
Article in Chinese | MEDLINE | ID: mdl-7656789

ABSTRACT

8-chloroadenosine showed marked activity against mice solid tumor hepatoma 22 (H22) and ascitic leukemia L-1210. At 100mg. Kg-1. /d x 7, the inhibition rate of H22 was 71.7 +/- 13.3% (P < 0.01) and 66.1 +/- 4.46% (P < 0.01), i.p. and i.v., respectively; at the same dose, the life-prolonging rate of mice bearing L-1210 was 124.0 +/- 22.1% (P < 0.01) and 104.2 +/- 20.1% (P < 0.01), i.p. and i.v., respectively. 8-chloroadenosine also showed activity against 3 human cancer cell lines in vitro. The IC50 values were determined by measuring cell growth using trypan blue dye exclusion. The results showed that HL-60 and K562, and human gastric cancer cell line MGc80-3 and IC50 values of 1. 8 mumol/L, 4.2 mumol/L and 1.56 mumol/L, respectively. The toxicity of 8-chloroadenosine was low, with LD50 of 1025.0 +/- 52.4 mg/kg for mice and 793.4 +/- 70.1 mg/kg for rats by single i.p. injection.


Subject(s)
2-Chloroadenosine/analogs & derivatives , Antineoplastic Agents/pharmacology , Leukemia L1210/drug therapy , Liver Neoplasms, Experimental/drug therapy , 2-Chloroadenosine/pharmacology , 2-Chloroadenosine/therapeutic use , 2-Chloroadenosine/toxicity , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Female , Lethal Dose 50 , Leukemia L1210/pathology , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred DBA , Neoplasm Transplantation , Rats , Rats, Wistar , Stomach Neoplasms/pathology , Tumor Cells, Cultured/drug effects
5.
Biochem J ; 304 ( Pt 3): 877-85, 1994 Dec 15.
Article in English | MEDLINE | ID: mdl-7818494

ABSTRACT

Incubation of human thymocytes with an optimum concentration of adenosine and its receptor site agonist, 2-chloroadenosine, induced increases in intracellular cyclic AMP (cAMP) (from a resting 0.6 +/- 0.1 to 4.1 +/- 0.2 pmol/10(7) cells within 5 min) and Ca2+ (from the resting 85 +/- 7 nM to a peak of 210 +/- 25 nM) levels and resulted in internucleosomal DNA fragmentation and cell death (apoptosis). Other adenosine analogues were also effective at inducing DNA fragmentation, the order of potency being 2-p-(carboxyethylphenylethylamino)-5'-carboxyamidoadenosine < 5'-(N-ethylcarboxamide)adenosine < or = cyclopentyladenosine < 2-chloroadenosine (2-CA). 2-CA treatment (with an optimum concentration of 40 microM) selectively depleted a thymocyte subpopulation (15-20% of the total cells) which expressed higher levels of the CD3 molecule and which was found mainly in the CD4+CD8+ double positive immature thymocyte population. DNA fragmentation was prevented by the addition of actinomycin D or cycloheximide to the thymocyte suspension, indicating that this process required both mRNA and protein synthesis. Endonuclease activation and cell killing were dependent on an early, sustained increase in cytosolic Ca2+ concentration, most of which was of extracellular origin and was a result of an adenosine-induced inositol trisphosphate release. Other agents known to elevate intracellular cAMP levels by different mechanisms failed to induce similar DNA fragmentation, but enhanced the effect of adenosine. This suggested a supporting role for cAMP in adenosine-induced DNA fragmentation. Phorbol dibutyrate, a protein kinase. C activator, previously shown to inhibit Ca(2+)-dependent DNA fragmentation and cell killing in human thymocytes [McConkey, Hartzell, Jondal and Orrenius (1989) J. Biol. Chem. 264, 13399-13402], at 60 ng/ml concentration also prevented adenosine-induced DNA fragmentation when added prior to adenosine. This suggested a complex cross-talk between the adenosine-triggered signal transduction cascade and the activation state of protein kinase C in regulating apoptosis of human thymocytes.


Subject(s)
Adenosine/toxicity , Apoptosis/drug effects , Apoptosis/physiology , Calcium/metabolism , DNA Damage , DNA/drug effects , DNA/metabolism , Thymus Gland/drug effects , Thymus Gland/metabolism , 2-Chloroadenosine/toxicity , Adenosine/analogs & derivatives , Adenylyl Cyclases/metabolism , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Child, Preschool , Cyclic AMP/metabolism , Enzyme Activation , Humans , Infant , Inositol Phosphates/metabolism , Intracellular Fluid/metabolism , Lymphocyte Count/drug effects , Protein Kinase C/metabolism , Purinergic P1 Receptor Agonists , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/metabolism , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/physiology , Stimulation, Chemical , T-Lymphocyte Subsets/drug effects , Thymus Gland/cytology
6.
Proc Natl Acad Sci U S A ; 89(7): 2970-4, 1992 Apr 01.
Article in English | MEDLINE | ID: mdl-1348362

ABSTRACT

2-Chlorodeoxyadenosine (CdA) is active in chronic lymphocytic leukemia, hairy-cell leukemia, and low-grade lymphomas. In part, this spectrum of activity may be attributable to the selective toxicity of CdA to nondividing lymphocytes and monocytes. However, CdA is unstable at acidic pH and is degraded by bacterial nucleoside phosphorylases. The present experiments demonstrate that the 2'-arabino-fluoro derivative of CdA, designated CAFdA, is also directly toxic to quiescent lymphocytes and macrophages. Unlike CdA, CAFdA was stable at pH 2 and resisted degradation by Escherichia coli nucleoside phosphorylase. Cell killing was preceded by the formation of DNA strand breaks and could be prevented by supplementation of the medium with deoxycytidine. The initial DNA damage initiated the pattern of oligonucleosomal DNA fragmentation characteristic of apoptosis. Mutant lymphoblasts, deficient in deoxycytidine kinase, with elevated cytoplasmic 5'-nucleotidase, or with expanded deoxynucleotide pools secondary to increased ribonucleotide reductase activity, were cross-resistant to both CAFdA and CdA toxicity. One-week oral treatment with CAFdA (1 mg/ml in drinking water) achieved an average plasma concentration of 0.56 microM and eliminated 90% of chronic lymphocytic leukemia cells transplanted into severe combined immunodeficiency (scid) mice. Under the same conditions, CdA was much less active. Collectively, these results suggest that CAFdA could be effective as an oral agent in indolent lymphoproliferative diseases and in autoimmune diseases where lymphocyte and monocyte depletion is desirable.


Subject(s)
2-Chloroadenosine/analogs & derivatives , Antimetabolites, Antineoplastic/toxicity , Arabinonucleosides/toxicity , Cell Death/drug effects , DNA Damage , Deoxyadenosines/toxicity , Lymphocytes/drug effects , Monocytes/drug effects , 2-Chloroadenosine/chemistry , 2-Chloroadenosine/pharmacokinetics , 2-Chloroadenosine/toxicity , Adenine Nucleotides , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics , Arabinonucleosides/chemistry , Arabinonucleosides/pharmacokinetics , Cladribine , Clofarabine , DNA/drug effects , Deoxyadenosines/chemistry , Deoxyadenosines/pharmacokinetics , Leukemia, Hairy Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Mice , Mice, SCID
7.
J Immunol ; 147(12): 4302-6, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1753100

ABSTRACT

Mononuclear phagocytes play a central role in the pathogenesis of chronic inflammatory diseases. It is therefore important to define chemotherapeutically exploitable metabolic pathways that distinguish monocytes from other cell types. Blood monocytes do not synthesize deoxynucleotides de novo, and their transformation to macrophages occurs without cell division. Whether or not monocytes can repair DNA damage, and whether or not DNA repair is necessary for their survival, is unknown. The present experiments demonstrate that normal human monocytes, unlike neutrophils, rapidly repair DNA strand breaks induced by gamma-irradiation. Monocyte extracts contain functional immunoreactive DNA polymerase-alpha. DNA repair synthesis in normal monocytes is blocked by aphidicolin, an inhibitor of DNA polymerase-alpha with respect to dCTP. Aphidicolin is also directly toxic to normal monocytes, but has no effect on nondividing lymphocytes or fibroblasts. Compared to most other cell types, monocytes and macrophages have very low dCTP pools, but abundant deoxycytidine kinase activity. This suggests that dCTP derived from salvage pathways is important for DNA repair in these cells. Consistent with this notion, exogenous deoxycytidine could partially protect monocytes from aphidicolin killing. The unexpected toxicity of aphidicolin toward normal human monocytes may be attributable to their high rate of spontaneous DNA strand break formation, to the importance of DNA polymerase-alpha for DNA repair in these cells, and to their minute dCTP pools.


Subject(s)
Cell Survival/physiology , DNA Repair/physiology , Monocytes/physiology , 2-Chloroadenosine/toxicity , Aphidicolin/toxicity , Cell Survival/drug effects , Cells, Cultured , DNA Polymerase II/antagonists & inhibitors , DNA Repair/drug effects , Deoxycytidine Kinase/analysis , Humans , Monocytes/drug effects , Monocytes/metabolism , Thymidine Kinase/analysis
8.
J Clin Invest ; 86(5): 1480-8, 1990 Nov.
Article in English | MEDLINE | ID: mdl-1700795

ABSTRACT

Lymphoid cells were thought to be uniquely susceptible to excess 2'-deoxyadenosine (dAdo), when exposed to inhibitors of adenosine deaminase (ADA). However, we now find that human monocytes are as sensitive as lymphocytes to dAdo or to the ADA-resistant congener 2-chloro-2'-deoxyadenosine (CldAdo). Monocytes exposed in vitro to CldAdo, or to dAdo plus deoxycoformycin rapidly developed DNA strand breaks. Both the DNA damage and the toxicity of CldAdo or dAdo toward monocytes were blocked by deoxycytidine, but not by inhibitors of poly(ADP-ribose) polymerase. A partial decrease in RNA synthesis and a gradual decline of cellular NAD were early biochemical events associated with monocyte DNA damage. Low CldAdo concentrations (5-20 nM) inhibited monocyte phagocytosis and reduced the release of interleukin 6. Higher CldAdo concentrations led to a dose- and time-dependent loss of monocyte viability. Circulating monocytes disappeared within 1 wk in patients with cutaneous T cell lymphoma or with rheumatoid arthritis during continuous CldAdo infusion. The marked sensitivity of human monocyte function and survival to CldAdo in vitro, together with the monocyte depletion in patients receiving CldAdo chemotherapy, suggests that CldAdo or other dAdo analogues offer a novel therapeutic strategy for chronic inflammatory and autoimmune diseases characterized by inappropriate monocyte deployment or function.


Subject(s)
2-Chloroadenosine/analogs & derivatives , Deoxyadenosines/toxicity , Monocytes/drug effects , 2-Chloroadenosine/therapeutic use , 2-Chloroadenosine/toxicity , Adenosine Triphosphate/metabolism , Cell Survival/drug effects , Cells, Cultured , Cladribine , DNA Damage , Deoxyadenosines/pharmacology , Deoxyadenosines/therapeutic use , Humans , Interleukin-6/metabolism , Lymphoma, T-Cell/blood , Lymphoma, T-Cell/drug therapy , Monocytes/cytology , Monocytes/metabolism , Mutagens , NAD/metabolism , Phagocytosis/drug effects , Protein Biosynthesis , RNA/biosynthesis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...