Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Acta Pol Pharm ; 62(3): 195-205, 2005.
Article in English | MEDLINE | ID: mdl-16193812

ABSTRACT

Tezacitabine (FMdC) is a new cytostatic/cytotoxic agent widely investigated in clinical trials and on the cellular level. In a previous paper (3) we worked on human and murine leukemia (L-1210, HL-60, and MOLT-4) cells, and in this paper we investigated the influence of FMdC on the cell cycle and apoptosis in vitro of three other leukemias (CCRF-SB, KG-1, and Jurkat), and human solid tumor (carcinoma) cell lines (COLO-205, MCF-7, and PC-3). We found that FMdC induces the G1 (at concentrations higher than 10 nM). and S-phase (at low concentration) leaky block of the cell cycle. FMdC also effectively induces apoptotic death of cells by the caspase 3/7 pathway. We found also that FMdC induces intensive changes in the protein metabolism. These changes are correlated with the cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , G1 Phase/drug effects , S Phase/drug effects , Animals , Cell Line, Tumor , Deoxycytidine/pharmacology , Humans , Jurkat Cells , Mice
2.
Cancer Lett ; 229(1): 67-75, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16157220

ABSTRACT

In vitro cytotoxicity tests currently in use applied in the developmental stages of anticancer drug discovery are able to select the most potent compounds, but are not predictive of their potential toxicity. In this study, we have demonstrated the applicability of neutral red uptake assay using mouse fibroblasts Balb/c 3T3 cell line (3T3 NRU assay) for in vitro toxicity testing of newly synthesized genistein glycosides, the compounds that appear to show anticancer activity. We have also proven the compatibility of in-house 3T3 NRU assay with the prediction model for acute rodent oral toxicity testing, endorsed by NIEHS-ICCVAM workshop. The combined results from the cytotoxicity and the in vitro toxicity tests facilitated the selection of the most promising genistein derivatives, compounds G21 and G23, which were the most active and selective towards cancer cells. The comparison of predicted LD50 values revealed that almost all genistein derivatives are at least two-fold less toxic than the chemotherapeutics currently used in cancer therapy, which is very promising for this new group of compounds.


Subject(s)
Genistein/pharmacology , Genistein/toxicity , Animals , Cell Culture Techniques , Coloring Agents/pharmacokinetics , Drug Screening Assays, Antitumor , Fibroblasts , Genistein/analogs & derivatives , Lethal Dose 50 , Mice , Mice, Inbred BALB C , Models, Theoretical , Neoplasms/drug therapy , Neutral Red/pharmacokinetics , Predictive Value of Tests , Sensitivity and Specificity
3.
Acta Biochim Pol ; 52(2): 561-5, 2005.
Article in English | MEDLINE | ID: mdl-15933759

ABSTRACT

The aim of the study was to determine the relation between the cytotoxic and cytostatic effects of tezacitabine and cladribine on a HL-60 cell line and the time of exposure of cells to these drugs. Cell viability and induction of apoptosis were assessed using flow cytometry methods. Apoptosis was confirmed by direct microscopic observation. Growth inhibition was examined by cell counting. After 24 h incubation tezacitabine was equally or less toxic compared to cladribine. However, toxicity of tezacitabine strongly rose after 48 h incubation leading to massive cell death at doses much lower than those of cladribine. Assessment of the effect of increased exposure time on the clinical efficacy of tezacitabine is indicated.


Subject(s)
Antineoplastic Agents/toxicity , Cladribine/toxicity , Deoxycytidine/analogs & derivatives , Apoptosis/drug effects , Cell Survival/drug effects , Deoxycytidine/toxicity , Flow Cytometry , HL-60 Cells , Humans
4.
Acta Pol Pharm ; 61(4): 297-305, 2004.
Article in English | MEDLINE | ID: mdl-15575597

ABSTRACT

Physiologic deoxynucleotides are required for an error-proof DNA replication, repair and synthesis. Any inaccuracy in this process results in a block in DNA synthesis until the error is corrected. If the cell enzymes are unable to correct the error, a signal for apoptosis is generated. This mechanism is the main target for anticancer nucleoside analogs. They also interact with the metabolism of physiological nucleosides, and consequently, have a large number of intracellular targets to induce cytotoxicity. In addition, it is now reported that some analogs may interfere directly with RNA synthesis. A great deal of synthesized nucleoside analogs provide the opportunity to understand the structure-based differences in their metabolism and mechanisms of action as well as to identify the specific intracellular targets and diseases, in which each of these newer nucleoside analogs acts most efficiently. This paper summarizes developments in the area of new nucleoside analogs undergoing clinical evaluation for the treatment of solid tumors, namely tezacitabine, troxacitabine, DMDC, CNDAC, ECyD, clofarabine, and decitabine.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , Cytarabine/analogs & derivatives , Cytidine/analogs & derivatives , Cytosine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Nucleosides/chemistry , Nucleosides/therapeutic use , Adenine Nucleotides , Animals , Antineoplastic Agents/pharmacology , Arabinonucleosides/chemistry , Arabinonucleosides/pharmacology , Arabinonucleosides/therapeutic use , Azacitidine/chemistry , Azacitidine/pharmacology , Azacitidine/therapeutic use , Clinical Trials as Topic , Clofarabine , Cytarabine/chemistry , Cytarabine/pharmacology , Cytarabine/therapeutic use , Cytidine/chemistry , Cytidine/pharmacology , Cytidine/therapeutic use , Cytosine/chemistry , Cytosine/pharmacology , Cytosine/therapeutic use , Decitabine , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Dioxolanes/chemistry , Dioxolanes/pharmacology , Dioxolanes/therapeutic use , Humans , Nucleosides/pharmacology
5.
Acta Pol Pharm ; 61(3): 223-32, 2004.
Article in English | MEDLINE | ID: mdl-15481249

ABSTRACT

Cytotoxic nucleoside analogs have a broad clinical use. They were among the first chemotherapeutic agents used in the treatment of malignant diseases. The anticancer nucleosides include analogs of physiologic pyrimidine and purine nucleosides. They are used in oncology in the treatment of both, solid tumors and hematological malignancies. These agents have many intracellular targets, e.g. they act as antimetabolites, competing with natural nucleosides during DNA or RNA synthesis and as inhibitors of key cell enzymes. Understanding of the mechanisms of action of these compounds and synthesis of new analogs provides the possibility to further expand the spectrum of their clinical use and enhance their antitumor activity. In this paper we describe mechanisms of action and possible clinical use in the treatment of hematological malignancies of these nucleoside analogs, which are now in different stages of clinical trials, namely tezacitabine, troxacitabine, clofarabine, nelarabine, decitabine, CNDAC and ECyD.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytarabine/analogs & derivatives , Cytosine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Hematologic Neoplasms/drug therapy , Purine Nucleosides/therapeutic use , Pyrimidine Nucleosides/therapeutic use , Adenine Nucleotides , Animals , Antineoplastic Agents/adverse effects , Arabinonucleosides/adverse effects , Arabinonucleosides/pharmacokinetics , Arabinonucleosides/therapeutic use , Clinical Trials as Topic , Clofarabine , Cytarabine/adverse effects , Cytarabine/therapeutic use , Cytosine/adverse effects , Cytosine/therapeutic use , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Dioxolanes/adverse effects , Dioxolanes/therapeutic use , Half-Life , Humans , Purine Nucleosides/adverse effects , Pyrimidine Nucleosides/adverse effects , Stereoisomerism
6.
Oncol Rep ; 12(3): 509-16, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15289829

ABSTRACT

Promoter hypermethylation within CpG islands plays an important role in the silencing of numerous genes involved in tumor growth including tumor suppressor genes and genes encoding proteins involved in the execution of apoptosis. Here we show that CpG islands are also found within the promoter regions of both human and mouse TNFR1 (TNFRSF1) genes. Selective inhibition of methyltransferases with 5-aza-2'-deoxycytidine increases the expression of TNFR1 in human (WM35) and murine (B16F10) melanoma cells and sensitizes them to TNF-induced apoptosis both in vitro and in vivo. Treatment of mice with the combination of 5-aza-2'-deoxycytidine and recombinant TNF leads to potentiated antitumor effects. Importantly the antitumor efficacy of the combination treatment is shown when both drugs are used in doses that do not exert any antitumor effects when used alone. Altogether our studies show that the combination treatment with 5-aza-2'-deoxycytidine and TNF might be effective in the treatment of melanoma.


Subject(s)
Antigens, CD/biosynthesis , Apoptosis , Azacitidine/analogs & derivatives , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Cell Line, Tumor , Cell Separation , CpG Islands , DNA Fragmentation , Decitabine , Flow Cytometry , Humans , In Situ Nick-End Labeling , Melanoma/drug therapy , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Necrosis , Neoplasm Transplantation , Promoter Regions, Genetic , RNA/metabolism , Receptors, Tumor Necrosis Factor, Type I , Recombinant Proteins/chemistry , Ribonucleases/metabolism , Time Factors
7.
Int J Oncol ; 24(5): 1149-57, 2004 May.
Article in English | MEDLINE | ID: mdl-15067336

ABSTRACT

Competitive inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase are commonly used in the clinic to treat hypercholesterolemia and have been reported to exert antitumor effects. Cerivastatin is a novel, synthetic and the most pharmacologically potent inhibitor of HMG-CoA reductase. We decided to examine the cytostatic/cytotoxic activity of cerivastatin against human breast cancer cell lines and to test whether the effects of cerivastatin could be potentiated by doxorubicin and cisplatin. Cytostatic/cytotoxic effects of cerivastatin used alone or in the combination with chemotherapeutics were measured with MTT assay. The cell cycle distribution and apoptosis induction were evaluated with flow cytometer. The expression of p21 and p27 cyclin-dependent kinase inhibitors was measured with Western blotting. Isobologram analysis was performed to study the drug interactions. We observed that cerivastatin exerts cytostatic/cytotoxic effects against four human tumor cell lines (T-47D, T4-2, MDA-MB-231, MCF-7). We also demonstrated that cerivastatin exerts growth inhibitory effect through induction of p21 cyclin-dependent kinase inhibitor and inhibition of cell cycle progression. In the two tumor cell lines studied, one sensitive (MDA-MB-231) and one moderately resistant (T4-2) to the cytostatic/cytotoxic effects of cerivastatin we examined the effects of combined treatment with cerivastatin and either doxorubicin or cisplatin. Cerivastatin potentiated cytostatic/cytotoxic effects of cisplatin against T4-2 cells and those of doxorubicin against both cell lines. In T4-2 cells the interaction between doxorubicin and cerivastatin and between cisplatin and cerivastatin was found to be synergistic. Altogether, these studies indicate that cerivastatin is another HMG-CoA reductase inhibitor with potent antitumor effects.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/pathology , Intracellular Signaling Peptides and Proteins , Blotting, Western , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cisplatin/administration & dosage , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Doxorubicin/administration & dosage , Drug Interactions , Drug Synergism , Female , Humans , Pyridines/administration & dosage , Tumor Cells, Cultured
8.
J Inorg Biochem ; 98(3): 553-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14987857

ABSTRACT

A novel class of water-soluble Pt(IV) complexes with histamine (Hist) and radioiodinated histamine ([(125)I/(131)I]Hist) has been synthesised with the goal of potential application for concomitant anticancer radio-chemotherapy of solid tumours. The prepared complex of 1:2 metal:ligand stoichiometry ([Pt(IV)(Hist)(2)(OH)(2)]Cl(2)) was characterised by microanalysis, mass spectrometry, and chromatographic methods. Cytotoxic/cytostatic activities of the complex were examined by flow cytometry method using the MCF-7 cells line. A slightly lower cytotoxicity of the Pt(IV) complex comparing to cisplatin was found (IC(50) 59 and 48 microM, respectively). Both cisplatin and the histamine complex show a cytostatic activity by blocking MCF-7 cells in S-phase of cell cycle. Biodistribution studies in normal rats revealed the highest accumulation of the (131)I-labelled complex in liver and kidneys (41.3% and 12.4% ID after 24 h post-intravenous injection (p.i.v.)). The similar pharmacokinetics was observed in tumour-bearing C3H/W mice, however, a lower accumulation in liver was observed following an intraperitoneal comparing to an intravenous administration. A concentration of the complex in tumour increased with time post-intraperitoneal injection (1.2 and 2.5%ID/g after 2 and 24 h (p.i.), respectively). An increasing tumour/muscle ratio was also observed (2.2 and 4.5 after 2 and 24 h p.i., respectively), and that suggests a penetration of the complex into the tumour cells, and a permanent binding with some cellular components, probably with the DNA.


Subject(s)
Histamine/analogs & derivatives , Iodine Radioisotopes/pharmacology , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/pharmacology , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Iodine Radioisotopes/chemistry , Isotope Labeling , Male , Mice , Mice, Inbred C3H , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacokinetics , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Solubility , Spectrometry, Mass, Electrospray Ionization , Tissue Distribution
9.
Cancer Lett ; 203(1): 59-69, 2004 Jan 08.
Article in English | MEDLINE | ID: mdl-14670618

ABSTRACT

Genistein, the principal soy isoflavone, is a molecule of great interest as an innovative chemotherapeutic agent or as a lead-compound in anticancer drug design. To enhance intrinsic activity of genistein and to explore its pharmacophoric potential, its glycosidic derivatives were synthesized. On the basis of structural features and calculated lipophilicity coefficient (ClogP) the derivatives were classified as hydrophilic (i.e. those containing free sugar moiety) or lipophilic (i.e. those with alkylated or acylated sugar hydroxyls). The in vitro cytostatic and cytotoxic studies showed hydrophilic glycosides to be practically inactive against human cancer cell lines when compared to the free aglycone. On the contrary, lipophilic glycosides were significantly more active than the parent isoflavone although the correlation between ClogP and the activity was not clear. On the basis of GI50 and LC50 values two of the most active glycosides were found to be several times more potent in their cytostatic and cytotoxic effect than genistein. Additionally all lipophilic glycosides were revealed to exhibit different mode of action in comparison to genistein. It may suggest that these compounds do not undergo rapid biodegradation, either in culture media or inside cells, and exert their biological effects primarily as intact molecules.


Subject(s)
Anticarcinogenic Agents/pharmacology , Genistein/analogs & derivatives , Genistein/pharmacology , Glycosides , Isoflavones/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Division/drug effects , Humans , Neoplasms/drug therapy , Soybean Proteins , Tumor Cells, Cultured
11.
Nucl Med Biol ; 29(2): 169-75, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11823121

ABSTRACT

Cisplatin chemotherapy in combination with external irradiation or with low-dose continuous internal radiotherapy produces significant supra-additive treatment effects towards several tumor cells. The purpose of our research is to develop a new class of platinum-based anticancer drugs containing moieties of synergistic potency such as platinum core and a radiotherapeutic isotope which, delivered directly to the tumorous cells by a specifically designed vectors, should produce a local enhancement of therapeutic dose. Thus, we have synthesized a new platinum-iodohistamine complex and its radioactive analogues labeled with I-125 and I-131. In the present study some biological properties of those compounds have been investigated. The in vitro screening study pointed out that non-radioactive platinum-iodohistamine complex possesses high cytostatic activity against COLO-205 cells, and moderate activity against HL-60 cell line. No cytotoxicity was observed against MOLT-4 and L-1210 cells, as well as against VERO normal cells. The biodistribution of intravenously administered radioactive platinum-[131I]-iodohistamine complex to normal rats revealed the highest accumulation in the liver (c.a. 40%ID). Intraperitoneal injections of the complex to tumor-bearing C3H mice resulted in scattering of the dose in the organs (mainly in GIT, liver, kidney). The retention of radioactive complex in neoplastic tissue was 3-4 times higher than in normal muscular tissue, although exhibited the tendency to decrease with time post injection. The results of the present study show promising features of the newly developed platinum-iodohistamine complexes and justify prospective investigation of in vivo anticancer potency on animal models of solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Histamine/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Organoplatinum Compounds/pharmacology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Antineoplastic Agents/pharmacokinetics , Cell Cycle/drug effects , Cell Cycle/radiation effects , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Drug Synergism , Female , Flow Cytometry , Histamine/analogs & derivatives , Histamine/pharmacokinetics , Humans , Injections, Intraperitoneal , Injections, Intravenous , Leukemia L1210/therapy , Male , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred C3H , Organoplatinum Compounds/pharmacokinetics , Radiotherapy, Adjuvant/methods , Rats , Rats, Wistar , Tissue Distribution , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...