Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Bull Exp Biol Med ; 158(4): 497-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25708334

ABSTRACT

Blood levels of vascular endothelial growth factor, the main marker of angiogenesis activity, and coagulation hemostasis were studied in CDF1 mice with transplanted P-388 lymphocytic leukemia. Blood levels of vascular endothelial growth factor increased by 168% in mice with tumors. The animals developed the hypercoagulation syndrome manifesting in shorter activated partial thromboplastin time and prothrombin time and development of hyperfibrinogenemia.


Subject(s)
Blood Coagulation Disorders/physiopathology , Leukemia P388/physiopathology , Neovascularization, Pathologic/physiopathology , Vascular Endothelial Growth Factor A/blood , Animals , Enzyme-Linked Immunosorbent Assay , Male , Mice , Neoplasm Transplantation/physiology , Neovascularization, Pathologic/blood , Partial Thromboplastin Time , Prothrombin Time , Statistics, Nonparametric
2.
Bull Exp Biol Med ; 147(4): 421-3, 2009 Apr.
Article in English, Russian | MEDLINE | ID: mdl-19704938

ABSTRACT

Exogenous NO donor 3,3-bis-(nitroxymethyl)oxetane (NMO) was synthesized at the Institute for Problems of Chemical Physics (Russian Academy of Sciences). This compound was shown to inhibit cell death (apoptosis and necrosis) in cyclophosphamide-sensitive and cyclophosphamide-resistant P388 murine tumor. p53 protein was expressed in both lines of tumor cells. NO donor NMO had little effect on p53 protein expression in cells of both stains. Our results suggest that the proapoptotic effect of NMO is mediated by the p53-independent molecular mechanisms.


Subject(s)
Apoptosis/drug effects , Ethers, Cyclic/pharmacology , Leukemia P388/drug therapy , Nitric Oxide Donors/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/physiology , Cell Count , Cell Line, Tumor , Cyclophosphamide/pharmacology , Drug Resistance, Neoplasm , Leukemia P388/pathology , Leukemia P388/physiopathology , Mice , Necrosis/drug therapy , Necrosis/physiopathology , Time Factors
3.
Proc Natl Acad Sci U S A ; 94(17): 9394-9, 1997 Aug 19.
Article in English | MEDLINE | ID: mdl-9256493

ABSTRACT

As previously reported, Listeria monocytogenes infection of P388D1 macrophages results in a rapid induction of NF-kappaB DNA-binding activity. Here we show that this induction of NF-kappaB activity occurs in a biphasic mode: first, a transient, IkappaBalpha degradation-dependent phase of activity, also induced by the nonvirulent species Listeria innocua, which is mediated by binding of the bacteria to the macrophage, or by adding Listeria-derived lipoteichoic acid to the macrophage; the second persistent phase of activation is only markedly induced when the bacteria enter the cytoplasm of the host cell and express the virulence genes plcA and plcB, encoding two phospholipases. We suggest that products of the enzymatic activity of phospholipases directly interfere with host cell signal transduction pathways, thus leading to persistent NF-kappaB activation via persistent IkappaBbeta degradation.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , Leukemia P388/physiopathology , Lipopolysaccharides/pharmacology , Listeria monocytogenes , Listeriosis/metabolism , Macrophages/microbiology , NF-kappa B/metabolism , Phospholipases/pharmacology , Teichoic Acids/pharmacology , Animals , Humans , Macrophages/metabolism , Mice , NF-KappaB Inhibitor alpha
4.
C R Acad Sci III ; 320(1): 13-8, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9099259

ABSTRACT

In order to study the role of Fos on the regulation of proliferation in the monocyte-macrophage lineage we realized a stable transfection of the murine P388D1 cell line by the murine c-fos gene under the control of the human metallothionein IIA promoter. Several clones have been selected by geneticin: they show a variable number of integrated transgene (two to ten copies). Their expression has been analyzed in the presence or absence of cadmium chloride as inducer (5 x 10(-6) M). In one clone especially, the c-fos mRNA and Fos protein levels were respectively 6- and 10-fold increased. The study of cell growth by tritiated thymidine incorporation indicates a negative effect of the overexpressed Fos protein in the absence of any other stimulus.


Subject(s)
Genes, fos/genetics , Macrophages/metabolism , Animals , Cadmium Chloride/pharmacology , Cell Division/drug effects , Clone Cells , Gene Expression/drug effects , Leukemia P388/physiopathology , Mice , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/pharmacology , RNA, Messenger/genetics , Transfection
5.
Life Sci ; 60(15): 1239-48, 1997.
Article in English | MEDLINE | ID: mdl-9096241

ABSTRACT

It has been found that certain antineoplastic drugs impart their function with a distinct duality. Besides being tumoricidal, they are capable of acting as immunopotentiator. This led us to investigate the effect of cytosine arabinoside (CA), vincristine sulphate (VS), cyclophosphamide (CS), mitomycin C (MMC), hydroxy urea (HU) and lipopolysaccharide (LPS) on a macrophage cell line P388D1. Supernatants collected from P388D1 cells treated with CA, VS, CS, MMC, HU or LPS demonstrated enhanced production of tumor necrosis factor (TNF) confirmed by bioassay on L929 tumor target cells and increased interleukin-1 (IL-1) production by standard thymocyte proliferation bioassay. Also, supernatants showed increased amounts of nitric oxide and lysozyme using Griess reaction and reduction in turbidity of Micrococcus lysodeikticus, respectively. The above findings demonstrate that these drugs may be used not only as chemotherapeutic agents but also as macrophage-activating agents.


Subject(s)
Antineoplastic Agents/pharmacology , Hydroxyurea/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/metabolism , Animals , Cell Line , Cell Survival , Interleukin-1/biosynthesis , Leukemia P388/physiopathology , Mice , Mice, Inbred DBA , Muramidase/biosynthesis , Nitric Oxide/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
6.
Cancer Res ; 56(18): 4171-9, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8797588

ABSTRACT

Overexpression of P-glycoprotein (Pgp) by tumors results in multidrug resistance (MDR) to structurally unrelated oncolytics. MDR cells may be sensitized to these oncolytics when treated with a Pgp modulator. The present study evaluates LY335979 as a modulator both in vitro and in vivo. LY335979 (0.1 microM) fully restored sensitivity to vinblastine, doxorubicin (Dox), etoposide, and Taxol in CEM/VLB100 cells. LY335979 modulated Dox cytotoxicity even when LY335979 (0.5 microM) was removed 24 h prior to the cytotoxicity assay. LY335979 blocked [3H]azidopine photoaffinity labeling of the M(r) approximately 170,000 Pgp in CEM/VLB100 plasma membranes and competitively inhibited equilibrium binding of [3H]vinblastine to Pgp (Ki of approximately 0.06 microM). Treatment of mice bearing P388/ADR murine leukemia cells with LY335979 in combination with Dox or etoposide gave a significant increase in life span with no apparent alteration of pharmacokinetics. LY335979 also enhanced the antitumor activity of Taxol in a MDR human non-small cell lung carcinoma nude mouse xenograft model. Thus, LY335979 is an extremely potent, efficacious modulator that apparently lacks pharmacokinetic interactions with coadministered anticancer drugs and is, therefore, an exciting new agent for clinical evaluation for reversal of Pgp-associated MDR.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Dibenzocycloheptenes/pharmacology , Drug Resistance, Multiple , Etoposide/toxicity , Leukemia P388/drug therapy , Leukemia P388/physiopathology , Lung Neoplasms/drug therapy , Paclitaxel/therapeutic use , Quinolines/pharmacology , Vinblastine/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Cell Survival/drug effects , Dibenzocycloheptenes/therapeutic use , Etoposide/metabolism , Etoposide/therapeutic use , Humans , Kinetics , Mice , Mice, Nude , Protein Binding , Quinolines/therapeutic use , Transplantation, Heterologous , Tumor Cells, Cultured , Vinblastine/metabolism , Vinblastine/therapeutic use
7.
Mol Pharmacol ; 49(5): 832-41, 1996 May.
Article in English | MEDLINE | ID: mdl-8622633

ABSTRACT

We studied the effect of doxorubicin (Dox) on cell cycle progression and its correlation with DNA damage and cytotoxicity in p53-mutant P388 cells. P388 cells synchronized in S and G2/M phases were > 3-fold more sensitive to Dox than were cells in G1 phase (Dox ID50 = 0.50 +/- 0.16 microM in cells synchronized in S phase versus 1.64 +/- 0.12 microM in asynchronized cells; drug exposure, 1 hr). Treatment of synchronized cells in early S phase with 1 microM Dox (2 x ID50) for 1 hr induced a marked cell arrest at G2/M phase at 6-12 hr after drug incubation. We then studied the effect of Dox on the p34cdc2/cyclin B1 complex because it plays a key role in regulating G2/M phase transition. In untreated control P388 cells, p34cdc2 kinase localizes in the nucleus and cytoplasms, particularly in the centrosomes, and p34cdc2 kinase activity is dependent on cell cycle progression, with the enzyme activity increasing steadily from G1/S to G2/M and markedly declining thereafter. Treatment of synchronized P388 cells in early S phase with 1 microM Dox for 1 hr did not affect the pattern of subcellular distribution of the enzyme but completely abrogated its function for > or = 10 hr. In a cell-free system, Dox did not inhibit p34cdc2 kinase activity, indicating that is has no direct effect on the enzyme function. In whole cells, Dox treatment prevented p34cdc2 kinase dephosphorylation without altering its synthesis, and this effect was due to neither down-regulation of cdc25C nor inhibition of protein-tyrosine phosphatase activity. In contrast, Dox treatment was found to induced cyclin B1 accumulation as a result of stimulating its synthesis and inhibiting its degradation. A good correlation was found between extent of DNA double-strand breaks and p34cdc2 kinase activity inhibition. Our results suggest that anthracycline-induced cytotoxicity is cell cycle dependent and is mediated, at least in part, by disturbance of the regulation of p34cdc2/cyclin B1 complex, thus leading to G2/M phase arrest.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Cyclin B , Cyclins/metabolism , Doxorubicin/pharmacology , Leukemia P388/physiopathology , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Compartmentation/drug effects , Cell Survival/drug effects , Cyclin B1 , Mice , Phosphorylation
8.
Eksp Klin Farmakol ; 58(3): 48-50, 1995.
Article in Russian | MEDLINE | ID: mdl-7663298

ABSTRACT

The thymuses and spleens from healthy and tumor-bearing (leukemia P 388 and Lewis' carcinoma) were morphometrically studied after a single administration of the antitumor agent nitrosomethylurea in doses of 96 and 4 mg/kg. When used both in the high and low doses, nitrosomethylurea had an immunomodulating effect, causing the activation of cell-mediated immunity in healthy and tumor-bearing (in early malignancy) animals when given in the small dose.


Subject(s)
Adjuvants, Immunologic/pharmacology , Methylnitrosourea/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/physiopathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Immunity, Cellular/drug effects , Leukemia P388/drug therapy , Leukemia P388/immunology , Leukemia P388/physiopathology , Male , Methylnitrosourea/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation , Spleen/drug effects , Spleen/immunology , Spleen/physiopathology , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/physiopathology , Time Factors
9.
Cancer Res ; 54(3): 730-7, 1994 Feb 01.
Article in English | MEDLINE | ID: mdl-7905786

ABSTRACT

Cross-resistance to chemotherapeutic drugs is a significant problem in the treatment of patients with cancer. The discovery that this phenomenon is associated with the overexpression of a membrane glycoprotein, P-glycoprotein, which acts as a drug efflux pump, has provided a new target for drug development. To develop a model for identifying new compounds which can block the function of P-glycoprotein, we infected P388 mouse leukemic cells with a retrovirus containing a cloned human MDR1 complementary DNA. The new cell line, P388/VMDRC.04, incorporated and overexpressed the human gene as evidenced by Southern blots, increased mRNA and protein synthesis, and recognition by the MRK16 monoclonal antibody. P388/VMDRC.04 was cross-resistant to colchicine, vincristine, and doxorubicin, and the degree of resistance correlated with a reduction in cellular drug accumulation. Unlike many cell lines selected for resistance by growth in increasing concentrations of drug for prolonged periods of time, these cells did not show alternative mechanisms of resistance such as increased synthesis of glutathione or alterations in topoisomerase II. In addition, the sensitivity of P388/VMDRC.04 cells was completely restored by cyclosporin A and trans-flupenthixol. P388/VMDRC.04 cells were subcloned and 10 clones were picked for in vivo evaluation. One subclone grew similarly to parental cells in female BALB/c x DBA/2 F1 mice and showed no responsiveness to therapeutic doses of vincristine or etoposide. The combination of vincristine with cyclosporin A significantly increased the survival of mice inoculated with P388/VMDRC.04 cells. The availability of a cell line that displays the MDR phenotype, overexpresses human P-glycoprotein, but does not contain alterations in at least two well-defined alternative mechanisms of resistance, and that can be grown in simple animal models should facilitate the development of new agents active against this form of chemotherapeutic drug resistance.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Drug Resistance/physiology , Leukemia P388/drug therapy , Leukemia P388/physiopathology , Membrane Glycoproteins/antagonists & inhibitors , Models, Biological , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Animals , Antineoplastic Agents/pharmacology , Carrier Proteins/physiology , Cell Division/drug effects , Cell Line, Transformed , DNA, Complementary/genetics , Drug Resistance/genetics , Drug Screening Assays, Antitumor , Female , Flupenthixol/pharmacology , Genetic Vectors/genetics , Humans , Leukemia P388/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , RNA, Messenger/genetics , Retroviridae/genetics , Sensitivity and Specificity , Terfenadine/pharmacology , Transformation, Genetic/genetics
10.
Cancer Lett ; 76(2-3): 139-45, 1994 Jan 30.
Article in English | MEDLINE | ID: mdl-7511983

ABSTRACT

Calphostin C is a potent and specific inhibitor of protein kinase C (PKC). In this investigation we examined the effect of Calphostin C (without prior exposure to light) on daunorubicin (DNR) accumulation and sensitivity to DNR in multidrug-resistant (MDR) murine leukemia P388/ADR and human myeloid leukemia HL60/AR cells. P388/ADR cells overexpress P-glycoprotein, whereas HL60/AR cells lack any expression of P-glycoprotein (both at mRNA and protein levels). Calphostin C, in a concentration-dependent manner, increased the accumulation of DNR in P388/ADR cells and partially reversed (threefold) the DNR resistance in P388/ADR cells but had no effect on either of the parameters in HL60/AR cells. Calphostin C-induced increased accumulation of DNR in P388/ADR cells was due to increased uptake and decreased efflux of DNR. Furthermore, Calphostin C increased the uptake and decreased the efflux of rhodamine 123 (a substrate for P-gp) in P388/ADR cells but had no such effect in P388 cells. In addition, Calphostin C without exposure to light did not inhibit PKC activity in any of the cell lines studied. Taken together, these data suggest that Calphostin C may reverse drug resistance via P-glycoprotein independently of its effect on PKC activity. Therefore, any data regarding the effect of Calphostin C on the reversal of MDR should be interpreted in the light of these findings.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Carrier Proteins/physiology , Daunorubicin/pharmacology , Leukemia P388/drug therapy , Leukemia, Experimental/drug therapy , Leukemia, Myeloid/drug therapy , Membrane Glycoproteins/physiology , Naphthalenes , Polycyclic Compounds/pharmacology , Protein Kinase C/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Animals , Coloring Agents/pharmacokinetics , Daunorubicin/pharmacokinetics , Drug Resistance , Drug Screening Assays, Antitumor , Humans , Leukemia P388/metabolism , Leukemia P388/physiopathology , Leukemia, Experimental/metabolism , Leukemia, Experimental/physiopathology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/physiopathology , Mice , Protein Kinase C/metabolism , Rhodamine 123 , Rhodamines/pharmacokinetics , Tumor Cells, Cultured/drug effects
11.
Biull Eksp Biol Med ; 114(11): 518-9, 1992 Nov.
Article in Russian | MEDLINE | ID: mdl-1290828

ABSTRACT

The study was performed to investigate the effect of ascitic fluid globulins of tumor on tumor growth and life span of mice. The globulins are shown to shorten the life span of Ehrlich tumor mice from 86.8 to 61.8 days, to increase 3-5-fold the growth rate of Ehrlich carcinoma and P388/DOX tumor. It was found that globulins of ascitic fluids and serum globulins of tumor have equal effects of tumor growth. It is proposed to use globulins of ascitic fluid to study the globulin role in tumor growth.


Subject(s)
Ascitic Fluid/physiopathology , Carcinoma, Ehrlich Tumor/physiopathology , Globulins/pharmacology , Leukemia P388/physiopathology , Animals , Carcinoma, Ehrlich Tumor/blood , Carcinoma, Ehrlich Tumor/mortality , Globulins/isolation & purification , Leukemia P388/blood , Leukemia P388/mortality , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Neoplasm Transplantation , Serum Globulins/isolation & purification , Serum Globulins/pharmacology , Time Factors
12.
Cancer Res ; 51(16): 4187-91, 1991 Aug 15.
Article in English | MEDLINE | ID: mdl-1651156

ABSTRACT

It is known that 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11), a semisynthesized derivative of camptothecin (CPT), has a potent antitumor activity in vivo, but 7-ethyl-10-hydroxycamptothecin (SN-38), a metabolite of CPT-11, shows much stronger cytotoxicity in vitro than CPT-11. In this study, we demonstrated that the relaxation of SV40 DNA plasmids by type I DNA topoisomerase prepared from P388 murine leukemia cells was inhibited by 50% by SN-38 at approximately 1 microM, although CPT-11 at 1 mM slightly inhibited the relaxation. SN-38 and CPT showed strong, time-dependent inhibitory activity against DNA synthesis of P388 cells. However, CPT-11 weakly inhibited DNA synthesis independently of time with coincident inhibition of the total thymidine uptake by the cells. By alkaline and neutral elution assays, it was demonstrated that SN-38 caused much more frequent DNA single-strand breaks in P388 cells than did CPT-11. The same content of SN-38 and a similar frequency of single-strand breaks were detected in the cells treated with SN-38 at 0.1 microM or with CPT-11 at 100 microM. Therefore, single-strand breaks by CPT-11 seem to be due to SN-38 produced from CPT-11 in cells. These results indicate that CPT-11 itself possesses a marginal antiproliferative effect but that SN-38 plays an essential role in the mechanism of action of CPT-11.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Cell Survival/drug effects , DNA Damage , DNA Replication/drug effects , Topoisomerase I Inhibitors , Animals , Camptothecin/metabolism , Camptothecin/pharmacology , DNA, Single-Stranded/drug effects , Irinotecan , Kinetics , Leukemia P388/pathology , Leukemia P388/physiopathology , Mice , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/drug effects , Thymidine/metabolism , Uridine/metabolism
13.
Prostaglandins ; 41(5): 501-13, 1991 May.
Article in English | MEDLINE | ID: mdl-1862229

ABSTRACT

Hyperthermia-induced cell lethality is thought to be mediated through injury to the cell membrane. Membrane perturbation results in the release of prostaglandins (PG) and leukotrienes (LT). These compounds are potent biological mediators and may modify the tumor microenvironment and therapeutic efficacy. Membrane composition and PG/LT release are influenced by the dietary fatty acids. The relationship between these variables and response to hyperthermia was examined in vitro using murine P388 leukemia cells grown as an ascites in mice provided either saturated fatty acid diet (SFA; 16% beef tallow) or unsaturated fatty acid diet (UFA; 16% safflower oil). Cells were harvested and exposed in vitro to either 37 degrees C or 43.5 degrees C for periods up to 2 hours. Hyperthermic exposure for 2 hours resulted in 40% cell lethality in SFA cells and 55% in UFA cells. The phospholipid and total cholesterol content was higher (33% and 50% respectively) in the UFA versus the SFA cells. Hyperthermia produced a six-fold increase in prostaglandin E2 PGE2 release by SFA cells and a 4.5-fold increase by UFA cells. No LTC4 was detected. Alteration of dietary fat affects cell lethality and PG release following hyperthermic treatment. The increase in phospholipid and cholesterol content of UFA cells may be a response to reduced membrane fluidity.


Subject(s)
Cell Survival , Dietary Fats/pharmacology , Dinoprostone/metabolism , Fatty Acids, Unsaturated/pharmacology , Hot Temperature , Leukemia P388/physiopathology , Animals , Cell Membrane/physiology , Cell Survival/drug effects , Cholesterol/analysis , Female , Leukemia P388/pathology , Mice , Mice, Inbred Strains , Phospholipids/analysis
14.
Arch Pharm (Weinheim) ; 324(3): 153-60, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1859250

ABSTRACT

The combined effects of amino acid antagonists with proven or potential inhibitory activities on aminoacyl-tRNA synthetases were investigated on the murine leukemic cell line P388 D1. As the best result a summation of the antiproliferative effects was observed. Combinations with established cytostatic agents like platinum complexes or other antitumor compounds also yielded partly additive effects. In experiments performed with asparaginase L-aspartic acid-beta-hydroxamate gave synergistic growth inhibition of P388 D1 cells in vitro, which was reflected by additive effects against murine leukemia P388 in vivo.


Subject(s)
Amino Acids/antagonists & inhibitors , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Cell Division/drug effects , Animals , Antineoplastic Agents/pharmacology , Drug Interactions , Leukemia P388/physiopathology , Mice
15.
Res Commun Chem Pathol Pharmacol ; 71(2): 235-8, 1991 Feb.
Article in English | MEDLINE | ID: mdl-2047569

ABSTRACT

The effect of cepharanthine on the cytotoxicity of doxorubicin (DOX) in p388 leukemia cells was studied in vitro and in vivo. Nontoxic levels of cepharanthine enhanced the sensitivity to doxorubicin in p388 cells in vitro. The potentiation of antitumor activity of DOX by cepharanthine against mice bearing p388 cells was also observed.


Subject(s)
Alkaloids/toxicity , Doxorubicin/toxicity , Leukemia P388/drug therapy , Tumor Cells, Cultured/drug effects , Animals , Benzylisoquinolines , Female , Leukemia P388/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation
16.
Article in English | MEDLINE | ID: mdl-2895540

ABSTRACT

The P388 lymphocytic leukemia and the L1210 lymphoid leukemia are used as test systems for putative cytotoxic drugs. These leukemias are also used to investigate the perturbation of cell cycle progression of various chemical compounds in more detail. There is little information on the normal growth kinetics in vivo of these leukemias. In the present report we therefore present the results from growth kinetic studies of P388 and L1210 leukemic cells growing in ascites form in mice. We used 3H-TdR autoradiography, DNA flow cytometry and the stathmokinetic method. During exponential growth both leukemias showed a growth fraction of unity. Whereas no significant cell loss was observed during the early growth phase of P388 cells, cell loss was indicated by a discrepancy between potential and actual doubling times during exponential growth of L1210 cells. During the phase of growth retardation, the proportion of G1 and G2 cells increased at the expence of a reduced S phase fraction in the P388 leukemia, whereas only small changes in cell cycle distributions were seen with time after inoculation of L1210 cells. An increasing discrepancy in the reduction of the S phase fraction and the 3H-TdRLI was seen in the P388 cells with time after inoculation. Thus, a majority of P388 cells with S phase DNA content were unlabelled during the late phase of growth restriction, indicating resting cells in S phase. A good correlation was found between the 3H-TdR LI and S phase fraction throughout the life history of L1210 cells, revealing considerable differences in in vivo growth kinetics between the two leukemias. Such differences should be considered when evaluating test results.


Subject(s)
Leukemia L1210/metabolism , Leukemia P388/metabolism , Leukemia, Experimental/metabolism , Animals , Autoradiography , Cell Cycle , Cell Line , DNA/analysis , Flow Cytometry , Growth , Kinetics , Leukemia L1210/physiopathology , Leukemia P388/physiopathology , Mice , Tritium
17.
In Vivo ; 1(1): 47-52, 1987.
Article in English | MEDLINE | ID: mdl-2979764

ABSTRACT

A line of P388 leukemia resistant to mitomycin C (MMC) was successfully developed in vivo by treating mice bearing parental P388 (P388/0) with MMC followed by serial passage of the surviving leukemic cells. From this P388/MMC line, a subline was derived by not treating the passage mice with MMC (P388/MMC-NP); resistance to MMC was stable for as many as 56 weeks of transplantation. The chemosensitivities of each P388 line to assorted anticancer drugs were compared in vivo. Both P388/MMC and P388/MMC-NP had similar patterns of drug cross-resistance and collateral sensitivity. With respect to alkylating agents (e.g. cyclophosphamide, Platinol and chlorambucil), there was generally a partial degree of cross-resistance, sometimes only detectable at suboptimal dose levels. With respect to DNA binders or intercalators (e.g. actinomycin D, luzopeptin A, amsacrine, doxorubicin), the extent of cross-resistance varied from none (dihydoxyanthraquinone) to marked (doxorubicin). Antimitotic inhibitors (vinblastine and vincristine) were completely cross-resistant, as were some miscellaneous natural agents (rebeccamycin, VP-16, sesbanimide, and elsamicin, a chartreusin analog). Antimetabolites (e.g. methotrexate and 6-thioguanine) showed no cross-resistance and even demonstrated some occasional evidence of collateral effectiveness.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance , Leukemia P388/drug therapy , Mitomycin/therapeutic use , Animals , Leukemia P388/physiopathology , Mice , Mice, Inbred DBA , Mice, Inbred Strains
18.
Cancer Res ; 46(9): 4352-6, 1986 Sep.
Article in English | MEDLINE | ID: mdl-3460695

ABSTRACT

A series of anthrapyrazoles was examined for their cytotoxic effect on P388 cells resistant (P388R) to anthracyclines, N-[4-(9-acridinylamino)-3-methoxyphenyl] methanesulfonamide, trimetrexate, and vinblastine. The degree of resistance of P388R cells to Adriamycin (ADR) and daunomycin was 50-fold and 38-fold, respectively, when compared to the parent cell line (P388S). The Adriamycin-resistant cells were highly cross-resistant to some anthrapyrazoles, but the degree of cross-resistance was not uniform and was less than 3-fold for one member of the series. The lipophilicity of these compounds appeared to correlate to some extent with the level of resistance. The calcium channel blockers verapamil (VER) and diltiazem and the calmodulin antagonist trifluoperazine potentiated the cytotoxicity of the anthrapyrazoles and ADR in P388R. This potentiating effect was concentration dependent with VER being the most efficacious. VER increased ADR cytotoxicity by greater than 10-fold and CI-937 by almost 40-fold. However, VER, diltiazem, and trifluoperazine had no effect on ADR or anthrapyrazole activity in P388S cells. The antiarrhythmic drug, quinidine, and the detergent, Tween 80, also potentiated ADR activity in P388R cells to the same extent as VER. Both the net accumulation and efflux of [3H]daunomycin were altered in P388R cells by nontoxic concentrations of Tween 80 in a fashion virtually identical to that demonstrated for VER. These data suggest that agents which potentiate drug cytotoxicity in P388R cells may do so by their interaction with the lipid domain of the plasma membrane. In addition, these results demonstrate that some members of the new series of DNA binding drugs, the anthrapyrazoles, may be active against anthracycline-resistant tumors and that, where cross-resistance to them occurs, it can be partially reversed by agents such as VER.


Subject(s)
Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calmodulin/antagonists & inhibitors , Drug Resistance/drug effects , Leukemia P388/physiopathology , Leukemia, Experimental/physiopathology , Pyrazoles/pharmacology , Animals , Antibiotics, Antineoplastic , Biological Transport/drug effects , Cell Line , Daunorubicin/metabolism , Mice , Naphthacenes/pharmacology , Polysorbates/pharmacology , Quinidine/pharmacology , Solubility , Structure-Activity Relationship
19.
Eur J Cancer Clin Oncol ; 22(1): 45-52, 1986 Jan.
Article in English | MEDLINE | ID: mdl-3754212

ABSTRACT

Daphnane diterpene esters have previously been shown to have antineoplastic activity in vivo against the growth of P-388 lymphocytic leukemia cells. These studies demonstrate cytotoxic activity of genkwadaphnin and gnidilatidin against P-388 lymphocytic leukemia, L-1210 lymphoid leukemia and human KB carcinoma cell growth in vitro. At the ED50 values in the respective tumor lines DNA synthesis was preferentially suppressed in all three cell lines. RNA synthesis was essentially unaffected by the agents. Protein synthesis inhibition by the two agents demonstrated selectivity, e.g. in P-388 cells significant inhibition, in L-1210 cells marginal inhibition and in KB cells no inhibition was observed at these concentrations. Multiple sites in DNA synthesis were found to be inhibited by the daphnane diterpene esters. Two to three times the ED50 concentration in the respective tumor lines was required to observe suppression of DNA synthesis. Purine de novo synthesis appeared to be the major site of inhibition, with inosine monophosphate dehydrogenase and phosphoribosyl pyrophosphate amido transferase activities being inhibited in all three tissue lines. Dihydrofolate reductase activity was inhibited, significant only in the P-388 and KB cells. The magnitude of the enzyme suppression by the agents varied with the tumor line. However, the degree of enzyme suppression was of sufficient magnitude to account for the observed purine and DNA synthesis inhibition by the daphnane diterpene esters.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Diterpenes/pharmacology , KB Cells/metabolism , Leukemia L1210/physiopathology , Leukemia P388/physiopathology , Leukemia, Experimental/physiopathology , Animals , Cell Survival/drug effects , Cells, Cultured , DNA, Neoplasm/biosynthesis , Humans , Leukemia L1210/metabolism , Leukemia P388/metabolism , Metaphase , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/biosynthesis , Tetrahydrofolate Dehydrogenase/biosynthesis
20.
J Natl Cancer Inst ; 73(4): 959-62, 1984 Oct.
Article in English | MEDLINE | ID: mdl-6592390

ABSTRACT

Macrophage-like P388D1 cells, when stimulated with lipopolysaccharide (LPS), produced a factor(s) whose activity was inhibitory to growth of Lewis lung carcinoma cells. Indomethacin, a prostaglandin-synthesis inhibitor, augmented the production of tumoristatic activity by LPS-stimulated P388D1 cells, whereas exogenous prostaglandin E2 (PGE2) suppressed its secretion. These results suggest that P388D1 cells regulate their antitumor abilities by producing PGE2, which inhibits production of soluble factor(s) with tumoristatic activities.


Subject(s)
Antineoplastic Agents/isolation & purification , Leukemia P388/physiopathology , Leukemia, Experimental/physiopathology , Macrophages/physiology , Prostaglandins E/physiology , Animals , Antineoplastic Agents/toxicity , Cell Line , DNA Replication/drug effects , Dinoprostone , Indomethacin/pharmacology , Lipopolysaccharides/pharmacology , Lung Neoplasms/pathology , Macrophages/drug effects , Mice , Prostaglandins E/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...