Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pigment Cell Res ; 4(2): 80-6, 1991 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1946213

RESUMO

Melanin contains melanin-free radicals and can both absorb and produce additional free radicals and active oxygen species on exposure to various stimuli. Yet its role in the radiation responses of malignant melanoma has been little studied. In this report, three subclones of Cloudman S91 mouse melanoma clone PC1A varying in constitutive melanin content were compared with respect to killing by gamma irradiation. Radiation responses correlated with melanin content. The least melanotic line, S91/amel, was most sensitive and the most melanotic line, S91/I3, was most resistant. Curve fitting using the linear-quadratic model suggests that S91/amel is killed only by single event inactivations; S91/I3, only by double event inactivations; and S91/M1B, with intermediate melanin and radiation response, by both types of inactivations. Split dose experiments confirmed a lack of immediate split dose recovery in S91/amel and its existence in S91/I3. Potentially lethal damage and its repair could be demonstrated in both S91/amel and S91/I3. Double strand break (DSB) induction was evaluated as a function of gamma ray dose in DNA of S91/I3 and S91/amel, as well as in EMT6, a mouse mammary cancer line that lacks tyrosinase and melanin. The rates of induction were proportional to cellular melanization, i.e., the rate of DSB induction was greatest in S91/I3, least in EMT6. Levels of thioredoxin reductase (TR), glutathione reductase (GR), superoxide dismutase (SOD), and catalase (CAT) were determined in S91/amel and S91/I3. TR was the same in both cell lines, while the other three enzymes were 3- to 4-fold lower in S91/amel.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Sobrevivência Celular/efeitos da radiação , Raios gama/efeitos adversos , Melaninas/metabolismo , Melanoma/patologia , Animais , Antioxidantes/metabolismo , Catalase/análise , Sobrevivência Celular/fisiologia , DNA/efeitos da radiação , Melaninas/análise , Camundongos , Radiação Ionizante , Superóxido Dismutase/análise , Tiorredoxinas/análise , Células Tumorais Cultivadas
2.
Cancer Chemother Pharmacol ; 26(6): 423-8, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2171796

RESUMO

Four drugs known to interact with topoisomerase II were assessed for their ability to enhance the cytotoxicity of cis-diamminedichloroplatinum(II) (CDDP) in Chinese hamster ovary (CHO) cell lines sensitive and resistant to VM-26. The combination treatments were analyzed by isobologram methodology. On 24 h exposure, there was no significant difference in the cytotoxicity of novobiocin or ciprofloxacin toward either cell line. The resistant cells were approximately 9-fold more resistant to 4'-(9-acridinylamino)methanesulfon-m-anisidide (m-AMSA) and approximately 170-fold more resistant to etoposide after a 24-h exposure. The combination of novobiocin and cisplatin produced greater than additive cell kill over the entire dose range of cisplatin tested in both cell lines. m-AMSA and CDDP produced cell kill that fell within the envelope of additivity. Etoposide and CDDP resulted in cytotoxicity that was slightly greater than additive at low CDDP concentrations and additive at the highest concentration of CDDP tested in the parental cell line and was slightly greater than additive in the resistant cell line. Ciprofloxacin and CDDP, like novobiocin, resulted in greater than additive cell kill in both cell lines. The enhancement of CDDP cytotoxicity by novobiocin that was seen in exponentially growing cells was lost in stationary-phase cultures. In these studies, novobiocin and, to a lesser degree, ciprofloxacin produced greater than additive cell kill in combination with CDDP in parental and epipodophyllotoxin-resistant CHO cells.


Assuntos
Cisplatino/toxicidade , Ovário/efeitos dos fármacos , Podofilotoxina/antagonistas & inibidores , Inibidores da Topoisomerase II , Animais , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Sinergismo Farmacológico , Feminino
3.
Cancer Res ; 49(3): 595-8, 1989 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-2910481

RESUMO

Our previous in vitro studies demonstrated marked synergy with alkylating agents when novobiocin was present during and after alkylating agent exposure. To determine whether this effect is observed in vivo, novobiocin was administered daily for 3 days prior to alkylating agent treatment, during alkylating agent treatment, and for 2 days after completion of alkylating agent treatment. When combined with cis-diamminedichloroplatinum(II), 1,3-bis(2-chloroethyl)-1-nitrosourea, or cyclophosphamide, there was significant enhancement of the growth delay of the FSaIIC fibrosarcoma implanted s.c. in C3H mice when compared with alkylating agents alone. In a second assay using ex vivo studies of tumor cells exposed in vivo, single doses of 100 mg/kg of novobiocin followed by cis-diamminedichloroplatinum(II) resulted in a 3- to 4-fold increase in tumor cell killing by cis-diamminedichloroplatinum(II). At a dose of 100 mg/kg of 1,3-bis(2-chloroethyl)-1-nitrosourea there was about a 7-fold increase in tumor cell kill upon addition of novobiocin. Cyclophosphamide showed a dose response effect with novobiocin, reaching 13-fold at a dose of 300 mg/kg of cyclophosphamide. In all cases bone marrow elements were affected less than were neoplastic cells, suggesting that the combination of novobiocin and alkylating agents may be a clinically useful strategy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Medula Óssea/efeitos dos fármacos , Fibrossarcoma/tratamento farmacológico , Novobiocina/farmacologia , Alquilantes/administração & dosagem , Alquilantes/farmacocinética , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carmustina/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Ciclofosfamida/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Novobiocina/administração & dosagem , Compostos Organoplatínicos/administração & dosagem
4.
Cancer Res ; 48(22): 6417-23, 1988 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-3180059

RESUMO

Dose-response studies were performed with the alkylating agents [nitrogen mustard, N,N'-bis(2-chloroethyl)-N-nitrosourea, melphalan, cisplatin (CDDP), 4-hydroperoxycyclophosphamide (4-HC), and trimethyleneiminethiophosphoramide] in both the MCF-7 human breast carcinoma cell line and the EMT6 and FSaIIC murine tumor lines. Increasing selection pressure with the alkylating agents CDDP, melphalan, and 4-HC in vitro produced low levels (6.5- to 9-fold) of drug resistance, despite an intensive and prolonged treatment program. The MCF-7 sublines made resistant to CDDP and 4-HC did not exhibit cross-resistance to other alkylating agents; however, the MCF-7 subline resistant to melphalan was partially cross-resistant to nitrogen mustard, 4-HC, and CDDP. A log-linear relationship was maintained between surviving fraction of MCF-7 cells in culture and drug concentration with alkylating agents, whereas for nonalkylating agents the survival curves tended to plateau at high drug concentrations. Log-linear tumor cell kill was also obtained over a wide dosage range with several alkylating agents in murine tumors treated in vivo. Tumor cell survival assay by colony formation indicated the continuing importance of dose in the action of the drugs even at high levels of tumor cell kill. With some agents, there was a difference between the slopes of the tumor cell killing curves in vivo as compared to in vitro. Cyclophosphamide was far more potent in vitro (4-HC) than in vivo (cyclophosphamide). Trimethyleneiminethiophosphoramide and N,N'-bis(2-chloroethyl)-N-nitrosourea were both more potent in vivo than in vitro. These differences may be explained by the various metabolic patterns of these drugs. Dose of alkylating agents is clearly a crucial variable particularly where multilog tumor cell kill is the goal, and in this regard, the effect of drug dose on the tumoricidal action of the alkylating agents is substantially greater than for nonalkylating agents.


Assuntos
Alquilantes/farmacologia , Antineoplásicos/farmacologia , Animais , Neoplasias da Mama/patologia , Carmustina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Ciclofosfamida/farmacologia , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Neoplasias Experimentais/tratamento farmacológico , Células Tumorais Cultivadas/efeitos dos fármacos
5.
Cancer ; 62(7): 1275-81, 1988 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-2843275

RESUMO

Normal Swiss Webster mice were treated with monocrotaline or high doses of three antitumor alkylating agents (BCNU, cyclophosphamide, or mitomycin C), all of which have been connected with hepatic veno-occlusive disease at our clinic. Prior administration of WR-2721 did not improve the survival of monocrotaline-treated animals. Glutathione (GSH) improved the survival of these animals to a small degree. Glutathione monoethyl ester (GSHet) almost completely protected animals from the toxicity of monocrotaline. Pretreatment with WR-2721 produced moderate increases in survival at the highest doses of BCNU, and at the lower BCNU doses none of the animals pretreated with WR-2721 died before they were killed on day 150. Pretreatment with GSHet gave good protection from BCNU toxicity at the highest dose of the drug, and there were no deaths in the groups of animals treated with GSHet 1 hour before BCNU. On a multiple dose schedule, GSH provided some protection from cyclophosphamide toxicity; GSHet gave a very good level of protection from cyclophosphamide. In none of these treatment groups were lesions suggestive of hepatic or pulmonary venoocclusive disease identified. In all three experimental protocols (monocrotaline, BCNU, and cyclophosphamide), there was a consistent decrease in hepatic toxicity after GSHet pretreatment; this was not observed in GSH- or WR-2721-pretreated animals. There was no evidence of protection of the FSaIIC fibrosarcoma growing in C3H mice as assayed by tumor growth delay or tumor cell survival in groups treated with two different doses of GSHet 1 hour before each drug injection compared to those treated with the BCNU or cyclophosphamide alone, or BCNU with cyclophosphamide. Pretreatment with GSHet did not alter the toxicity of these drugs to bone marrow. GSHet appears to be an effective protector of critical normal tissue and does not appear to protect tumor.


Assuntos
Carmustina/toxicidade , Ciclofosfamida/toxicidade , Glutationa/análogos & derivados , Fígado/efeitos dos fármacos , Amifostina/farmacologia , Animais , Linhagem Celular , Interações Medicamentosas , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/patologia , Glutationa/farmacologia , Fígado/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C3H , Mitomicina , Mitomicinas/toxicidade , Monocrotalina , Alcaloides de Pirrolizidina/toxicidade
6.
Cancer Res ; 48(18): 5101-5, 1988 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-3409235

RESUMO

The effect of concomitant hyperthermia on the cytotoxicities of cis-diamminedichloroplatinum(II) (CDDP), a newly synthesized drug, Pt(Rh-123)2, and its chemical components, K2PtCl4 and rhodamine 123, was examined in vitro in a squamous cell tumor line of human origin (SCC-25) and in a CDDP-resistant subline (SCC-25/CP). No difference in the cytotoxicity of hyperthermia alone was observed between these cell lines. The dose-dependent cytotoxicities of 1-h exposures to CDDP and Pt(Rh-123)2 were markedly increased at 42 degrees C and 43 degrees C in comparison to 37 degrees C, and this effect was of the same magnitude in both cell lines (enhancements of approximately 1.5 logs at 42 degrees C and 2.5 logs at 43 degrees C for CDDP and 1.5 logs at 42 degrees C and greater than 3 logs at 43 degrees C for Pt(Rh-123)2). The use of hyperthermia with CDDP, however, did not lower survivals in the SCC-25/CP cells even to the levels seen in the parent line at 37 degrees C. The cytotoxicities of K2PtCl4 and rhodamine 123 were essentially the same in the CDDP-sensitive and -resistant cells at all temperatures tested. The magnitude of the temperature effect was significantly greater for Pt(Rh-123)2 than for its chemical components. No significant effect on CDDP or Pt(Rh-123)2 accumulation was observed at 42, 43, 44 or 45 degrees C in either cell line. DNA lesions, measured by alkaline elution, were significantly enhanced for CDDP in the SCC-25 cells at 42 degrees C. These results suggest that treatment with hyperthermia and either CDDP or Pt(Rh-123)2 should result in supraadditive anti-tumor effects, although the efficacy of CDDP plus hyperthermia will be significantly less once resistance to CDDP has developed. Since resistance to CDDP does not imply cross-resistance to Pt(Rh-123)2, and since the effect of hyperthermia is somewhat greater for Pt(Rh-123)2 than for CDDP at 43 degrees C, Pt(Rh-123)2 may be more selectively toxic to tumor cells when used with local hyperthermia versus normal cells outside the treated area, especially if resistance to CDDP has already developed.


Assuntos
Carcinoma de Células Escamosas/patologia , Cisplatino/farmacologia , Hipertermia Induzida , Compostos Organometálicos/farmacologia , Compostos Organoplatínicos , Sobrevivência Celular/efeitos dos fármacos , Resistência a Medicamentos , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Rodamina 123 , Rodaminas/farmacologia
7.
J Natl Cancer Inst ; 80(8): 599-603, 1988 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-2453675

RESUMO

In an attempt to improve the antitumor efficacy of bleomycin, the effects of the oxygen-carrying emulsion Fluosol-DA and increased levels of inspired oxygen were tested in the mouse FSaIIC fibrosarcoma system. The dose-dependent cytotoxicity of bleomycin toward the FSaIIC cells in vitro was significantly decreased under hypoxic conditions, but it increased in a 95% O2-5% CO2 (carbogen) atmosphere as compared with the cytotoxicity of bleomycin in normally oxygenated cells. Investigations on the FSaIIC tumor in vivo also demonstrated that growth delays induced by bleomycin (10 mg/kg ip given on days 6, 10, 13, and 16) were significantly increased when one of the following treatments was given with each bleomycin injection: carbogen breathing for 2 hours (4.7 days), carbogen breathing for 6 hours (5.7 days), and breathing 3 atm of hyperbaric oxygen (6.3 days) versus normal air (3.3 days). When Fluosol-DA (12 mL/kg iv) was administered just before each bleomycin injection, the following growth delays were produced: 4.8 days with air breathing, 14.6 days with carbogen breathing for 2 hours, 14.9 days with carbogen breathing for 6 hours, and 19.7 days with breathing 100% O2 at 3 atm for 1 hour. Excision studies on the FSaIIC tumor also demonstrated that the cytotoxicity increased approximately fivefold when Fluosol-DA and carbogen breathing for 2 hours were combined with a single treatment with 10 mg of bleomycin/kg. In contrast, no measurable bone marrow toxicity was evident with this combined regimen. These results suggest that the use of Fluosol-DA plus carbogen breathing could add substantially to the clinical antitumor effects of bleomycin.


Assuntos
Bleomicina/uso terapêutico , Dióxido de Carbono/farmacologia , Fibrossarcoma/tratamento farmacológico , Fluorocarbonos/farmacologia , Oxigênio/farmacologia , Animais , Medula Óssea/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Combinação de Medicamentos/farmacologia , Interações Medicamentosas , Fibrossarcoma/patologia , Derivados de Hidroxietil Amido , Oxigenoterapia Hiperbárica , Camundongos , Células Tumorais Cultivadas/efeitos dos fármacos
8.
Cancer Chemother Pharmacol ; 21(4): 281-5, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-3131032

RESUMO

The tumor growth delay produced by the combination of etoposide with the alkylating agent CDDP or BCNU and Fluosol-DA with carbogen breathing in three model tumor systems was examined. The addition of Fluosol-DA to etoposide treatment increased tumor growth delay 2.8-fold, 3.3-fold and 2.2-fold in the FSaIIC fibrosarcoma, the Lewis lung carcinoma and the SW2 small-cell xenograft, respectively. In both the FSaIIC fibrosarcoma and the Lewis lung carcinoma the combination of etoposide treatment with CDDP produced an additive effect. When Fluosol-DA was added to this combination the tumor growth delay increased 1.9-fold and 1.4-fold in the FSaIIC fibrosarcoma and the Lewis lung carcinoma, respectively. Adding Fluosol-DA to a treatment regimen with etoposide and BCNU produced a 2.2-fold, 2.0-fold and 1.6-fold increase in the tumor growth delay of the FSaIIC fibrosarcoma, the Lewis lung carcinoma and the SW2 small-cell xenograft, respectively. The effect of these various treatment combinations on tumor cell survival was assessed in the FSaIIC fibrosarcoma. When the alkylating agents CDDP or BCNU were prepared in Fluosol-DA, there was an additional increase in tumor cell kill, so that with CDDP there was 2.1-fold and 4.7-fold increase in tumor cell kill and with BCNU there was 1.5-fold and 1.2-fold increase in tumor cell kill compared to the drug plus Fluosol-DA and the drug plus Fluosol-DA/carbogen breathing, respectively. The combination of etoposide and CDDP led to less than additive cell killing, and the combination of etoposide and BCNU appeared to be additive, as predicted by simple product summation, in all of the treatment conditions examined. Both etoposide + CDDP and etoposide + BCNU produced additive or less than additive toxicity to bone marrow as measured by CFU-GM.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dióxido de Carbono/farmacologia , Fluorocarbonos/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Oxigênio/farmacologia , Alquilantes/administração & dosagem , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Medula Óssea/efeitos dos fármacos , Dióxido de Carbono/administração & dosagem , Carmustina/administração & dosagem , Divisão Celular , Sobrevivência Celular , Combinação de Medicamentos/administração & dosagem , Combinação de Medicamentos/farmacologia , Etoposídeo/administração & dosagem , Fluorocarbonos/administração & dosagem , Derivados de Hidroxietil Amido , Masculino , Camundongos , Camundongos Endogâmicos , Neoplasias Experimentais/patologia , Compostos Organoplatínicos/administração & dosagem , Oxigênio/administração & dosagem
9.
Cancer Chemother Pharmacol ; 21(4): 286-91, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-3131033

RESUMO

The addition of Fluosol-DA followed by carbogen breathing increased the antitumor effect of cyclophosphamide as measured by both tumor growth delay and tumor cell survival assays. Under air breathing condition, cyclophosphamide (100 mg/kg) administered i.p. five times on alternate days produced a tumor growth delay in the FSaIIC fibrosarcoma of 8.0 +/- 0.8 days. Adding Fluosol-DA (0.3 ml) to treatment with cyclophosphamide followed by carbogen breathing increased tumor growth delay to 11.4 +/- 3.6 days, which was not statistically significantly different from that obtained with the drug plus carbogen breathing without Fluosol-DA. As the dose of Fluosol-DA was increased and administered with drug treatment followed by carbogen breathing for 6 h, increasing tumor growth delays of 15.0 +/- 1.5 days, 18.1 +/- 1.7 days and 29.4 +/- 2.2 days were observed with 0.1 ml, 0.2 ml and 0.3 ml Fluosol-DA, respectively. When 0.1 ml Fluosol-DA was administered in combination with cyclophosphamide and immediately followed by 1 h of hyperbaric oxygen (3 atm), a tumor growth delay of 13.7 +/- 1.2 days was observed. With 0.2 ml Fluosol-DA under these conditions, the tumor growth delay increased to 23.2 +/- 1.6 days, and with 0.3 ml Fluosol-DA the tumor growth delay was 35.6 +/- 3.2 days. Single doses of cyclophosphamide with and without Fluosol-DA (0.3 ml) and various conditions of oxygenation were used in an FSaIIC fibrosarcoma tumor cell survival assay. The addition of Fluosol-DA to this single-dose protocol produced a five- to tenfold increase in tumor cell kill compared to air-breathing drug-treated animals. There was no significant difference in the toxic effect of any of the treatment conditions on bone marrow.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dióxido de Carbono/farmacologia , Fluorocarbonos/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Oxigênio/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Medula Óssea/efeitos dos fármacos , Dióxido de Carbono/administração & dosagem , Divisão Celular , Sobrevivência Celular , Terapia Combinada , Ciclofosfamida/administração & dosagem , Combinação de Medicamentos/administração & dosagem , Combinação de Medicamentos/farmacologia , Fluorocarbonos/administração & dosagem , Derivados de Hidroxietil Amido , Oxigenoterapia Hiperbárica , Masculino , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Experimentais/patologia , Oxigênio/administração & dosagem
10.
Cancer Res ; 48(1): 94-100, 1988 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-3121169

RESUMO

In vitro and in vivo studies with the drug combination thioTEPA and cyclophosphamide (CPA) were carried out using the MCF-7 human breast carcinoma cell line and the EMT6 mouse mammary carcinoma cell line. In vitro, survival curves were essentially linear. The EMT6 cell line was less sensitive to thioTEPA than the MCF-7 cell line, with concentrations which reduce cell survival to 10% of 440 and 140 microM, respectively. The response of both cell lines to 4-hydroperoxycyclophosphamide was similar. Simultaneous and immediate sequential treatments with these drugs produced supraadditive cell killing of both cell lines, although the magnitude of the supraadditivity was greater in the MCF-7 cell line than in the EMT6 cell line. Both of these drugs appeared to be as effective as thiol-depleting agents as is diethyl maleate. By DNA alkaline elution, there was a pattern of increasing DNA cross-linking similar to the increasing levels of cytotoxicity of this drug combination with increasing thioTEPA concentrations. In the EMT6 tumor in vivo, the maximally tolerated combination therapy (5 mg/kg x 6 thioTEPA and 100 mg/kg x 3 CPA) produced about 25 days of tumor growth delay which was not significantly different than expected for additivity of the individual drugs. The survival of EMT6 tumor cells after treatment of the animals with various single doses of thioTEPA and CPA was assayed. Tumor cell killing by thioTEPA produced a very steep, linear survival curve through 5 logs. The tumor cell survival curve for CPA out to 500 mg/kg gave linear tumor cell kill through almost 4 logs. In all cases, the combination treatment tumor cell survivals fell well within the envelope of additivity. Both of these drugs are somewhat less toxic toward bone marrow cells by the granulocyte-macrophage colony-forming unit in vitro assay method than to tumor cells. The combination treatments were subadditive or additive in bone marrow granulocyte-macrophage colony-forming unit killing. When bone marrow is the dose-limiting tissue, there is a therapeutic advantage to the use of this drug combination.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclofosfamida/administração & dosagem , Tiotepa/administração & dosagem , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Medula Óssea/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclofosfamida/análogos & derivados , Humanos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Células Tumorais Cultivadas/efeitos dos fármacos
11.
Cancer Res ; 47(19): 5036-41, 1987 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-3113727

RESUMO

The cytotoxicity of melphalan toward exponentially growing FSaIIC fibrosarcoma cells under hypoxia, normal aeration, hyperoxygenation, and stationary phase normally oxygenated cells was examined. Through 4 logs of cell kill by melphalan, there was no difference in survival of FSaIIC cells under any of the four conditions. In the fifth and sixth logs of cell kill, melphalan was most cytotoxic toward normally aerated cells. DNA alkaline elution was performed in FSaIIC cells treated for 1 h with melphalan under the various atmospheres. Both upon immediate elution and after a 6-h delay period the greatest number of DNA cross-links were formed in the normally oxygenated cells. Tumor growth delay studies of the FSaIIC fibrosarcoma treated with melphalan were performed under four levels of oxygenation. From air breathing to 100% oxygen at 3 atm, the tumor growth delay produced by melphalan increased from about 3 days to about 9 days. With the addition of Fluosol-DA, there was an increase in tumor growth delay by melphalan from about 6.5 days with air breathing to about 13 days with 100% oxygen at 3 atm (1 h). When FSaIIC fibrosarcoma tumors were treated with melphalan, and tumor cell survival was measured by colony formation in culture, increasing doses of melphalan produced increasing levels of tumor cell kill in a relatively log linear manner. The addition of Fluosol-DA to treatment with melphalan produced approximately 1 log greater tumor cell kill than melphalan and air breathing under the various oxygenation conditions. There was approximately a 4-fold increase in toxicity to bone marrow granulocyte-macrophage colony-forming units under both extended carbogen breathing conditions (6 h) and hyperbaric oxygenation conditions (100% oxygen, 1 h, 3 atm). The response of the spleen to these various treatment regimens appeared to be immediate and shortlived. Necrotic cells were seen on day 1 posttreatment, with a substantial reduction by day 4 posttreatment. Mitotic figures were essentially absent from the liver on day 1 posttreatment, but by day 4 were significantly increased in treatment groups receiving Fluosol-DA, with the largest number seen in the melphalan/Fluosol-DA with carbogen-breathing group. In conclusion, Fluosol-DA and 1 h of carbogen breathing significantly increases the antitumor activity of melphalan without a concomitant increase in normal tissue toxicity. Although increasing the oxygenation level increased the response of the tumor, normal tissue toxicity was also increased.


Assuntos
Fluorocarbonos/farmacologia , Melfalan/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Oxigênio/farmacologia , Animais , Dióxido de Carbono/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Combinação de Medicamentos/farmacologia , Sinergismo Farmacológico , Derivados de Hidroxietil Amido , Masculino , Melfalan/toxicidade , Camundongos , Neoplasias Experimentais/patologia , Baço/efeitos dos fármacos
12.
Int J Radiat Oncol Biol Phys ; 13(8): 1217-24, 1987 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3610710

RESUMO

The complex of tetrachloroplatinate and rhodamine-123, Pt(Rh-123)2 has demonstrated cytotoxic and antitumor effects and radiosensitizing potential in vitro and in vivo. Using the FSaIIC in vivo-in vitro tumor system, tumor cell survival indicated a dose modifying factor (DMF) of 1.51 due to the addition of Pt(Rh-123)2. Pt(Rh-123)2 was added to two fractionated radiation protocols. The drug was administered by i.p. injection on three different multiple injection schedules, each reaching a cumulative dose of 75 mg/kg in those groups receiving the maximum treatment. Radiation was delivered in 3 Gy fractions in the morning and afternoon or daily for five days. Pt(Rh-123)2 was administered daily at 25 or 15 mg/kg or on alternate days at 25 mg/kg. The DMFs obtained ranged from 1.8 +/- 0.2 to 1.25 +/- 0.1. The pharmacokinetics of [195mPt]-Pt(Rh-123)2 after i.p. injection of 100 mg/kg of the drug were characterized in mice bearing the Lewis lung carcinoma using a two compartment model. The total exposure of the lung carcinoma to the drug as reflected by the area under the concentration vs. time curve was 1.5 times greater than that of the normal lung tissue. The formation of DNA cross-links and single strand breaks in SCC-25 cells exposed to Pt(Rh-123)2 (100 microM) and 6 Gy under normally oxygenated and hypoxic conditions immediately following treatment or 6 hours later were measured by DNA alkaline elution. Over the time course, the level of DNA cross-linking increased in the normally oxygenated cells by 1.5-fold and the hypoxic cells by 4-fold. The overall effects of Pt(Rh-123)2 in the presence of radiation results from both DNA cross-linking and single strand breaks.


Assuntos
Neoplasias Experimentais/radioterapia , Compostos Organoplatínicos/uso terapêutico , Radiossensibilizantes/uso terapêutico , Animais , Terapia Combinada , Cinética , Masculino , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Compostos Organoplatínicos/metabolismo , Radiossensibilizantes/metabolismo , Dosagem Radioterapêutica , Rodaminas/metabolismo , Rodaminas/uso terapêutico , Distribuição Tecidual
13.
J Clin Invest ; 79(5): 1524-8, 1987 May.
Artigo em Inglês | MEDLINE | ID: mdl-3033027

RESUMO

DNA-DNA crosslinks are the lethal cellular mechanism of bifunctional alkylating agent cytotoxicity. Novobiocin, an inhibitor of DNA topoisomerase II, impairs eukaryotic DNA repair of alkylating agent adducts and may increase the number of adducts and their resultant cytotoxicity in malignant cells. The effect of novobiocin on clonogenic survival and DNA crosslinking due to cisplatin (cDDP) and carmustine (BCNU) was studied. Novobiocin caused synergistic cytotoxicity in Chinese hamster ovary cells exposed to cDDP or BCNU. Novobiocin and cDDP increased the formation of DNA-DNA interstrand crosslinks six-fold greater than cDDP alone. The effect was schedule dependent. Novobiocin and cDDP or BCNU markedly reduced in vivo growth of a murine fibrosarcoma without increased host toxicity. As a modulating agent of cytotoxicity due to DNA-DNA crosslinking, novobiocin may enhance the clinical effectiveness of the alkylating agents in human cancer and offer insight into new therapeutic strategies.


Assuntos
Alquilantes/uso terapêutico , DNA/efeitos dos fármacos , Novobiocina/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carmustina/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/uso terapêutico , Cricetinae , Cricetulus , Sinergismo Farmacológico , Feminino , Fibrossarcoma/tratamento farmacológico , Leucemia L1210/tratamento farmacológico , Camundongos , Ovário/citologia , Ovário/efeitos dos fármacos , Inibidores da Topoisomerase II
14.
Radiat Res ; 109(1): 36-46, 1987 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3809391

RESUMO

The radiosensitizing potential in hypoxic EMT6 cells of several complexes of Co(III) and Fe(III) has been examined. The cytotoxicity of each of the agents toward oxygenated and hypoxic EMT6 cells was tested over the concentration range of 1 to 500 micron for 1-h drug exposure. There was no statistically significant difference between the cytotoxicity of these complexes toward oxygenated and hypoxic cells. Based on these findings, 100 micron was selected as the drug concentration for the initial assessment of radiosensitizing potential. The radiation survival of EMT6 cells in the presence of 100 microM drug for a series of Co(III) complexes in which the number of nitro ligands was varied showed that the hexanitro and the triamine-trinitro complexes are very effective radiosensitizers. The trans-tetrammine dinitro complex was a more effective radiosensitizer than the corresponding cis-dinitro complex. The diethylenetriamine and 1,10-phenanthroline complexes were very effective radiosensitizers, producing dose-modifying factors of 2.4. The trans-tetrammine dichloro complex was moderately effective, giving a dose-modifying factor of 1.9. On the other hand, the hexammine and triammine tricyano complexes and the trans-dinitro complex with negatively charged acetylacetonate ligands were ineffective as radiosensitizers in this system. Finally, three complexes with cyclopentadienyl ligands were examined. The ferricenium salt itself was a moderately effective radiosensitizer, giving a dose-modifying factor of 2.0. However, both the dimethylferricenium salt and the analogous cobalt complex were ineffective. The FSaIIC fibrosarcoma was used to study radiosensitizing potential in vivo. The trans-tetramminedinitro complex was administered at doses of 100, 200, or 300 mg/kg as a single ip injection 1 h prior to irradiation or as three daily ip injections. There was increasing dose modification with increasing drug dosage. With a fractionated radiation protocol in which five daily fractions of 2, 3, or 4 Gy were administered to the tumor-bearing limb with ip drug injections of 100 or 200 mg/kg given 1 h prior to irradiation, a dose-modifying effect of 1.6 was observed with 5 X 200 mg/kg of the drug.


Assuntos
Cobalto , Ferro , Radiossensibilizantes , Animais , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Hipóxia/fisiopatologia , Masculino , Camundongos , Sarcoma Experimental/radioterapia , Relação Estrutura-Atividade , Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...