Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Natl Cancer Inst ; 68(5): 697-701, 1982 May.
Artigo em Inglês | MEDLINE | ID: mdl-6951083

RESUMO

A heterogeneous response to X-ray and far UV (254 nm) light irradiations was found in cultured skin fibroblast lines from 2 separate families with Gardner's syndrome. When compared to 2 normal control cultures and cultures from 2 patients with nonfamilial colon cancer, cultures from 4 clinically affected members of family 1 showed increased sensitivity to the lethal effects of both X-ray and UV light irradiations. These cells also showed a delayed pattern of X-ray potentially lethal damage repair (PLDR) and absent UV PLDR. In contrast, cultures from 3 members of family 2 (2 of whom were clinically affected) showed a normal response of survival and PLDR to both X-ray and UV light irradiations. Thus increased sensitivity of cultured skin fibroblasts to X-ray and UV light irradiations was not a consistent in vitro finding in patients with Gardner's syndrome. However, in families with Gardner's syndrome who demonstrate in vitro radiosensitivity, additional studies are needed to assess the usefulness of these techniques in detecting affected individuals prior to the development of colon carcinoma and other manifestations.


Assuntos
Síndrome de Gardner/genética , Pele/efeitos da radiação , Adolescente , Adulto , Sobrevivência Celular , Células Cultivadas , Reparo do DNA , Síndrome de Gardner/patologia , Humanos , Recém-Nascido , Cariotipagem , Pessoa de Meia-Idade , Fenótipo , Tolerância a Radiação , Risco , Pele/patologia , Raios Ultravioleta , Raios X
2.
Cancer Res ; 52(7): 1687-94, 1992 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1532343

RESUMO

The combination of fluoropyrimidines and radiation has resulted in increased control of colorectal cancer in the clinic, but the basic mechanism of the interaction is not understood clearly. Preliminary work in our laboratory showed that 2-h exposures of HT 29 human colon carcinoma cells to relatively low levels of 5-fluorodeoxyuridine resulted in extended thymidylate synthase inhibition after the drug was removed (up to 30 h after treatment with 0.5 microM 5-fluorodeoxyuridine). The low cytotoxicity associated with this treatment simplified efforts to test the effects of extended thymidylate synthase inhibition on radiosensitivity of HT 29 cells. Although thymidylate synthase was completely inhibited at the end of the 2-h exposure, an increase in the radiosensitivity of the cells was not evident until 16 h after the removal of drug. Flow cytometric analysis showed that cells accumulated in early S phase over time, and the increase in radiation sensitivity of the entire population followed the increase of the proportion of cells in early S, a relatively radiosensitive phase of the cell cycle. This treatment schedule was compared with 24-h continuous exposure, and we found that the same maximum increase in radiosensitivity was achieved by both treatment strategies. However, more cytotoxicity was associated with continuous exposure. This study provides evidence that radiosensitization by 5-fluorodeoxyuridine is in part due to alteration of cell kinetics and redistribution of cells throughout the cycle. This information may be useful in the design of less toxic combined chemo- and radiotherapy treatment strategies by limiting systemic exposure to fluoropyrimidines.


Assuntos
Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Floxuridina/farmacologia , Radiossensibilizantes/farmacologia , Ribonucleotídeos/metabolismo , Timidilato Sintase/antagonistas & inibidores , Ciclo Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Citometria de Fluxo , Humanos , Cinética , Fase S/efeitos dos fármacos , Fatores de Tempo , Raios X
3.
Cancer Res ; 58(10): 2245-52, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9605773

RESUMO

Ribonucleotide reductase (RR), the rate-limiting enzyme in the de novo synthesis of deoxynucleotide triphosphates (dNTPs), is a potential target for cancer therapy. We characterized the response of RR in a human cervical carcinoma cell line, Caski, after damage by ionizing radiation (IR). We also investigated the cell cycle regulation of both the regulatory (R1) and catalytic (R2) RR subunits in an attempt to distinguish between a direct DNA damage induction of RR by IR and a cell cycle-dependent expression of RR after IR. Confluent, growth-arrested Caski cells showed a > or = 5-fold increase in R2 mRNA and an 18-fold increase in R2 protein as cells entered S phase after serum stimulation. The R2 protein levels peaked in late S phase and returned to lower basal levels in G2-M. No changes in R1 mRNA and protein levels occurred with progression through the cell cycle after serum stimulation. In growth-arrested Caski cells treated with IR (6 Gy) without serum stimulation, a similar rise (17-fold) in R2 protein was evident at 24 h after IR and was associated with a 4-fold increase in in situ RR enzyme activity, but no increases in R1 and R2 mRNA nor R1 protein were found. E2 promoter binding factor 1 mRNA and protein levels also showed no change after IR. Growth-arrested controls (no IR and no serum stimulation) showed <4-fold elevation in R2 protein. These data suggest that RR plays a role in IR-mediated damage responses in Caski cells, which appears different than RR regulation after a proliferation (serum) stimulus. Such a response to IR in human tumor cells has not been reported previously. The use of specific R2 protein or RR enzyme inhibitors after IR may enhance IR cytotoxicity by altering this potential RR-mediated repair pathway.


Assuntos
Carcinoma/enzimologia , Proteínas de Neoplasias/efeitos da radiação , Ribonucleotídeo Redutases/efeitos da radiação , Neoplasias do Colo do Útero/enzimologia , Carcinoma/patologia , Ciclo Celular/fisiologia , Meios de Cultura , Feminino , Humanos , Proteínas de Neoplasias/metabolismo , Ribonucleotídeo Redutases/metabolismo , Células Tumorais Cultivadas/efeitos da radiação , Neoplasias do Colo do Útero/patologia
4.
Cancer Res ; 43(6): 2806-11, 1983 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-6303568

RESUMO

The in vitro response to radiation and chemotherapeutic drugs of cell lines established from 7 patients with small cell (SC) lung cancer were tested using a soft agarose clonogenic assay. Five cell lines retained the typical morphological and biochemical amine precursor uptake decarboxylation characteristics of SC, while two cell lines had undergone "transformation" to large cell (LC) morphological variants with loss of amine precursor uptake decarboxylation cell characteristics of SC. The radiation survival curves for the SC lines were characterized by D0 values ranging from 51 to 140 rads and extrapolation values (n) ranging from 1.0 to 3.3. While the D0 values of the radiation survival curves of the LC variants were similar (91 and 80 rads), the extrapolation values were 5.6 and 11.1 In vitro chemosensitivity testing of the cell lines revealed an excellent correlation between prior treatment status of the patient and in vitro sensitivity or resistance. No correlation was observed between in vitro chemosensitivity and radiation response. These data suggest that transformation of SC to LC with loss of amine precursor uptake and decarboxylation characteristics is associated with a marked increase in radiation resistance (n) in vitro. The observation of a 2- to 5-fold increase in survival of the LC compared to the SC lines following 200 rads suggests that the use of larger daily radiation fractions and/or radiation-sensitizing drugs might lead to a significantly greater clinical response in patients with LC morphology. This clinical approach may have a major impact on patient response and survival.


Assuntos
Carcinoma de Células Pequenas/terapia , Neoplasias Pulmonares/terapia , Antineoplásicos , Carmustina/uso terapêutico , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Doxorrubicina/uso terapêutico , Humanos , Técnicas In Vitro , Metotrexato/uso terapêutico , Vincristina/uso terapêutico
5.
Cancer Res ; 54(10): 2701-6, 1994 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8168100

RESUMO

We investigated the effect of iododeoxyuridine (IdUrd) exposure on cis-diamminedichloroplatinum (CDDP) cytotoxicity in the human bladder cancer cell line 647V. Following a 48-h incubation with 2-20 microM IdUrd, a 1-h exposure to 0-120 microM CDDP produced a dose-dependent increase in CDDP cytotoxicity as measured by clonogenic survival. IdUrd exposure of 2, 5, 10, and 20 microM prior to CDDP resulted in dose-modifying factors at 10% survival of 1.2, 1.6, 2.0, and 3.5, respectively. The increase in CDDP cytotoxicity appears to be associated with the level of DNA thymidine replacement in DNA by IdUrd over the range of 13-36%. Atomic absorption spectrophotometric analysis of DNA extracted from 647V cells showed that IdUrd substitution did not affect the total amount of platinum bound to the DNA or the persistence of the bound platinum over a 24-h period post-CDDP exposure versus control cells. IdUrd, unlike thymidine, was found to form two monofunctional adducts with CDDP both in vitro and in vivo. IdUrd was also found to form a mixed bifunctional adduct with deoxyguanosine (dGua) and CDDP in vitro. 1H NMR analysis of purified IdUrd-Pt and IdUrd-Pt-dGua adducts confirmed the identity of these adducts. High pressure liquid chromatography analysis of [3H]IdUrd-labeled 647V DNA digests exposed to CDDP showed the presence of two monofunctional adducts. Unlike the free solution production of adducts in vitro, the predominant adduct formed was not IdUrd-Pt. Results using 125IdUrd-labeled 647V DNA suggests that this adduct is 5-Pt-deoxyuridine. We were not able, however, to detect the presence of the bifunctional adducts IdUrd-Pt-dGua or dUrd-Pt-dGua. This was most likely due to the extremely low proportion of mixed bifunctional adducts produced in vivo. Nonetheless, these results suggest that IdUrd DNA incorporation may enhance CDDP cytotoxicity through the increase of available sites for Pt adduct formation. A Phase I clinical trial of this approach is planned.


Assuntos
Cisplatino/metabolismo , DNA de Neoplasias/metabolismo , Idoxuridina/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Cromatografia Líquida de Alta Pressão , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Humanos , Idoxuridina/farmacologia , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/tratamento farmacológico
6.
Cancer Res ; 55(13): 2800-5, 1995 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-7796406

RESUMO

Biochemical modulation of iododeoxyuridine (IdUrd) incorporation into the DNA of tumor cells is a potential clinical strategy to enhance radiosensitivity and to simultaneously differentiate the sensitivity of rapidly proliferating tumor cells and more slowly proliferating adjacent normal tissues to radiation. The interactions of hydroxyurea (HU) and IdUrd were studied in a human bladder cancer cell line, 647V. Exposure of exponentially growing 647V cells to HU concentrations of 10-100 microM for one cell population doubling (24 h) resulted in no cytotoxicity as assessed by clonogenic survival. Flow cytometric analysis showed a significant increase in an early S-phase population after a 12-h exposure but a return to a normal cell cycle distribution after a 24-h exposure to 100 microM HU. Incorporation of IdUrd into DNA was increased 2-fold by coincubation with HU (100 microM) and a clinically achievable concentration of IdUrd (2 microM) for 24 h. To elucidate the mechanism of modulation, IdUTP pools were compared in 647V cells treated with 2 microM IdUrd with or without 100 microM HU. A 2-fold increase in IdUTP pools was evident within 2 h when this drug combination was used. With the use of multivariate statistical analysis, the radiosensitivity of 647V cells was compared after a 24-h exposure to various concentrations of IdUrd (0 and 2 microM) and HU (0, 10, and 100 microM). A 24-h exposure to 100 microM HU alone or to 2 microM IdUrd alone before irradiation resulted in significant (P < 0.02) radiosensitization with sensitizer enhancement ratios of 1.15 and 1.27, respectively. A 24-h exposure to 100 microM HU + 2 microM IdUrd resulted in even more significant (P = 0.001) radiosensitization, which was found to be a greater than additive response (sensitizer enhancement ratio, 1.76 observed compared with 1.37 expected). No radiosensitization was found with a 12-h exposure to 100 microM HU alone. The mechanism of biochemical modulation of IdUrd by a noncytotoxic dose of HU is proposed as increasing the IdUTP pools by stimulating enzymes in the thymidine salvage pathway and subsequently enhancing IdUrd incorporation and radiosensitization.


Assuntos
Hidroxiureia/administração & dosagem , Idoxuridina/administração & dosagem , Radiossensibilizantes , Neoplasias da Bexiga Urinária/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Interações Medicamentosas , Humanos , Técnicas In Vitro , Timidina Monofosfato/metabolismo , Células Tumorais Cultivadas
7.
Cancer Res ; 46(4 Pt 1): 1703-6, 1986 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3004702

RESUMO

Micrococcus luteus endonuclease sensitive sites were measured by alkaline elution in normal human and ataxia-telangiectasia (AT) fibroblasts after ionizing radiation. Due to the sensitivity of this assay, repair of base damage after 3 to 6 kilorads has been measured after oxic or hypoxic radiation. With 5.5 kilorads of oxic radiation, more than 50% of the base damage was removed after 1.5 h of repair incubation in all cells, including exr+ and exr- AT cells, and approximately 75% was removed by 4 h. After 3 or 4.5 kilorads of hypoxic X-irradiation, repair was equivalent in normal and exr- AT cells. This study included three exr- AT strains which have been reported to be deficient in the removal of gamma-ray base damage at higher doses. Since these strains repaired ionizing radiation base damage normally at lower doses, which are more relevant to survival, it is concluded that the X-ray hypersensitivity of AT cells is probably not related to the repair of base damage.


Assuntos
Ataxia Telangiectasia/metabolismo , Reparo do DNA , DNA/efeitos da radiação , Células Cultivadas , Endonucleases/farmacologia , Raios gama , Humanos
8.
Cancer Res ; 43(3): 987-91, 1983 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-6825118

RESUMO

Chinese hamster V79 cells were made thermotolerant by either continuous heating at 42.5 degrees or by fractionated 43 degrees exposures with interfraction incubation at 37 degrees. For both methods of thermotolerance induction, elevations in cellular glutathione (GSH) were observed. Additionally, GSH was also shown to be elevated following a 1-hr exposure to 6% ethanol, which also induces thermotolerance. These elevations in cellular GSH preceded thermotolerance induction in regard to cell survival. To determine if a reduction in cellular GSH prior to or during heating at 42.5 degrees would influence thermotolerance, GSH levels were reduced by either pretreatment with diethylmaleate, an agent that binds GSH, or treatment during heating with buthionine sulfoximine, an agent that inhibits GSH synthesis. Both depleting protocols resulted in thermosensitization. These data suggest that GSH may be important in the early cellular response to thermal stress.


Assuntos
Glutationa/metabolismo , Temperatura Alta , Pulmão/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Cricetinae , Cricetulus , Fatores de Tempo
9.
Cancer Res ; 60(1): 92-100, 2000 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-10646859

RESUMO

Recent studies of fluoropyrimidine (FP)-mediated radiosensitization (RS) have focused on the molecular mechanisms underlying regulation of the cell cycle, particularly at the G1-S transition. Although thymidylate synthase (TS) inhibition by FP is necessary, we hypothesize that FP-RS is temporally dependent on progression of cells into S-phase under conditions of altered deoxynucleotide triphosphate pools, particularly an increased dATP:dTTP ratio, which subsequently results in enhanced DNA fragmentation and cell death. To better understand the mechanism of FP-RS, we characterized the cellular and biochemical responses to ionizing radiation (IR) alone, using different synchronization techniques in two isogenic, TS-deficient mutant cell lines, JH-1 (TS-) and JH-2 (Thy4), derived previously from a human colon cancer cell line. After G0 synchronization by leucine deprivation, these clones differ under subsequent growth conditions and dThd withdrawal: JH-2 cells have an intact G1 arrest (>72 h) and delayed cell death (>96 h), whereas JH-1 cells progress rapidly into early S-phase and undergo acute cell death (<24 h). No difference in the late S-phase and G2-M cell populations were noted between these growth-stimulated, G0-synchronized TS-deficient cell lines with dThd withdrawal. Biochemically, the intracellular ratio of dATP:dTTP increased substantially in JH-1 cells as cells progressed into early S-phase compared with JH-2 cells, which remained in G1 phase. Synchronized JH-1 cells showed significantly decreased clonogenic survival and an increase in DNA fragmentation after IR when compared with JH-2 cells. RS was demonstrated by an increase in alpha and decrease in beta, using linear quadratic analyses. An alternative synchronization technique used mimosine to induce a block in late G1, close to G1-S border. Both JH-1 and JH-2 cells, synchronized in late G1 and following growth stimulation, now progressed into S-phase identically (<24 h), with similarly increased dATP:dTTP ratios under dThd withdrawal conditions. These late G1-synchronized JH-1 and JH-2 cells also showed a comparable reduction in clonogenic survival and similar patterns of increased DNA fragmentation following IR. We suggest, based on the cellular and biochemical differences in response to IR between G0- and late G1-synchronized cells, that S-phase progression through the G1 restriction point under an altered (increased) dATP:dTTP ratio is a major determinant of FP-RS.


Assuntos
Desoxirribonucleosídeos/metabolismo , Fase G1/efeitos da radiação , Proteínas de Neoplasias/deficiência , Fase de Repouso do Ciclo Celular/efeitos da radiação , Fase S/efeitos da radiação , Timidilato Sintase/deficiência , Trifosfato de Adenosina/metabolismo , Apoptose , Sobrevivência Celular , Dano ao DNA , Desoxirribonucleosídeos/farmacologia , Citometria de Fluxo , Fase G1/efeitos dos fármacos , Fase G1/genética , Humanos , Pirimidinas , Tolerância a Radiação , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/genética , Fase S/efeitos dos fármacos , Nucleotídeos de Timina/metabolismo
10.
Cancer Res ; 46(12 Pt 1): 6509-12, 1986 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-3779659

RESUMO

Iododeoxyuridine (IdUrd) competes with thymidine for incorporation into DNA. In order to measure the incorporation of the drug in vivo, granulocytes were isolated from peripheral blood of patients at various times during and after 9- to 14-day IdUrd i.v. continuous infusions. DNA was extracted and enzymatically hydrolyzed. Both thymidine and IdUrd were separated and measured by high-performance liquid chromatography. Thymidine substitution by IdUrd was less than 1% prior to the fifth day of infusion and then increased rapidly to achieve a maximal value between 7 and 17% at the end of the infusion. In our protocol, thrombocytopenia was the most frequent dose-limiting systemic toxicity. For our group of patients, there was a clear overall relationship between the extent of substitution by IdUrd and the hematological toxicity. To our knowledge, these data provide the first direct quantitative determination of substitution by a drug into DNA in vivo without the use of radiolabeled compounds. The ability to directly monitor drug incorporation into DNA may provide the basis for monitoring and improving the selectivity of therapy.


Assuntos
DNA/metabolismo , Granulócitos/metabolismo , Idoxuridina/metabolismo , Humanos , Contagem de Leucócitos
11.
Cancer Res ; 50(16): 4962-7, 1990 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-2199032

RESUMO

5' Amino-5'-deoxythymidine (5'-AdThd) has been demonstrated previously to antagonize dTTP-mediated feedback inhibition of purified thymidine kinase from 647V, a human bladder cancer cell line. Low concentrations of 5'-AdThd (3-30 microM) have also been shown to stimulate cellular uptake of iododeoxyuridine (IdUrd) in 647V cells at clinically relevant IdUrd concentrations (2 microM). We report that the combination of 30 microM 5'-AdThd plus 2 microM IdUrd results in a significant increase of IdUrd replacement of thymidine (dThd) (18%) in the DNA of 647V cells over that obtained by exposure to 2 microM IdUrd alone (7.9%). However, increasing the 5'-AdThd concentration to 300 microM inhibited the incorporation of IdUrd into DNA (3%). IdUrd-induced radiosensitization of 647V cells, as measured by clonogenic survival, was enhanced by coincubation with 30 microM 5'-AdThd, while 300 microM 5'-AdThd reduced the IdUrd radiosensitization. Additionally, radiation-induced single strand break generation when IdUrd was incorporated into 647V DNA, as measured by rapid alkaline elution, was also enhanced by coincubation with 30 microM 5'-AdThd, while 300 microM 5'-AdThd resulted in a decrease in the number of single strand breaks produced. In T24, another bladder cancer cell line, and SV-HUC-TT1, a tumorigenic cell line derived from SV-HUC, 3-10 microM 5'-AdThd was also able to enhance IdUrd replacement of dThd in DNA. However, no stimulation of dThd replacement by 5'-AdThd occurred in SV-HUC, a prototypic "normal" bladder urothelial cell line. Since 5'-AdThd is not a substrate for mammalian thymidine kinase and has little or no cytotoxicity in vitro and in vivo, it may be a selective modulator of IdUrd radiosensitization of human bladder carcinoma and should be tested in vivo.


Assuntos
Sobrevivência Celular/efeitos da radiação , DNA de Neoplasias/biossíntese , Idoxuridina/metabolismo , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/efeitos da radiação , Neoplasias da Bexiga Urinária/metabolismo , Radioisótopos de Carbono , Sobrevivência Celular/efeitos dos fármacos , Didesoxinucleosídeos/farmacologia , Humanos , Idoxuridina/farmacologia , Cinética , Técnica de Diluição de Radioisótopos , Timidina/metabolismo , Trítio , Células Tumorais Cultivadas/citologia , Bexiga Urinária
12.
Cancer Res ; 61(13): 5193-201, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11431359

RESUMO

DNA mismatch repair (MMR) is an efficient system for the detection and repair of mismatched and unpaired bases in DNA. Deficiencies in MMR are commonly found in both hereditary and sporadic colorectal cancers, as well as in cancers of other tissues. Because fluorinated thymidine analogues (which through their actions might generate lesions recognizable by MMR) are widely used in the treatment of colorectal cancer, we investigated the role of MMR in cellular responses to 5-fluorouracil and 5-fluoro-2'-deoxyuridine (FdUrd). Human MLH1(-) and MMR-deficient HCT116 colon cancer cells were 18-fold more resistant to 7.5 microM 5-fluorouracil (continuous treatment) and 17-fold more resistant to 7.5 microM FdUrd in clonogenic survival assays compared with genetically matched, MLH1(+) and MMR-proficient HCT116 3-6 cells. Likewise, murine MLH1(-) and MMR-deficient CT-5 cells were 3-fold more resistant to a 2-h pulse of 10 microM FdUrd than their MLH1(+) and MMR-proficient ME-10 counterparts. Decreased cytotoxicity in MMR-deficient cells after treatment with various methylating agents and other base analogues has been well reported and is believed to reflect a tolerance to DNA damage. Synchronized HCT116 3-6 cells treated with a low dose of FdUrd had a 2-fold greater G(2) cell cycle arrest compared with MMR-deficient HCT116 cells, and asynchronous ME-10 cells demonstrated a 4-fold greater G(2) arrest after FdUrd treatment compared with CT-5 cells. Enhanced G(2) arrest in MMR-proficient cells in response to other agents has been reported and is believed to allow time for DNA repair. G(2) cell cycle arrest as determined by propidium iodide staining was not a result of mitotic arrest, but rather a true G(2) arrest, as indicated by elevated cyclin B1 levels and a lack of staining with mitotic protein monoclonal antibody 2. Additionally, p53 and GADD45 levels were induced in FdUrd-treated HCT116 3-6 cells. DNA double-strand break (DSB) formation was 2-fold higher in MMR-proficient HCT116 3-6 cells after FdUrd treatment, as determined by pulsed-field gel electrophoresis. The formation of DSBs was not the result of enhanced apoptosis in MMR-proficient cells. FdUrd-mediated cytotoxicity was caused by DNA-directed and not RNA-directed effects, because administration of excess thymidine (and not uridine) prevented cytotoxicity, cell cycle arrest, and DSB formation. hMLH1-dependent responses to fluoropyrimidine treatment, which may involve the action of p53 and the formation of DSBs, clearly have clinical relevance for the use of this class of drugs in the treatment of tumors with MMR deficiencies.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Reparo do DNA/fisiologia , Floxuridina/farmacologia , Fluoruracila/farmacologia , Proteínas de Neoplasias/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ciclina B/metabolismo , Ciclina B1 , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Fase G2/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Mitose/efeitos dos fármacos , Mitose/fisiologia , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/deficiência , Proteínas Nucleares , Proteínas/metabolismo , Coloração e Rotulagem/métodos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteínas GADD45
13.
Cancer Res ; 54(10): 2695-700, 1994 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8168099

RESUMO

5-iodo-2-pyrimidinone-2'-deoxyribose (IPdR) was recently reported to be converted to 5-iodo-2'-deoxyuridine (IUdR) by an aldehyde oxidase, most concentrated in liver tissue. We questioned whether IPdR could be used as a p.o. hepatotropic prodrug to increase the percentage of IUdR-DNA incorporation into liver tumors compared to normal liver with acceptable systemic toxicity. Athymic nude mice with human colon cancer (HCT-116) xenograft tumors as liver metastases and s.c. flank tumors received daily p.o. boluses (via gastric tubes) of IUdR or IPdR for 6 days. The maximum tolerated dose of IUdR was 250 mg/kg/day and was associated with a > 10% weight loss and a high percentage of IUdR-DNA incorporation (> 5%) into normal bone marrow and intestine. In contrast, animals tolerated escalating doses of IPdR to 1 gm/kg/day without weight loss and with less (1.5-4%) IUdR-DNA incorporation in normal tissues. Pharmacokinetic analysis of p.o. IPdR showed peak plasma levels of IPdR and IUdR within 15-45 min, suggesting efficient conversion of IPdR to IUdR. Aldehyde oxidase activity was found in normal liver tissue but not in other normal or tumor tissues. Additionally, we found a 2-3 times greater percentage of IUdR-DNA incorporation in tumor with IPdR than IUdR at the highest doses used. However, no differential effect in the percentage of IUdR-DNA incorporation was noted between liver metastases and s.c. tumors with either IPdR or IUdR. We conclude that p.o. IPdR offers a greater therapeutic index for tumor incorporation (and presumably radiosensitization) than a similar schedule of IUdR.


Assuntos
DNA/metabolismo , Idoxuridina/farmacocinética , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Nucleosídeos de Pirimidina/farmacocinética , Administração Oral , Animais , Neoplasias do Colo/metabolismo , Feminino , Humanos , Idoxuridina/administração & dosagem , Idoxuridina/toxicidade , Neoplasias Hepáticas/secundário , Camundongos , Transplante de Neoplasias , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/toxicidade , Distribuição Tecidual
14.
Cancer Res ; 48(10): 2933-7, 1988 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-2965970

RESUMO

The amount of iododeoxyuridine (IdUrd) incorporated into DNA determines the degree of radiosensitization. Fluorodeoxyuridine (FdUrd) has been shown to biochemically modulate IdUrd incorporation into DNA in vitro and in vivo. In this Phase I study, these drugs were coadministered to patients during 14-day continuous i.v. infusion periods in order to investigate whether the incorporation of IdUrd into DNA in vivo could be increased without increasing the dose of IdUrd. IdUrd plasma concentrations and incorporation of IdUrd into DNA of granulocytes were measured by high-performance liquid chromatography. Up to 8.8% substitution of thymidine by IdUrd was observed. Even at 3.5 mg/m2/day FdUrd for 14 days (78% of the maximum-tolerated dose as a single agent), no clinically relevant enhancement of incorporation of IdUrd into DNA of granulocytes was observed. Also, no changes in plasma levels of IdUrd were observed with escalating doses of FdUrd. Toxicity patterns (stomatitis, diarrhea, and bone marrow depression) and isobologram analysis suggested that IdUrd and FdUrd had additive, rather than synergistic, effects.


Assuntos
DNA/metabolismo , Floxuridina/farmacologia , Granulócitos/metabolismo , Idoxuridina/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Medula Óssea/efeitos dos fármacos , Feminino , Floxuridina/administração & dosagem , Humanos , Idoxuridina/administração & dosagem , Masculino , Neoplasias/tratamento farmacológico
15.
Cancer Res ; 60(20): 5773-80, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11059773

RESUMO

Mismatch repair (MMR) deficiency, which underlies hereditary nonpolyposis colorectal cancer, has recently been linked to a number of sporadic human cancers as well. Deficiency in this repair process renders cells resistant to many clinically active chemotherapy agents. As a result, it is of relevance to find an agent that selectively targets MMR-deficient cells. We have recently shown that the halogenated thymidine (dThd) analogues iododeoxyuridine (IdUrd) and bromodeoxyuridine (BrdUrd) selectively target MutL homologue-1 (MLH1)-deficient human cancer cells for radiosensitization. The levels of IdUrd and BrdUrd in cellular DNA directly correlate with the ability of these analogues to increase the sensitivity of cells and tissues to ionizing radiation, and data from our laboratory have demonstrated that MLH1-mediated MMR status impacts dThd analogue DNA levels, and consequently, analogue-induced radiosensitization. Here, we have extended these studies and show that, both in human and murine cells, MutS homologue-2 (MSH2) is also involved in processing dThd analogues in DNA. Using both E1A-transformed Msh2+/+ and Msh2-/- murine embryonic stem (ES)-derived cells (throughout this report we use Msh2+/+ and Msh2-/- to refer to murine ES-derived cell lines that are wild type or mutant, respectively, for the murine Msh2 gene) and human endometrial cancer cells differing in MSH2 status, we see the classic cytotoxic response to 6-thioguanine (6-TG) in Msh2+/+ and human HEC59/2-4 (MSH2+) MMR-proficient cells, whereas Msh2-/- cells and human HEC59 (MSH2-/-) cells are tolerant (2-log difference) to this agent. In contrast, there is very little cytotoxicity in Msh2+/+ ES-derived and HEC59/2-4 cells to IdUrd, whereas Msh2-/- and HEC59 cells are more sensitive to IdUrd. High-performance liquid chromatography analysis of IdUrd and BrdUrd levels in DNA suggests that this differential cytotoxicity may be due to lower analogue levels in MSH2+ murine and human tumor cells. The DNA levels of IdUrd and BrdUrd continue to decrease over time in Msh2+/+ cells following incubation in drug-free medium, whereas they remain high in Msh2-/- cells. This trend was also found in MSH2-deficient human endometrial cancer cells (HEC59) when compared with HEC59/2-4 (hMsh2-corrected) cells. As a result of higher analogue levels in DNA, Msh2-/- cells are selectively targeted for radiosensitization by IdUrd. Fluorescence-activated cell-sorting analysis of Msh2+/+ and Msh2-/- cells shows that selective toxicity of the halogenated nucleotide analogues is not correlated with a G2-M cell cycle arrest and apoptosis, as is found for selective killing of Msh2+/+ cells by 6-TG. Together, these data demonstrate MSH2 involvement in the processing of IdUrd and BrdUrd in DNA, as well as the differential cytotoxicity and cell cycle effects of the halogenated dThd analogues compared with 6-TG. Therefore, IdUrd and BrdUrd may be used clinically to selectively target both MLH1- and MSH2-deficient, drug-resistant cells for radiosensitization.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Bromodesoxiuridina/farmacologia , Proteínas de Ligação a DNA , DNA/metabolismo , Idoxuridina/farmacologia , Proteínas Proto-Oncogênicas/fisiologia , Radiossensibilizantes/farmacologia , Tioguanina/farmacologia , Proteínas E1A de Adenovirus/genética , Animais , Pareamento Incorreto de Bases , Bromodesoxiuridina/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Transformada , DNA/genética , Reparo do DNA , Nucleotídeos de Desoxicitosina/metabolismo , Relação Dose-Resposta a Droga , Humanos , Idoxuridina/metabolismo , Cinética , Camundongos , Camundongos Knockout , Proteína 2 Homóloga a MutS , Proteínas Proto-Oncogênicas/genética , Nucleotídeos de Timina/metabolismo
16.
Cancer Res ; 59(8): 1840-5, 1999 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10213489

RESUMO

Deficiency in DNA mismatch repair (MMR) is found in some hereditary (hereditary nonpolyposis colorectal cancer) and sporadic colon cancers as well as other common solid cancers. MMR deficiency has recently been shown to impart cellular resistance to multiple chemical agents, many of which are commonly used in cancer chemotherapy. It is therefore of interest to find an approach that selectively targets cells that have lost the ability to perform MMR. In this study, we examine the response of MMR-proficient (hMLH1+) and MMR-deficient (hMLH1-) colon carcinoma cell lines to the halogenated thymidine (dThd) analogues iododeoxyuridine (IdUrd) and bromodeoxyuridine (BrdUrd) before and after irradiation. These dThd analogues are used clinically as experimental sensitizing agents in radioresistant human cancers, and there is a direct correlation between the levels of dThd analogue DNA incorporation and tumor radiosensitization. In contrast to the well-characterized, marked increase in cytotoxicity (> 1 log cell kill) found with 6-thioguanine exposures in HCT116/3-6 (hMLH1+) cells compared to HCT116 (hMLH1-) cells, we found only modest cytotoxicity (10-20% cell kill) in both cell lines when treated with IdUrd or BrdUrd for 1 population doubling. Upon further analysis, the levels of halogenated dThd analogues in DNA were significantly lower (two to three times lower) in HCT116/3-6 cells than in HCT116 cells, and similar results were found in Mlh1+/+ spontaneously immortalized murine embryonic fibroblasts and fibroblasts from Mlh1 knockout mice. As a result of the higher levels of the dThd analogue in DNA, there was an increase in radiation sensitivity in HCT116 cells but not in HCT116/3-6 cells after pretreatment with IdUrd or BrdUrd when compared to treatment with radiation alone. Additionally, we found no differences in the cellular metabolic pathways for dThd analogue DNA incorporation because the enzyme activities of dThd kinase and thymidylate synthase, as well as the levels of triphosphate pools, were similar in HCT116 and HCT116/3-6 cells. These data suggest that the hMLH1 protein may participate in the recognition and subsequent removal of halogenated dThd analogues from DNA. Consequently, whereas MMR-deficient cells and tumor xenografts have shown intrinsic resistance to a large number of chemotherapeutic agents, the 5-halogenated dThd analogues appear to selectively target such cells for potential enhanced radiation sensitivity.


Assuntos
Neoplasias do Colo/genética , Proteínas de Neoplasias/metabolismo , Timidina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Bromouracila/análogos & derivados , Proteínas de Transporte , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Reparo do DNA , DNA de Neoplasias/biossíntese , DNA de Neoplasias/efeitos dos fármacos , Desoxirribonucleotídeos/metabolismo , Humanos , Proteína 1 Homóloga a MutL , Proteínas Nucleares , Radiação Ionizante , Tioguanina/metabolismo , Tioguanina/farmacologia , Timidina/análogos & derivados , Timidina Quinase/metabolismo , Timidilato Sintase/metabolismo , Células Tumorais Cultivadas , Uridina/análogos & derivados , Uridina/metabolismo , Uridina/farmacologia
17.
Cancer Res ; 44(4): 1702-5, 1984 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-6704976

RESUMO

Previously, 5-bromodeoxyuridine (BrdUrd) has been shown to be an effective radiosensitizing agent in rapidly dividing cells. As part of a Phase I/II study to evaluate BrdUrd as a radiosensitizer in gliomas, the pharmacology was studied in eight patients. BrdUrd was infused using an i.v. route as a 12-hr constant infusion each day for as long as 14 days. BrdUrd steady-state arterial levels are described for three different infusional rates: 1.6 mumol/sq m/min (350 mg/sq m/12 hr) produced a steady-state arterial level of 0.7 microM; 3.2 mumol/sq m/min (700 mg/sq m/12 hr) resulted in 2.1 microM; 5.9 mumol/sq m/min (650 mg/sq m/6 hr) showed a level of 3.9 microM. Because of myelosuppression, the highest tolerable dose for this intermittent long-term infusional therapy with BrdUrd appears to be 700 mg/sq m/12 hr. Contrary to the nonlinear pharmacokinetics of thymidine, 5-fluorouracil, and 5-fluorodeoxyuridine described previously, BrdUrd shows linear behavior in the range studied. BrdUrd still has promise as a radiosensitizer for gliomas in humans, but an alternative means of safe delivery into the carotid artery is needed. Because of an estimated 11- to 16-fold-higher local concentration, use of the intraarterial route could deliver optimum levels of BrdUrd to the tumor with minimal systemic toxicity.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Bromodesoxiuridina/administração & dosagem , Glioma/tratamento farmacológico , Bromodesoxiuridina/uso terapêutico , Esquema de Medicação , Humanos , Infusões Parenterais , Cinética
18.
Cancer Res ; 45(5): 2110-5, 1985 May.
Artigo em Inglês | MEDLINE | ID: mdl-3986765

RESUMO

The optimum integration of chemotherapy and irradiation is of potential clinical significance in the treatment of ovarian cancer. A series of human ovarian cancer cell lines have been developed in which dose-response relationships to standard anticancer drugs have been determined, and the patterns of cross-resistance between these drugs and irradiation have been established. By stepwise incubation with drugs, sublines of A2780, a drug-sensitive cell line, have been made 100-fold, 10-fold, and 10-fold more resistant to Adriamycin (2780AD), melphalan (2780ME), and cisplatin (2780CP). Two additional cell lines, NIH:OVCAR-3nu(Ag+) and NIH:OVCAR-4(Ag+), were established from drug-refractory patients. 2780ME, 2780CP, OVCAR-3nu(Ag+), and OVCAR-4(Ag+) are all cross-resistant to irradiation, with DOS of 146, 187, 143, and 203, respectively. However, 2780AD remains sensitive to radiation, with a DO of 111, which is similar to that of A2780 (101). Glutathione (GSH) levels are elevated in 2780ME, 2780CP, OVCAR-3nu(Ag+), and OVCAR-4(Ag+) to 4.58, 6.13, 12.10, and 15.14 nmol/10(6) cells as compared to A2780, with 1.89 nmol/10(6) cells. However, the GSH level in 2780AD is only minimally higher than that in A2780 (2.94 nmol/10(6) cells). Buthionine sulfoximine, a specific inhibitor of GSH synthesis, significantly increases the radiation sensitivity of 2780ME (changing the DO from 143 to 95) and 2780CP to a lesser extent, suggesting that intracellular GSH levels may play an important role in the radiation response of certain neoplastic cells. These results suggest that the sequential use of irradiation following chemotherapy with melphalan and cisplatin may be less effective than a combined modality approach, which integrates radiation and chemotherapy prior to the development of drug resistance and cross-resistance to irradiation.


Assuntos
Antineoplásicos/uso terapêutico , Metionina Sulfoximina/análogos & derivados , Neoplasias Ovarianas/radioterapia , Butionina Sulfoximina , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Resistência a Medicamentos , Feminino , Glutationa/análise , Humanos , Metionina Sulfoximina/farmacologia , Neoplasias Ovarianas/patologia
19.
Cancer Res ; 58(4): 767-78, 1998 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9485033

RESUMO

A role for the Mut L homologue-1 (MLH1) protein, a necessary component of DNA mismatch repair (MMR), in G2-M cell cycle checkpoint arrest after 6-thioguanine (6-TG) exposure was suggested previously. A potential role for MLH1 in G1 arrest and/or G1-S transition after damage was, however, not discounted. We report that MLH1-deficient human colon carcinoma (HCT116) cells showed decreased survival and a concomitant deficiency in G2-M cell cycle checkpoint arrest after ionizing radiation (IR) compared with genetically matched, MMR-corrected human colon carcinoma (HCT116 3-6) cells. Similar responses were noted between murine MLH1 knockout compared to wild-type primary embryonic fibroblasts. MMR-deficient HCT116 cells or embryonic fibroblasts from MLH1 knockout mice also demonstrated classic DNA damage tolerance responses after 6-TG exposure. Interestingly, an enhanced p53 protein induction response was observed in HCT116 3-6 (MLH1+) compared with HCT116 (MLH1-) cells after IR or 6-TG. Retroviral vector-mediated expression of the E6 protein did not, however, affect the enhanced G2-M cell cycle arrest observed in HCT116 3-6 compared with MLH1-deficient HCT116 cells. A role for MLH1 in G2-M cell cycle checkpoint control, without alteration in G1, after IR was also suggested by similar S-phase progression between irradiated MLH1-deficient and MLH1-proficient human or murine cells. Introduction of a nocodazole-induced G2-M block, which corrected the MLH1-mediated G2-M arrest deficiency in HCT116 cells, clearly demonstrated that HCT116 and HCT116 3-6 cells did not differ in G1 arrest or G1-S cell cycle transition after IR. Thus, our data indicate that MLH1 does not play a major role in G1 cell cycle transition or arrest. We also show that human MLH1 and MSH2 steady-state protein levels did not vary with damage or cell cycle changes caused by IR or 6-TG. MLH1-mediated G2-M cell cycle delay (caused by either MMR proofreading of DNA lesions or by a direct function of the MLH1 protein in cell cycle arrest) may be important for DNA damage detection and repair prior to chromosome segregation to eliminate carcinogenic lesions (possibly brought on by misrepair) in daughter cells.


Assuntos
Ciclo Celular/efeitos da radiação , Neoplasias do Colo/genética , Proteínas de Neoplasias/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte , Sobrevivência Celular , Células Cultivadas , Reparo do DNA , Fibroblastos , Humanos , Camundongos , Camundongos Knockout , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/genética , Proteínas Nucleares , Tioguanina/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/fisiologia
20.
Cancer Res ; 61(22): 8290-7, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11719462

RESUMO

Our previous data demonstrated that cells deficient in MutL homologue-1 (MLH1) expression had a reduced and shorter G(2) arrest after high-dose-rate ionizing radiation (IR), suggesting that the mismatch re pair (MMR) system mediates this cell cycle checkpoint. We confirmed this observation using two additional isogenetically matched human MLH1 (hMLH1)-deficient and -proficient human tumor cell systems: human ovarian cancer cells, A2780/CP70, with or without ectopically expressed hMLH1, and human colorectal carcinoma cells, RKO, with or without azacytidine treatment to reexpress hMLH1. We also examined matched MutS homologue-2 (hMSH2)-deficient and -proficient human endometrial carcinoma HEC59 cell lines to determine whether hMSH2, and MMR in general, is involved in IR-related G(2) arrest responses. As in MLH1-deficient cells, cells lacking hMSH2 demonstrated a similarly altered G(2) arrest in response to IR (6 Gy). These differences in IR-induced G(2) arrest between MMR-proficient and -deficient cells were found regardless of whether synchronized cells were irradiated in G(0)/G(1) or S phase, indicating that MMR indeed dramatically affects the G(2)-M checkpoint arrest. However, unlike the MMR-dependent damage tolerance response to 6-thioguanine exposures, no significant difference in the clonogenic survival of MMR-deficient cells compared with MMR-proficient cells was noted after high-dose-rate IR. In an attempt to define the signal transduction mechanisms responsible for MMR-mediated G(2) arrest, we examined the levels of tyrosine 15 phosphorylation of cdc2 (phospho-Tyr15-cdc2), a key regulator of the G(2)-M transition. Increased phospho-Tyr15-cdc2 levels were observed in both MMR-proficient and -deficient cell lines after IR. However, the levels of the phospho-Tyr15-cdc2 rapidly decreased in MMR (hMLH1 or hMSH2)-deficient cell lines at times coincident with progress from the IR-induced G(2) arrest through M phase. Thus, differences in the levels of phospho-Tyr15-cdc2 after high-dose-rate IR correspond temporally with the observed differences in the IR-induced G(2) arrest, suggesting that MMR proteins may exert their effect on IR-induced G(2) arrest by signaling the cdc2 pathway. Although MMR status does not significantly affect the survival of cells after high-dose-rate IR, it seems to regulate the G(2)-M checkpoint and might affect overall mutation rates.


Assuntos
Pareamento Incorreto de Bases , Proteína Quinase CDC2/fisiologia , Reparo do DNA/fisiologia , Fase G2/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Proteína Quinase CDC2/metabolismo , Proteínas de Transporte , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Humanos , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/deficiência , Proteínas Nucleares , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação , Fase S/efeitos dos fármacos , Fase S/fisiologia , Fase S/efeitos da radiação , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Tioguanina/farmacologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA