Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Pharmacol Exp Ther ; 330(2): 423-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19423841

RESUMEN

P-glycoprotein (Pgp), a member of the ATP-binding cassette transporter family, is one of the major causes for multidrug resistance (MDR). We report using confocal microscopy to study the roles of Pgp in mediating the efflux of the anticancer agent mitoxantrone and the reversal of MDR by the specific Pgp inhibitor valspodar (PSC833). The net uptake and efflux of mitoxantrone and the effect of PSC833 were quantified and compared in Pgp-expressing human cancer MDA-MB-435 (MDR) cells and in parental wild-type cells. The MDR cells, transduced with the human Pgp-encoding gene MDR1 construct, were approximately 8-fold more resistant to mitoxantrone than the wild-type cells. Mitoxantrone accumulation in the MDR cells was 3-fold lower than that in the wild-type cells. The net uptake of mitoxantrone in the nuclei and cytoplasm of MDR cells was only 58 and 67% of that in the same intracellular compartment of the wild-type cells. Pretreatment with PSC833 increased the accumulation of mitoxantrone in the MDR cells to 85% of that in the wild-type cells. In living animals, the accumulation of mitoxantrone in MDA-MB-435mdr xenograft tumors was 61% of that in the wild-type tumors. Administration of PSC833 to animals before mitoxantrone treatment increased the accumulation of mitoxantrone in the MDR tumors to 94% of that in the wild-type tumors. These studies have added direct in vitro and in vivo visual information on how Pgp processes anticancer compounds and how Pgp inhibitors modulate MDR in resistant cancer cells.


Asunto(s)
Ciclosporinas/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Mitoxantrona/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular Tumoral , Ciclosporinas/metabolismo , Resistencia a Múltiples Medicamentos/fisiología , Resistencia a Antineoplásicos/fisiología , Femenino , Humanos , Ratones , Ratones Desnudos , Mitoxantrona/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
2.
J Pharmacol Exp Ther ; 324(1): 95-102, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17947497

RESUMEN

P-glycoprotein (Pgp), a membrane transporter encoded by the MDR1 gene in human cells, mediates drug efflux from cells, and it plays a major role in causing multidrug resistance (MDR). Confocal microscopy was used to study in vitro and in vivo drug accumulation, net uptake and efflux, and MDR modulation by P-glycoprotein inhibitors in MDR1-transduced human MDA-MB-435mdr (MDR) cancer cells. The MDR cells were approximately 9-fold more resistant to the anticancer drug doxorubicin than their parental wild-type MDA-MB-435wt (WT) cells. Doxorubicin accumulation in the MDR cells was only 19% of that in the WT cells. The net uptake of doxorubicin in the nuclei of the MDR cells was 2-fold lower than that in the nuclei of the WT cells. Pgp inhibitors verapamil, cyclosporine A, or PSC833 increased doxorubicin accumulation in the MDR cells up to 79%, and it reversed drug resistance in these cells. In living animals, doxorubicin accumulation in MDA-MB-435mdr xenograft tumors was 68% of that in the wild-type tumors. Administration of verapamil, cyclosporine A, or PSC833 before doxorubicin treatment of the animals increased doxorubicin accumulation in the MDR tumors up to 94%. These studies have added direct in vitro and in vivo information on the capacity of the transporter protein Pgp to efflux doxorubicin and on the reversal of MDR by Pgp inhibitors in resistant cancer cells.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antibióticos Antineoplásicos/metabolismo , Doxorrubicina/metabolismo , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclosporina/farmacología , Ciclosporinas/farmacología , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidad , Femenino , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/metabolismo , Verapamilo/farmacología
3.
Mol Cancer Ther ; 2(12): 1321-9, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14707273

RESUMEN

The major mechanism of tumor cell resistance to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) is the DNA repair protein O(6)-methylguanine DNA methyltransferase (MGMT). This repair system can be temporarily inhibited by the free base O(6)-benzylguanine (BG), which depletes cellular MGMT activity and sensitizes tumor cells and xenografts to BCNU. In clinical studies, the combination of BG and BCNU enhanced the myeloid toxicity of BCNU, thereby reducing the maximum tolerated dose. We have shown previously that retroviral expression of the P140K mutant of MGMT (MGMT-P140K) in murine and human hematopoietic cells produces significant resistance of bone marrow cells to low-dose, combination BG and BCNU treatment in vivo. In the current study, we investigated the ability of bone marrow transplantation with MGMT-P140K-transduced hematopoietic cells to protect against an intensive antitumor treatment regimen of combination BG and BCNU in non-obese diabetic/severe combined immunodeficient (NOD/SCID) mice. The donor marrow cells underwent in vivo BG and BCNU selection before transplantation, allowing infusion of a highly selected population of transduced cells. Tolerance to the intensive BG and BCNU treatment was markedly improved in secondary MGMT-P140K-transplanted mice (n = 19) compared to untransplanted mice (n = 15), as indicated by blood counts and survival rate. The dose-intensified BG and BCNU therapy produced significant growth delays of glioma xenografts in MGMT-P140K-transplanted mice, extending the tumor doubling time by >40 days. These results demonstrate that MGMT-P140K-transduced bone marrow protects against BG and BCNU combination therapy in vivo and allows dose-intensified treatment of tumor xenografts.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Médula Ósea/enzimología , Neoplasias Encefálicas/tratamiento farmacológico , Carmustina/uso terapéutico , Glioma/tratamiento farmacológico , Guanina/análogos & derivados , Guanina/uso terapéutico , O(6)-Metilguanina-ADN Metiltransferasa/genética , Animales , Antineoplásicos Alquilantes/farmacología , Carmustina/farmacología , Guanina/farmacología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Transducción Genética , Trasplante Heterólogo
4.
Hum Gene Ther ; 14(18): 1703-14, 2003 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-14670122

RESUMEN

Strategies that increase the ability of human hematopoietic stem and progenitor cells to repair alkylator-induced DNA damage may prevent the severe hematopoietic toxicity in patients with cancer undergoing high-dose alkylator therapy. In the context of genetic diseases, this approach may allow for selection of small numbers of cells that would not otherwise have a favorable growth advantage. No studies have tested this approach in vivo using human hematopoietic stem and progenitor cells. Human CD34(+) cells were transduced with a bicistronic oncoretrovirus vector that coexpresses a mutant form of O(6)-methylguanine DNA methyltransferase (MGMT(P140K)) and the enhanced green fluorescent protein (EGFP) and transplanted into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. Mice were either not treated or treated with O(6)-benzylguanine (6BG) and 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). At 8-weeks postinjection, a 2- to 8-fold increase in the percentage of human CD45(+)EGFP(+) cells in 6BG/BCNU-treated versus nontreated mice was observed in the bone marrow and was associated with increased MGMT(P140K)-repair activity. Functionally, 6BG/BCNU-treated mice demonstrated multilineage differentiation in vivo, although some skewing in the maturation of myeloid and B cells was observed in mice transplanted with granulocyte-colony stimulating factor (G-CSF)-mobilized peripheral blood compared to umbilical cord blood. Expansion of human cells in 6BG/BCNU-treated mice was observed in the majority of mice previously transplanted with transduced umbilical cord blood cells. In addition, a significant increase in the number of EGFP(+) progenitor colonies in treated versus nontreated mice were observed in highly engrafted mice indicating that selection and maintenance of human progenitor cells can be accomplished by expression of MGMT(P140K) and treatment with 6BG/BCNU.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Carmustina/efectos adversos , Diferenciación Celular , Daño del ADN , Metilasas de Modificación del ADN/genética , Reparación del ADN , Células Madre Hematopoyéticas/inmunología , Animales , Antígenos CD34 , División Celular , Femenino , Terapia Genética/métodos , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones SCID , Neoplasias/tratamiento farmacológico , Selección Genética , Transducción Genética , Trasplante Heterólogo
5.
Nucl Med Biol ; 30(4): 405-15, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12767398

RESUMEN

Novel radiolabeled O(6)-benzylguanine (O(6)-BG) derivatives, 2-amino-6-O-[(11)C]-[(methoxymethyl)benzyloxy]-9-methyl purines ([(11)C]p-O(6)-AMMP, 1a; [(11)C]m-O(6)-AMMP, 1b; [(11)C]o-O(6)-AMMP, 1c), 2-amino-6-O-benzyloxy-9-[(11)C]-[(methoxycarbonyl)methyl]purine ([(11)C]ABMMP, 2), and 2-amino-6-O-benzyloxy-9-[(11)C]-[(4'-methoxycarbonyl)benzyl]purine ([(11)C]ABMBP, 3), have been synthesized for evaluation as new potential positron emission tomography (PET) imaging agents for the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) in breast cancer. The appropriate precursors for radiolabeling were obtained in two to three steps from starting material 2-amino-6-chloropurine with moderate to excellent chemical yields. Tracers were prepared by O-[(11)C]methylation of hydroxymethyl or acid precursors using [(11)C]methyl triflate. Pure target compounds were isolated by solid-phase extraction (SPE) purification procedure in 45-65% radiochemical yields (decay corrected to end of bombardment), and a synthesis time of 20-25 min. The activity of unlabeled standard samples of 1-3 was evaluated via an in vitro AGT oligonucleotide assay. Preliminary findings from biological assay indicate the synthesized analogs have similar strong inhibitory effectiveness on AGT in comparison with the parent compound O(6)-BG. The results warrant further evaluation of these radiotracers as new potential PET imaging agents for the DNA repair protein AGT in breast cancer in vivo.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Compuestos de Bencilo/síntesis química , Guanina , Radiofármacos/síntesis química , Compuestos de Bencilo/farmacocinética , Neoplasias de la Mama/enzimología , Reparación del ADN/efectos de los fármacos , Guanina/análogos & derivados , Guanina/síntesis química , Guanina/farmacología , Radiofármacos/farmacología , Relación Estructura-Actividad , Tomografía Computarizada de Emisión
6.
J Neurosurg ; 98(3): 591-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12650433

RESUMEN

OBJECT: Temozolomide (TMZ)-induced O6-methylguanine (MG) DNA lesions, if not removed by MG-DNA methyltransferase (MGMT), mispair with thymine, trigger rounds of futile mismatch repair (MMR), and in glioma cells lead to prolonged G2-M arrest and ultimately cell death. Depletion of MGMT by O6-benzylguanine (BG) sensitizes tumor cells to TMZ, and this combination is currently used in clinical trials. The use of the TMZ+BG combination in gliomas, however, is complicated by the prolonged TMZ-induced G2-M arrest, which may delay activation of poorly defined cell death pathways and allow for MGMT repletion and reversal of toxicity. METHODS: To address these issues, the actions of TMZ were monitored in DNA MMR-proficient SF767 glioma cells depleted of MGMT by BG, and in cells in which BG was removed at various times after TMZ exposure. In MGMT-depleted cells, TMZ exposure led to DNA single-strand breaks and phosphorylation of cdc2, followed by G2-M arrest, induction of p53/p21, and DNA double-strand breaks. Although DNA single-strand breaks, phosphorylation of cdc2, and G2-M arrest could be reversed by repletion of MGMT up to 5 days after TMZ exposure, TMZ-induced cytotoxicity could only be prevented if MGMT was replenished within 24 hours of the onset of G2-M arrest, and before the creation of DNA double-strand breaks. CONCLUSIONS: These results indicate that although SF767 glioma cells undergo a prolonged G2-M arrest in response to TMZ, their ability to escape TMZ-induced cytotoxicity by MGMT repletion is limited to an approximately 24-hour period after the onset of G2-M arrest.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Glioma/patología , Guanina/análogos & derivados , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Fase G2/efectos de los fármacos , Glioma/metabolismo , Guanina/farmacología , Humanos , Mitosis/efectos de los fármacos , Temozolomida , Factores de Tiempo , Células Tumorales Cultivadas/efectos de los fármacos
7.
Bioorg Med Chem ; 13(20): 5779-86, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15993610

RESUMEN

A novel fluorine-18-labeled O6-benzylguanine (O6-BG) derivative, O6-[4-(2-[18F]fluoroethoxymethyl)benzyl]guanine (O6-[18F]FEMBG, [18F]1), has been synthesized for evaluation as a potential positron emission tomography (PET) probe for the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT) in cancer chemotherapy. The appropriate radiolabeling precursor N(2,9)-bis(p-anisyldiphenylmethyl)-O6-[4-(hydroxymethyl)benzyl]guanine (6) and reference standard O6-[4-(2-fluoroethoxymethyl)benzyl]guanine (O6-FEMBG, 1) were synthesized from 1,4-benzenedimethanol and 2-amino-6-chloropurine in four or six steps, respectively, with moderate to excellent chemical yields. The target tracer O6-[18F]FEMBG was prepared in 20-35% radiochemical yields by reaction of MTr-protected precursor 6 with [18F]fluoroethyl bromide followed by quick deprotection reaction and purification with a simplified Silica Sep-Pak method. Total synthesis time was 60-70 min from the end of bombardment. Radiochemical purity of the formulated product was >95%, with a specific radioactivity of >1.0 Ci/micromol at the end of synthesis. The activity of unlabeled O6-FEMBG was evaluated via an in vitro AGT oligonucleotide assay. Preliminary findings from biological assay indicate that the synthesized analogue has similarly strong inhibiting effect on AGT in comparison with O6-BG and O6-4-fluorobenzylguanine (O6-FBG). The results warrant further in vivo evaluation of O6-[18F]FEMBG as a new potential PET probe for AGT.


Asunto(s)
Antineoplásicos/farmacología , Guanina/análogos & derivados , Sondas Moleculares , O(6)-Metilguanina-ADN Metiltransferasa/química , Tomografía de Emisión de Positrones , Cromatografía Líquida de Alta Presión , Guanina/síntesis química , Guanina/farmacología , Espectroscopía de Resonancia Magnética , Espectrometría de Masas
8.
J Pharmacol Exp Ther ; 309(2): 506-14, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14742745

RESUMEN

O(6)-Methylguanine DNA methyltransferase (MGMT) protects tumor cells from the cytotoxic effects of DNA-alkylating agents such as 1,3-bis-(2-chloroethyl)-1-nitrosourea (BCNU). To improve the therapeutic index of BCNU, biochemical strategies to inhibit MGMT temporarily by systemic administration of small molecules, such as O(6)-benzylguanine, have been developed and are showing promise in clinical trials. In this study, an alternative molecular strategy for modulating BCNU resistance was explored using hammerhead ribozymes (Rz) designed to degrade the long-lived MGMT mRNA. We had previously identified several ribozymes capable of decreasing MGMT levels in HeLa cells. Using colony formation assays, the BCNU-induced cell kill was shown to be increased by 1 to 3 logs in the HeLa/Rz clones compared with wild-type HeLa cells at a BCNU dose of 100 microM. In the current study, 10 randomly selected clones of Rz161, 212, and a reconstructed Rz178/212 were assayed for MGMT activity, MGMT mRNA, and sensitivity to BCNU. The 30 clones exhibited almost identical results in the three assays, i.e., nearly undetectable MGMT activity, greatly diminished MGMT mRNA, and comparable sensitivity to BCNU using the 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1) viability assay. The effects of catalytically inactive ribozymes carrying a single point mutation were compared with their active counterparts in vitro and in stably transfected clones to determine whether antisense inhibition was a contributor to the inhibition of MGMT activity we observed. Collectively, these results suggest that the hammerhead ribozymes characterized in this study will be excellent candidates for future gene therapy approaches targeting MGMT.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Carmustina/farmacología , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , ARN Catalítico/metabolismo , Northern Blotting , Western Blotting , División Celular/efectos de los fármacos , Extractos Celulares/química , Células HeLa , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/genética , ARN/metabolismo , Células Madre/efectos de los fármacos , Sales de Tetrazolio , Transfección , Células Tumorales Cultivadas
9.
Bioorg Med Chem Lett ; 13(4): 641-4, 2003 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-12639548

RESUMEN

Novel radiolabeled O(6)-benzylguanine derivatives, 6-O-[(11)C]-[(methoxymethyl)benzyl]guanines ([(11)C]p-O(6)-MMBG, 1a; [(11)C]m-O(6)-MMBG, 1b; ([(11)C]o-O(6)-MMBG, 1c), have been synthesized for evaluation as new potential positron emission tomography (PET) breast cancer imaging agents for DNA repair protein, O(6)-alkylguanine-DNA alkyltransferase (AGT).


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Inhibidores Enzimáticos , Guanina/análogos & derivados , O(6)-Metilguanina-ADN Metiltransferasa/antagonistas & inhibidores , Tomografía Computarizada de Emisión/métodos , Neoplasias de la Mama/diagnóstico , Línea Celular Tumoral , Inhibidores Enzimáticos/síntesis química , Guanina/síntesis química , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Unión Proteica , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA