Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Br J Cancer ; 111(8): 1634-45, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25167227

RESUMEN

BACKGROUND: The level of plasma-derived naturally circulating anti-glycan antibodies (AGA) to P1 trisaccharide has previously been shown to significantly discriminate between ovarian cancer patients and healthy women. Here we aim to identify the Ig class that causes this discrimination, to identify on cancer cells the corresponding P1 antigen recognised by circulating anti-P1 antibodies and to shed light into the possible function of this glycosphingolipid. METHODS: An independent Australian cohort was assessed for the presence of anti-P1 IgG and IgM class antibodies using suspension array. Monoclonal and human derived anti-glycan antibodies were verified using three independent glycan-based immunoassays and flow cytometry-based inhibition assay. The P1 antigen was detected by LC-MS/MS and flow cytometry. FACS-sorted cell lines were studied on the cellular migration by colorimetric assay and real-time measurement using xCELLigence system. RESULTS: Here we show in a second independent cohort (n=155) that the discrimination of cancer patients is mediated by the IgM class of anti-P1 antibodies (P=0.0002). The presence of corresponding antigen P1 and structurally related epitopes in fresh tissue specimens and cultured cancer cells is demonstrated. We further link the antibody and antigen (P1) by showing that human naturally circulating and affinity-purified anti-P1 IgM isolated from patients ascites can bind to naturally expressed P1 on the cell surface of ovarian cancer cells. Cell-sorted IGROV1 was used to obtain two study subpopulations (P1-high, 66.1%; and P1-low, 33.3%) and observed that cells expressing high P1-levels migrate significantly faster than those with low P1-levels. CONCLUSIONS: This is the first report showing that P1 antigen, known to be expressed on erythrocytes only, is also present on ovarian cancer cells. This suggests that P1 is a novel tumour-associated carbohydrate antigen recognised by the immune system in patients and may have a role in cell migration. The clinical value of our data may be both diagnostic and prognostic; patients with low anti-P1 IgM antibodies present with a more aggressive phenotype and earlier relapse.


Asunto(s)
Antígenos de Neoplasias/inmunología , Glicoesfingolípidos/inmunología , Metástasis de la Neoplasia/inmunología , Neoplasias Ováricas/inmunología , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/aislamiento & purificación , Línea Celular Tumoral , Cromatografía de Afinidad , Femenino , Citometría de Flujo , Humanos , Neoplasias Ováricas/patología
2.
Br J Cancer ; 107(8): 1399-408, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-22976800

RESUMEN

BACKGROUND: Tumours are responsive to temozolomide (TMZ) if they are deficient in O(6)-methylguanine-DNA methyltransferase (MGMT), and mismatch repair (MMR) proficient. METHODS: The effect of TMZ on medulloblastoma (MB) cell killing was analysed with clonogenic survival assays. Expression of DNA repair genes and enzymes was investigated using microarrays, western blot, and immunohistochemistry. DNA sequencing and promoter methylation analysis were employed to investigate the cause of loss of the expression of MMR gene MLH1. RESULTS: Temozolomide exhibited potent cytotoxic activity in D425Med (MGMT deficient, MLH1 proficient; IC(50)=1.7 µM), moderate activity against D341Med (MGMT proficient, MLH1 deficient), and DAOY MB cells (MGMT proficient, MLH1 proficient). MGMT inhibitor O(6)-benzylguanine sensitised DAOY, but not D341Med cells to TMZ. Of 12 MB cell lines, D341Med, D283Med, and 1580WÜ cells exhibited MMR deficiency due to MLH1 promoter hypermethylation. DNA sequencing of these cells provided no evidence for somatic genetic alterations in MLH1. Expression analyses of MMR and MGMT in MB revealed that all patient specimens (n=74; expression array, n=61; immunostaining, n=13) are most likely MMR proficient, whereas some tumours had low MGMT expression levels (according to expression array) or were totally MGMT deficient (3 out of 13 according to immunohistochemistry). CONCLUSION: A subset of MB may respond to TMZ as some patient specimens are MGMT deficient, and tumours appear to be MMR proficient.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Cerebelosas/genética , Reparación de la Incompatibilidad de ADN/genética , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos/genética , Meduloblastoma/genética , Proteínas Nucleares/genética , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Línea Celular Tumoral , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/metabolismo , Niño , Preescolar , Metilasas de Modificación del ADN/biosíntesis , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/biosíntesis , Enzimas Reparadoras del ADN/genética , Dacarbazina/uso terapéutico , Femenino , Humanos , Masculino , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/metabolismo , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/biosíntesis , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Temozolomida , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
3.
J Leukoc Biol ; 58(5): 519-25, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7595052

RESUMEN

Using two different cytometers, an Epics Profile II and a FACScan, we determined the extent to which changes in forward and right angle scatter are a reliable measure for changes in polymorphonuclear leukocyte (PMN) shape, volume, and actin polymerization and whether distinct types of shape changes in PMNs can be recognized. PMN stimulation can substantially change the positions of PMNs in the scatter diagram of the FACScan but not of the Epics Profile II. Within the limits of the experiments, it has been possible to determine whether or not a shape change has taken place using the FACScan but not using the Epics Profile II. However, using either cytometer, it has not been possible to determine which type of shape change (e.g., spherical vs. polarized vs. nonpolar cells) has taken place. Furthermore, forward or right angle scatter changes are not a reliable measure for changes in cell volume or actin polymerization of human PMNs.


Asunto(s)
Neutrófilos/ultraestructura , Citoesqueleto de Actina/ultraestructura , Actinas/ultraestructura , Secuencia de Aminoácidos , Tamaño de la Célula , Factores Quimiotácticos/farmacología , Citometría de Flujo , Humanos , Datos de Secuencia Molecular , Activación Neutrófila/efectos de los fármacos , Oligopéptidos/farmacología , Dispersión de Radiación
4.
Ann Oncol ; 18(9): 1493-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17761705

RESUMEN

BACKGROUND: Routine adjuvant administration of trastuzumab (T) has been implemented in most centers, but its economic impact has not yet been well examined. METHODS: A Markov model was constructed based on clinical data of the Herceptin Adjuvant (HERA) and the Finland Herceptin (FinHer) trials. Costs from the perspective of a Swiss health care provider were calculated based on resource use. RESULTS: On the basis of HERA data, our model yielded an overall survival rate of 71.8% for the T group versus 62.8% for the control group [risk ratio (RR) = 0.87) after 10 years and 62.9% versus 52.7% (RR = 0.84) after 15 years. Cost-effectiveness resulted in 40505 Euros (EUR) per life years gained (LYG) after 10 years and 19673 EUR per LYG after 15 years. For the FinHer regimen, overall survival after 10 and 15 years resulted in 81.8% versus 66.1% (RR = 0.81) and 73.6% versus 57.0% (RR = 0.77). Costs of 8497 EUR per patient could be saved after 10 years and 9256 EUR after 15 years compared with the control group. CONCLUSION: In a long-term perspective, adjuvant T based on the HERA regimen can be considered cost-effective. The regimen used in the FinHer trial is even cost saving, but estimations are based on a single small trial.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Cadenas de Markov , Anticuerpos Monoclonales Humanizados , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/mortalidad , Quimioterapia Adyuvante , Ensayos Clínicos como Asunto , Análisis Costo-Beneficio , Supervivencia sin Enfermedad , Femenino , Humanos , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Trastuzumab , Estudios de Validación como Asunto
5.
Cell Motil Cytoskeleton ; 37(4): 326-37, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9258505

RESUMEN

The putative role of cellular or solvent volume in protrusive activity and locomotion has been investigated in blebbing Walker carcinosarcoma cells using hypertonic media. Blebbing, locomotion, and cell polarity are completely suppressed by 0.2 M sorbitol. The response occurs in two steps. In a first step, i.e. within 10 sec after the addition of sorbitol, blebbing and locomotion are inhibited and this is associated with an average cell volume reduction by 17% (corresponding to a reduction in solvent volume by 38%). It clearly precedes suppression of cell polarity (pre-existing protrusions, tail) occurring in a second step within 5 to 10 min after addition of sorbitol without additional reduction in the cell or solvent volume. The relative amount of F-actin does not correlate with the decrease in cell volume, suppression of blebbing, locomotion, and cell polarity. A significant decrease in the relative amount of F-actin is found only at volume reductions which are higher than those required to completely suppress blebbing, locomotion, and cell polarity. F-actin staining occurs preferentially along the cell membrane in isotonic as well as in hypertonic media. The results are best compatible with the hypothesis that hydrostatic pressure rather than actin polymerization at the front is the direct force driving the membrane forward during bleb formation. Cells with lamellipodia show a similar response to hypertonic media, suggesting that basically similar mechanisms may operate in both forms of protrusions.


Asunto(s)
Actinas/fisiología , Movimiento Celular , Soluciones Hipertónicas , Animales , Carcinoma 256 de Walker , Polaridad Celular , Tamaño de la Célula , Medios de Cultivo , Orgánulos/fisiología , Células Tumorales Cultivadas
6.
Cell Motil Cytoskeleton ; 44(1): 44-57, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10470018

RESUMEN

Capping in cells developing polarity has been reinterpreted on the basis of a quantitative analysis of Concanavalin A (Con A) redistribution and cell movement in Walker carcinosarcoma cells. Several new features emerged. Based on the developing asymmetry in the distribution of surface-bound Con A, the direction of cell movement and the prospective front-tail polarity can already be predicted when the cell is spherical. Development of polarity by an initially spherical cell is associated with formation of two parts. The concentrically contracting part (prospective uropod) characterized by surface-associated Con A decreases in size, while the other part is cleared from Con A and grows into formerly unoccupied space. Surface-bound Con A shows isotropic centripetal movement towards the initial position of the centroid of the spherical cell rather than rearward movement. Therefore, the centroid of fluorescence intensity remains either stationary or moves marginally forward with respect to the initial position of the spherical cell. The amount and direction of cell movement measured correlates closely with values predicted by a theoretical model that assumes a unidirectional transfer of volume from a stationary contracting compartment into a protruding compartment. The results suggest that isotropic (cortical) contraction of the initially spherical cells and one-sided relaxation rather than unidirectional retrograde movement of ligand-receptor complexes produces movement in cells developing polarity. Reversible accumulation of surface-bound Con A at the uropod occurring to a similar extent in untreated and colchicine-treated cells is partly due to membrane folding and partly to movement in the plane of the membrane.


Asunto(s)
Membrana Celular/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Concanavalina A/metabolismo , Animales , Carcinoma 256 de Walker/metabolismo , Carcinoma 256 de Walker/patología , Carcinoma 256 de Walker/fisiopatología , Membrana Celular/fisiología , Tamaño de la Célula/fisiología , Microscopía Confocal , Microscopía por Video , Modelos Biológicos , Células Tumorales Cultivadas
7.
Br J Cancer ; 89(8): 1559-65, 2003 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-14562032

RESUMEN

Defects in DNA mismatch repair (MMR) are associated with a predisposition to tumorigenesis and with drug resistance owing to high mutation rates and failure to engage DNA-damage-induced apoptosis. DNA minor groove binders (MGBs) are a class of anticancer agents highly effective in a variety of human cancers. Owing to their mode of action, DNA MGB-induced DNA damage may be a substrate for DNA MMR. This study was aimed at investigating the effect of loss of MMR on the sensitivity to brostallicin (PNU-166196), a novel synthetic alpha-bromoacrylic, second-generation DNA MGB currently in Phase II clinical trials and structurally related to distamycin A. Brostallicin activity was compared to a benzoyl mustard derivative of distamycin A (tallimustine). We report that the sensitivities of MLH1-deficient and -proficient HCT116 human colon carcinoma cells were comparable after treatment with brostallicin, while tallimustine resulted in a three times lower cytotoxicity in MLH1-deficient than in -proficient cells. MSH2-deficient HEC59 parental endometrial adenocarcinoma cells were as sensitive as the proficient HEC59+ch2 cells after brostallicin treatment, but were 1.8-fold resistant after tallimustine treatment as compared to the MSH2-proficient HEC59+ch2 counterpart. In addition, p53-deficient mouse fibroblasts lacking PMS2 were as sensitive to brostallicin as PMS2-proficient cells, but were 1.6-fold resistant to tallimustine. Loss of neither ATM nor DNA-PK affected sensitivity to brostallicin in p53-deficient mouse embryonic fibroblasts, indicating that brostallicin-induced cytotoxicity in a p53-deficient genetic background does not seem to require these kinases. These data show that, unlike other DNA MGBs, MMR-deficient cells retain their sensitivity to this new alpha-bromoacrylic derivative, indicating that brostallicin-induced cytotoxicity does not depend on functional DNA MMR. Since DNA MMR deficiency is common in numerous types of tumours, brostallicin potentially offers the advantage of being effective against MMR-defective tumours that are refractory to several anticancer agents.


Asunto(s)
Disparidad de Par Base , Neoplasias del Colon/patología , Reparación del ADN , Proteínas de Unión al ADN , Guanidinas/farmacología , Proteínas Proto-Oncogénicas , Pirroles/farmacología , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma/patología , Animales , Antineoplásicos/farmacología , Proteínas Portadoras , Muerte Celular , Distamicinas/farmacología , Resistencia a Antineoplásicos , Fibroblastos , Humanos , Ratones , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Compuestos de Mostaza Nitrogenada/farmacología , Proteínas Nucleares , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor
8.
Int J Cancer ; 93(4): 571-6, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11477562

RESUMEN

Sporadic breast carcinomas demonstrate microsatellite instability, reflecting the presence of DNA mismatch repair-deficient cells, in about one fourth of cases at the time of diagnosis. Loss of DNA mismatch repair has been reported to result in resistance not only to cisplatin and alkylating agents but also to the topoisomerase II poison doxorubicin, suggesting an association between DNA mismatch repair and topoisomerase II poison-induced cytotoxicity. Our study investigates the relationship between loss of MSH2 or MLH1 function and sensitivity to the topoisomerase I and II poisons, and to the taxanes, 2 classes of cytotoxic drugs commonly used in breast cancer. Two pairs of cell lines proficient and deficient in mismatch repair due to loss of either MSH2 or MLH1 function were used. Loss of either MSH2 or MLH1 function resulted in resistance to the topoisomerase II poisons doxorubicin, epirubicin and mitoxantrone, whereas only loss of MLH1 function was associated with low-level resistance to the topoisomerase I poisons camptothecin and topotecan. In contrast, there was no resistance to docetaxel and paclitaxel. Our data support the hypothesis that both MSH2 and MLH1 are involved in topoisomerase II poison-mediated cytotoxicity, whereas only MLH1 is involved in topoisomerase I poison-mediated cytotoxicity. Since our study shows that loss of DNA mismatch repair does not result in resistance to the taxanes, these drugs can be recommended for use in breast cancer deficient in mismatch repair.


Asunto(s)
Disparidad de Par Base , Reparación del ADN , Proteínas de Unión al ADN , Inhibidores Enzimáticos/farmacología , Taxoides , Inhibidores de Topoisomerasa I , Inhibidores de Topoisomerasa II , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Antibióticos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Proteínas Portadoras , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Docetaxel , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/genética , Epirrubicina/farmacología , Femenino , Humanos , Sustancias Intercalantes/farmacología , Mitoxantrona/farmacología , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/fisiología , Proteínas Nucleares , Paclitaxel/análogos & derivados , Paclitaxel/farmacología , Proteínas Proto-Oncogénicas/fisiología , Gemcitabina
9.
Br J Cancer ; 87(9): 1027-33, 2002 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-12434296

RESUMEN

A large fraction of human tumours carries mutations in the p53 gene. p53 plays a central role in controlling cell cycle checkpoint regulation, DNA repair, transcription, and apoptosis upon genotoxic stress. Lack of p53 function impairs these cellular processes, and this may be the basis of resistance to chemotherapeutic regimens. By virtue of the involvement of DNA mismatch repair in modulating cytotoxic pathways in response to DNA damaging agents, we investigated the effects of loss of Pms2 on the sensitivity to a panel of widely used anticancer agents in E1A/Ha-Ras-transformed p53-null mouse fibroblasts either proficient or deficient in Pms2. We report that lack of the Pms2 gene is associated with an increased sensitivity, ranging from 2-6-fold, to some types of anticancer agents including the topoisomerase II poisons doxorubicin, etoposide and mitoxantrone, the platinum compounds cisplatin and oxaliplatin, the taxanes docetaxel and paclitaxel, and the antimetabolite gemcitabine. In contrast, no change in sensitivity was found after treatment with 5-fluorouracil. Cell cycle analysis revealed that both, Pms2-deficient and -proficient cells, retain the ability to arrest at the G2/M upon cisplatin treatment. The data indicate that the concomitant loss of Pms2 function chemosensitises p53-deficient cells to some types of anticancer agents, that Pms2 positively modulates cell survival by mechanisms independent of p53, and that increased cytotoxicity is paralleled by increased apoptosis. Tumour-targeted functional inhibition of Pms2 may be a valuable strategy for increasing the efficacy of anticancer agents in the treatment of p53-mutant cancers.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Antineoplásicos/farmacología , Línea Celular Transformada/efectos de los fármacos , Enzimas Reparadoras del ADN , Proteínas de Unión al ADN/fisiología , Fibroblastos/efectos de los fármacos , Taxoides , Proteína p53 Supresora de Tumor/deficiencia , Animales , Apoptosis/efectos de los fármacos , Disparidad de Par Base , Hidrocarburos Aromáticos con Puentes/farmacología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Transformada/metabolismo , Reparación del ADN , Resistencia a Antineoplásicos , Fibroblastos/metabolismo , Fibroblastos/patología , Fluorouracilo/farmacología , Homocigoto , Ratones , Ratones Noqueados , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Compuestos Organoplatinos/farmacología , Azul de Tripano
10.
Ann Oncol ; 14(6): 938-45, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12796033

RESUMEN

BACKGROUND: Ataxia-telangiectasia is a pleiotropic autosomal recessive disorder caused by mutations in the ATM gene. In addition to a profound cancer predisposition, another hallmark of ataxia-telangiectasia is radiosensitivity. Recently, p53-null mouse fibroblasts have been reported to be radiosensitised by the concurrent loss of ATM. MATERIALS AND METHODS: We compared the sensitivity of atm(+/+)/p53(-/-) and atm(-/-)/p53(-/-) mouse embryonic fibroblasts to different classes of chemotherapeutic agents using the MTT assay, Trypan Blue exclusion and fluorescence-activated cell sorting for cell cycle and apoptosis analyses. RESULTS: Loss of ATM function in p53-deficient cells resulted in a 2- to 4-fold increase in sensitivity to the topoisomerase I poisons camptothecin and topotecan, to the topoisomerase II poisons doxorubicin, epirubicin and etoposide, and to the antimetabolites 5-fluorouracil and gemcitabine, but not to the platinum compounds cisplatin, carboplatin and oxaliplatin, the taxanes docetaxel and paclitaxel, or to busulfan. Loss of ATM function did not result in increased apoptosis, but resulted in increased Trypan Blue staining in response to epirubicin, suggesting that processes other than apoptosis may mediate cytotoxicity. ATM deficiency did not alter the extent of G(1)/S or G(2)/M cell cycle phase accumulation produced by epirubicin, suggesting that enhanced sensitivity was not due to failure of checkpoint activation. CONCLUSIONS: We provide further evidence that ATM is involved in regulating cellular defences against some cytotoxic agents in the absence of p53. Tumour-targeted functional inhibition of ATM may be a valuable strategy for increasing the efficacy of anticancer agents in the treatment of p53-mutant cancers.


Asunto(s)
Antineoplásicos/toxicidad , Fibroblastos/efectos de los fármacos , Eliminación de Gen , Proteínas Serina-Treonina Quinasas/fisiología , Inhibidores de Topoisomerasa II , Proteína p53 Supresora de Tumor/deficiencia , Animales , Antimetabolitos/toxicidad , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular , Línea Celular , Proteínas de Unión al ADN , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/toxicidad , Fibroblastos/metabolismo , Homocigoto , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Azul de Tripano , Proteínas Supresoras de Tumor
11.
Br J Cancer ; 86(7): 1130-5, 2002 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-11953861

RESUMEN

Loss of DNA mismatch repair is a common finding in hereditary nonpolyposis colon cancer as well as in many types of sporadic human tumours. DNA mismatch repair-deficient cells have been reported to be resistant to many chemotherapeutic agents and to radiotherapy, and to have the potential of rapidly acquiring additional mutations leading to tumour progression. Photodynamic therapy is a new treatment modality using light to activate a photosensitiser that preferentially localises in tumour cells. An oxygen dependent photochemical reaction ensues, resulting in selective tumour necrosis. The effect of loss of DNA mismatch repair activity on the sensitivity to photodynamic therapy was tested using pairs of cell lines proficient or deficient in mismatch repair due to loss of either MLH1 or MSH2 protein function. Cells were incubated with the photosensitiser 5,10,15,20-meta-tetra(hydroxyphenyl)chlorin and exposed to laser light at 652 nm with various optical doses ranging from 0-1 J cm(-2). Cell survival was assessed using the clonogenic assay. Loss of MLH1 or MSH2 function was not associated with resistance to photodynamic therapy. MCF-7 cells repeatedly treated with photodynamic therapy expressed parental levels of MLH1, MSH2, MSH6, and PMS2. DNA mismatch repair-deficient and -proficient cells showed similar subcellular distributions of meta-tetra(hydroxyphenyl)chlorin as analysed by laser scanning and fluorescence microscopy. Therefore, repeated exposure of tumour cells to photodynamic therapy does not seem to result in loss of DNA mismatch repair, and loss of mismatch repair, in turn, does not seem to contribute to resistance to photodynamic therapy. Our results suggest recommending photodynamic therapy as a strategy for circumventing resistance due to loss of DNA mismatch repair.


Asunto(s)
Disparidad de Par Base , Reparación del ADN , Proteínas de Unión al ADN , Resistencia a Antineoplásicos , Proteínas de Neoplasias/genética , Fotoquimioterapia , Proteínas Proto-Oncogénicas/genética , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma/patología , Neoplasias de la Mama/patología , Proteínas Portadoras , Neoplasias Colorrectales/patología , Humanos , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/biosíntesis , Proteínas Nucleares , Proteínas Proto-Oncogénicas/biosíntesis , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda