Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int. braz. j. urol ; 46(3): 353-362, May-June 2020. tab, graf
Article in English | LILACS | ID: biblio-1090612

ABSTRACT

ABSTRACT Purpose: Testicular germ cells tumor (TGCT) are associated with a high cure rate and are treated with platinum-based chemotherapy. However, a group of testicular cancer patients may have a very unfavorable evolution and insensitivity to the main therapeutic agent chemotherapy (CT) cisplatin. The aim of this study was to evaluate the risk of recurrence and overall survival related to the expression of nuclear factor kappa-B (NF-κB), transglutaminase 2 (TG2) and excision repair cross-complementation group 1 (ERCC1) in patients with TGCT treated with platinum combinations. Patients and Methods: A retrospective study was performed with TGCT patients treated with platinum-based chemotherapy. Immunohistochemical analysis was performed and the expression was correlated with clinical and laboratory data. Results: Fifty patients were included, the mean age was 28.4 years (18 to 45), and 76% were non-seminoma. All patients were treated with standard cisplatin, etoposide and bleomycin or cisplatin, and etoposide. Patient's analyzed immunodetection for NF-κB, TG2, and ERCC1 were positive in 76%, 54% and 42%, respectively. Multivariate analysis identified that positive expressions to ERCC1 and NF-κB are independent risk factors for higher recurrence TGCT after chemotherapy (RR 2.96 and 3.16, respectively). Patients with positive expression of ERCC1 presented a poor overall survival rate for 10-year follow (p=0.001). Conclusions: The expression of ERCC1 and NF-κB give a worse prognosis for relapse, and only ERCC1 had an influence on the overall survival of TGCT patients treated with platinum-based chemotherapy. These may represent markers that predict poor clinical outcome and response to cisplatin.


Subject(s)
Humans , Male , Adult , Testicular Neoplasms , Transglutaminases/metabolism , NF-kappa B/metabolism , GTP-Binding Proteins/metabolism , Lung Neoplasms , Prognosis , Antineoplastic Combined Chemotherapy Protocols , Retrospective Studies , Cisplatin , Drug Resistance, Neoplasm/physiology , DNA-Binding Proteins , DNA Repair , Endonucleases
2.
Arch. endocrinol. metab. (Online) ; 63(6): 568-575, Nov.-Dec. 2019. graf
Article in English | LILACS | ID: biblio-1055025

ABSTRACT

ABSTRACT Tumor development is a multistep process whereby local mechanisms enable somatic mutations during preneoplastic stages. Once a tumor develops, it becomes a complex organ composed of multiple cell types. Interactions between malignant and non-transformed cells and tissues create a tumor microenvironment (TME) comprising epithelial cancer cells, cancer stem cells, non-tumorous cells, stromal cells, immune-inflammatory cells, blood and lymphatic vascular network, and extracellular matrix. We review reports and present a hypothesis that postulates the involvement of growth hormone (GH) in field cancerization. We discuss GH contribution to TME, promoting epithelial-to-mesenchymal transition, accumulation of unrepaired DNA damage, tumor vascularity, and resistance to therapy. Arch Endocrinol Metab. 2019;63(6):568-75


Subject(s)
Humans , DNA Damage/physiology , Drug Resistance, Neoplasm/physiology , Human Growth Hormone/physiology , Epithelial-Mesenchymal Transition/physiology , Tumor Microenvironment/physiology , Neovascularization, Pathologic/physiopathology
3.
Biol. Res ; 51: 58, 2018. graf
Article in English | LILACS | ID: biblio-1011402

ABSTRACT

BACKGROUND: Studies have demonstrated that transforming growth factor beta-1 (TGF-ß1) exhibits oncogenic activity in different types of cancer, including ovarian cancer (OC). However, its regulatory mechanism in OC and whether TGF-ß1 is involved in chemosensitivity regulation remains unclear. Thus, the aim of this study was to investigate the role of TGF-ß1 in OC. METHODS: The OC cell line SKOV3 was employed, and TGF-ß1 overexpression or knockdown vectors were constructed. The cell proliferation of SKOV3 was evaluated with the cell counting kit (CCK8) kit after treatment with different concentrations of cis-platinum. Western blot and protein immunoprecipitation were employed to detect changes in BRCA1 and Smad3 expression and their interactions. Tumor growth in nude mice was evaluated. RESULTS: The results showed that TGF-ß1 knockdown increased chemosensitivity by promoting BRCA1 expression and Smad3 phosphorylation. In vivo studies showed that TGF-ß1 knockdown significantly inhibited the growth of tumors, also by upregulating BRCA1 expression and Smad3 phosphorylation. CONCLUSION: Taken together, our results suggest that TGF-ß1 knockdown inhibits tumor growth and increases chemosensitivity by promotion of BRCA1/Smad3 signaling.


Subject(s)
Humans , Animals , Male , Female , Ovarian Neoplasms/metabolism , Down-Regulation/physiology , Genes, BRCA1/physiology , Smad3 Protein/physiology , Transforming Growth Factor beta1/physiology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Immunohistochemistry , Cells, Cultured , Blotting, Western , Drug Resistance, Neoplasm/physiology , Tumor Suppressor Proteins/physiology , Cell Line, Tumor , Cell Proliferation , Smad3 Protein/analysis , Transforming Growth Factor beta1/analysis , Gene Knockdown Techniques , Real-Time Polymerase Chain Reaction , Mice, Inbred BALB C
4.
Braz. j. med. biol. res ; 50(5): e6359, 2017. graf
Article in English | LILACS | ID: biblio-839294

ABSTRACT

MicroRNAs (miRNAs) play an important role in drug resistance and modulate the efficiency of chemotherapy. A recent study indicated that miR-340 functions as a tumor suppressor in various types of cancer. However, the role of miR-340 in chemotherapy has not been reported yet. In this study, we found that miR-340 enhanced cisplatin (CDDP)-induced cell death. Induction of miR-340-5p expression decreased the IC50 of CDDP and increased the apoptosis of CDDP-resistant MG-63 and Saos-2 cells. Moreover, miR-340-5p decreased the accumulation of MRP1 and MDR1. We further explored the mechanism underlying the promoting effects of miR-340-5p on CDDP-induced cell death. We identified a potential target of miR-340 in the 3′ untranslated region of lysophosphatidic acid acyltransferase (LPAATβ) using the online program Targetscan (http://www.microrna.org). Luciferase reporter assays showed that miR-340 binds to the 3′UTR of LPAATβ. Enforced expression of miR-340-5p decreased the accumulation of LPAATβ in both MG-63 and Saos-2 cells. Silencing LPAATβ decreased the IC50 of CDDP and increased the apoptosis of CDDP-resistant MG-63 and Saos-2 cells, which is consistent with the effect of miR-340-5p on CDDP-induced cell death. Moreover, induced expression of LPAATβ compromised the effects of miR-340-5p on CDDP-induced cell death and accumulation of MRP1 and MDR1. Taken together, our data indicated that miR-340-5p enhanced the sensitivity to CDDP by targeting LPAATβ.


Subject(s)
Humans , Acyltransferases/physiology , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm/physiology , MicroRNAs/physiology , Osteosarcoma/drug therapy , Acyltransferases/analysis , Acyltransferases/drug effects , Apoptosis/drug effects , Blotting, Western , Bone Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Luciferases , MicroRNAs/analysis , MicroRNAs/drug effects , Osteosarcoma/physiopathology , Real-Time Polymerase Chain Reaction
5.
J. bras. nefrol ; 36(4): 476-481, Oct-Dec/2014. tab, graf
Article in Portuguese | LILACS | ID: lil-731152

ABSTRACT

Introdução: Dados nacionais sobre diálise crônica têm tido impacto no planejamento do tratamento. Objetivo: Apresentar dados do inquérito da Sociedade Brasileira de Nefrologia sobre os pacientes com doença renal crônica em tratamento dialítico em julho de 2013 e comparar com dados de 2011- 12. Métodos: Levantamento de dados de unidades de diálise do país. A coleta de dados foi feita utilizando questionário preenchido on-line pelas unidades de diálise. Resultados: Trezentos e trinta e quatro (51%) unidades responderam ao inquérito. Em julho de 2013, o número total estimado de pacientes em diálise foi de 100.397. As estimativas nacionais das taxas de prevalência e de incidência de tratamento dialítico foram de 499 (variação: 284 na região Norte e 622 na Sul) e 170 pacientes por milhão da população, respectivamente. O número estimado de pacientes que iniciaram tratamento em 2013 foi 34.161. A taxa anual de mortalidade bruta foi de 17,9%. Dos pacientes prevalentes, 31,4% tinham idade ≥ 65 anos, 90,8% estavam em hemodiálise e 9,2% em diálise peritoneal, 31.351 (31,2%) estavam em fila de espera para transplante, 30% tinham diabetes, 17% tinham PTH > 600 pg/ml e 23% hemoglobina < 10 g/dl. Cateter venoso era usado como acesso em 15,4% dos pacientes em hemodiálise. Conclusão: O número absoluto de pacientes em diálise tem aumentado 3% ao ano nos últimos 3 anos. As taxas de prevalência e incidência de pacientes em diálise ficaram estáveis, e a taxa de mortalidade tendeu a diminuir em relação a 2012. Houve tendência a melhor controle da anemia e dos níveis de PTH. .


Introduction: National chronic dialysis data have had impact in the treatment planning. Objective: To report data of the annual survey of the Brazilian Society of Nephrology about chronic kidney disease patients on dialysis in July 2013 and compare with 2011-12. Methods: A survey based on data of dialysis units from the whole country. The data collection was performed by using a questionnaire filled out on-line by the dialysis units. Results: Three hundred thirty four (51%) of the dialysis units in the country answered the questionnaire. In July 2013, the total estimated number of patients on dialysis was 100,397. The estimated prevalence and incidence rates of chronic maintenance dialysis were 449 (range: 284 in the North region and 622 in the South) and 170 patients per million population, respectively. The estimated number of new patients starting dialysis in 2013 was 34,161. The annual gross mortality rate was 17.9%. For prevalent patients, 31.4% were aged 65 years or older, 90.8% were on hemodialysis and 9.2% on peritoneal dialysis, 31,351 (31.2%) were on a waiting list of renal transplant, 30% were diabetics, 17% had PTH levels > 600 pg/ml and 23% hemoglobin < 10 g/ dl. A venous catheter was the vascular access for 15.4% of the hemodialysis patients. Conclusion: The absolute number of patients on dialysis has increased 3% per year. The prevalence and incidence rates of patients on dialysis leveled off, while the mortality rate tended to decrease compared with 2012. There was a trend towards a better control of the anemia and PTH levels. .


Subject(s)
Animals , Mice , Cellular Senescence/physiology , /physiology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/genetics , /physiology , Ubiquitin-Protein Ligases , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/genetics , Apoptosis/physiology , Biomarkers , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , /genetics , Cyclophosphamide/therapeutic use , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Lymphoma, B-Cell/drug therapy , Mice, Knockout , Mice, Mutant Strains , Mutation , Prognosis , Proto-Oncogene Proteins c-cbl , /metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Cells, Cultured , /genetics , /physiology , /genetics
6.
Rev. méd. Chile ; 142(5): 599-605, mayo 2014. ilus, tab
Article in Spanish | LILACS | ID: lil-720669

ABSTRACT

Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm related to the presence of the BCR-ABL1 fusion gene, linked to t (9;22) (q34;q11). It is originated from an abnormal hematopoietic stem cell, which is characterized as its normal counterparts by long-term self-renewal and multi-lineage differentiation. Both leukemic and quiescent normal hematopoietic stem cells preferentially reside in the osteoblastic niche. Mesenchymal stromal cells (MSC) are located near them, playing a critical role in their regulation. Currently, with tyrosine kinase inhibitor (TKI) therapy, long term clinical responses are achieved in most CML cases. However, late treatment failures may be observed related to the persistence of leukemic stem cells. The interactions between the leukemic stem cell and the microenvironment may be responsible in part for these events. We review the interactions between the leukemic stem cell and BM stroma and its potential clinical and therapeutic implications.


Subject(s)
Humans , Bone Marrow/physiopathology , Drug Resistance, Neoplasm/physiology , Hematopoietic Stem Cells/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Mesenchymal Stem Cells/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
7.
Biol. Res ; 47: 1-9, 2014. ilus
Article in English | LILACS | ID: biblio-950762

ABSTRACT

As regards their morphology and biology, tumours consist of heterogeneous cell populations. The cancer stem cell (CSC) hypothesis assumes that a tumour is hierarchically organized and not all of the cells are equally capable of generating descendants, similarly to normal tissue. The only cells being able to self-renew and produce a heterogeneous tumour cell population are cancer stem cells. CSCs probably derive from normal stem cells, although progenitor cells may be taken into consideration as the source of cancer stem cells. CSCs reside in the niche defined as the microenvironment formed by stromal cells, vasculature and extracellular matrix. The CSC assays include FACS sorting, xenotransplantation to immunodeficient mice (SCID), incubation with Hoechst 33342 dye, cell culture in non-adherent conditions, cell culture with bromodeoxyuridine. CSCs have certain properties that make them resistant to anticancer therapy, which suggests they may be the target for potential therapeutic strategies.


Subject(s)
Animals , Mice , Neoplastic Stem Cells/pathology , Cell Differentiation/physiology , Drug Resistance, Neoplasm/physiology , Tumor Microenvironment/physiology , Carcinogenesis/pathology , Cell Self Renewal/physiology , Prognosis , Biomarkers, Tumor/therapeutic use , Mice, SCID , Stromal Cells/pathology , Extracellular Matrix/pathology , Microvessels/physiopathology , Clonal Evolution/physiology , Flow Cytometry , Fluorescent Dyes
8.
Braz. j. med. biol. res ; 46(8): 650-658, ago. 2013. graf
Article in English | LILACS | ID: lil-684524

ABSTRACT

Cisplatin resistance remains one of the major obstacles when treating epithelial ovarian cancer. Because oxaliplatin and nedaplatin are effective against cisplatin-resistant ovarian cancer in clinical trials and signal transducer and activator of transcription 3 (STAT3) is associated with cisplatin resistance, we investigated whether overcoming cisplatin resistance by oxaliplatin and nedaplatin was associated with the STAT3 pathway in ovarian cancer. Alamar blue, clonogenic, and wound healing assays, and Western blot analysis were used to compare the effects of platinum drugs in SKOV-3 cells. At an equitoxic dose, oxaliplatin and nedaplatin exhibited similar inhibitory effects on colony-forming ability and greater inhibition on cell motility than cisplatin in ovarian cancer. Early in the time course of drug administration, cisplatin increased the expression of pSTAT3 (Tyr705), STAT3α, VEGF, survivin, and Bcl-XL, while oxaliplatin and nedaplatin exhibited the opposite effects, and upregulated pSTAT3 (Ser727) and STAT3β. The STAT3 pathway responded early to platinum drugs associated with cisplatin resistance in epithelial ovarian cancer and provided a rationale for new therapeutic strategies to reverse cisplatin resistance.


Subject(s)
Animals , Humans , Rats , Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/physiology , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , /metabolism , Signal Transduction/drug effects , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Migration Assays/methods , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Gene Expression/drug effects , Inhibitor of Apoptosis Proteins/genetics , Organoplatinum Compounds/administration & dosage , Oxazines/pharmacology , Vascular Endothelial Growth Factor A/genetics , Xanthenes/pharmacology , bcl-X Protein/genetics
9.
Indian J Cancer ; 2013 July-Sept; 50(3): 195-199
Article in English | IMSEAR | ID: sea-148648

ABSTRACT

BACKGROUND: Chemoresistance is an important factor determining the response of tumor to neoadjuvant chemotherapy (NACT). P-glycoprotein (P-gp) expression-mediated drug efflux is one of the mechanisms responsible for multi-drug resistance. Our study was aimed to determine the role of P-gp expression as a predictor of response to NACT in locally advanced breast cancer (LABC) patients. MATERIALS AND METHODS: P-gp expression was performed by real-time quantitative polymerase chain reaction [qRT-PCR] in 76 patients with LABC. Response to adriamycin-based regimen was assessed both clinically and with contrast enhanced computed tomography (CECT) scan before and after NACT. The significance of correlation between tumor and P-gp levels was determined with Chi-square test. RESULTS: Twenty-one had high and 55 had low P-gp expression. On analyzing P-gp expression with response by World Health Organization (WHO) criteria, statistical significance was obtained (P = 0.038). Similarly, assessment of P-gp expression with response by Response Evaluation in Solid Tumors (RECIST) criteria in 48 patients showed statistical significance (P = 0.0005). CONCLUSION: This study proves that P-gp expression is a determinant factor in predicting response to NACT. Finally, detection of P-gp expression status before initiation of chemotherapy can be used as a predictive marker for NACT response and will also aid in avoiding the toxic side effects of NACT in non-responders.


Subject(s)
Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant , Drug Resistance, Neoplasm/physiology , Female , Humans , Middle Aged , Neoadjuvant Therapy/methods , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Real-Time Polymerase Chain Reaction , Biomarkers, Tumor/analysis
10.
West Indian med. j ; 60(6): 666-668, Dec. 2011.
Article in English | LILACS | ID: lil-672831

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignant tumours and its five-year survival rate remains low. Autophagy is a catabolic process conserved among all eukaryotes ranging from yeast to mammals. Recently, many studies show that tumour cells can utilize autophagy as a cellular defence mechanism when facing metabolic stress. Thus, we hypothesize that autophagy may play an important role in the resistance of hepatocellular carcinomas to therapy. Although the exact role of autophagy on tumour cells is still complex and further studies are needed to prove the impact of autophagy on HCC, it suggests that autophagy may be a new therapeutic target for the resistance to therapy of HCC.


El carcinoma hepatocelular (CHC) es uno de los tumores malignos más comunes, y su tasa de super-vivencia a los cinco años sigue siendo baja. La autofagia es un proceso catabólico conservado en todos los eucariotas, que abarca desde las levaduras hasta los mamíferos. Recientemente, numerosos estudios han demostrado que las células tumorales pueden utilizar la autofagia como un mecanismo celular de defensa frente al estrés metabólico. De este modo, sostenemos la hipótesis de que la autofagia puede desempeñar un papel importante en la resistencia de los carcinomas hepatocelulares a la terapia. Aunque el papel exacto de la autofagia en las celulares tumorales sigue siendo complejo, y se requieren más estudios a fin de probar el impacto de la autofagia en el CHC, hay indicios de que la autofagia puede ser un nuevo objetivo terapéutico para la resistencia a la terapia del CHC.


Subject(s)
Animals , Humans , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/physiopathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Liver Neoplasms/drug therapy , Liver Neoplasms/physiopathology
11.
Experimental & Molecular Medicine ; : 233-242, 2004.
Article in English | WPRIM | ID: wpr-173480

ABSTRACT

Tumor hypoxia contributes to the progression of a malignant phenotype and resistance to ionizing radiation and anticancer drug therapy. Many of these effects in hypoxic tumor cells are mediated by expression of specific set of genes whose relation to therapy resistance is poorly understood. In this study, we revealed that DNA-dependent protein kinase (DNA-PK), which plays a crucial role in DNA double strand break repair, would be involved in regulation of hypoxia inducible factor-1 (HIF-1). HIF-1beta-deficient cells showed constitutively reduced expression and DNA-binding activity of Ku, the regulatory subunit of DNA-PK. Under hypoxic condition, the expression and activity of DNA- PK were markedly induced with a concurrent increase in HIF-1alpha expression. Our result also demonstrated that DNA-PK could directly interact with HIF- and especially DNA-PKcs, the catalytic subunit of DNA-PK, could be involved in phosphorylation of HIF-1alpha, suggesting the possibility that the enhanced expression of DNA- PK under hypoxic condition might attribute to modulate HIF-1alpha stabilization. Thus, the correlated regulation of DNA-PK with HIF-1 could contribute to therapy resistance in hypoxic tumor cells, and it provides new evidence for developing therapeutic strategies enhancing the efficacy of cancer therapy in hypoxic tumor cells.


Subject(s)
Humans , Antibodies/immunology , Cell Hypoxia , Cell Line, Tumor , DNA Helicases/immunology , DNA-Binding Proteins/genetics , Deferoxamine/pharmacology , Drug Resistance, Neoplasm/physiology , Immunoprecipitation , Neoplasms/enzymology , Nuclear Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Up-Regulation
12.
Article in English | IMSEAR | ID: sea-119614

ABSTRACT

Cancer as a multifactorial disease results in gain of immortality due to defective apoptosis. The primary mode of cell death by apoptosis induced by various modes of treatment often fail in vivo. The in vitro environment is less complex while the in vivo environment is influenced by various external regulatory signals besides the existence of multiple, parallel and independent apoptotic pathways. Further, specific preference for an apoptotic pathway in a certain cell type would significantly alter the apoptotic responses. Identification of defects in preferred pathways and choosing alternative and potentially inducible pathways would help in deciding on apoptosis-based treatment protocols. Mechanisms involved in the execution of apoptosis may also not be unique to apoptotic pathways since similar events, possibly with strict control, do occur during mitosis. Further evaluation may yield new dimensions to apoptosis and apoptosis-based therapy.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Mitochondria/physiology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/physiology , Signal Transduction/physiology , Tumor Suppressor Protein p53/physiology
13.
Experimental & Molecular Medicine ; : 87-92, 1998.
Article in English | WPRIM | ID: wpr-70154

ABSTRACT

MDR1 promoter has been shown to contain heat shock elements (HSE), and it has been reported that FM3A/M and P388/M MDR cells show a constitutively activated heat shock factor (HSF), suggesting that HSF might be an important target for reversing the multidrug resistance. Therefore, it was examined whether quercetin, which has been shown to interfere with the formation of the complex between HSE and HSF, and to downregulate the level of HSF1, can sensitize MDR cells against anticancer drugs by inhibition of HSF DNA-binding activity. In this study, quercetin appeared to inhibit the constitutive HSF DNA-binding activity and the sodium arsenite-induced HSF DNA-binding activity in the MDR cells. The basal and sodium arsenite-induced MDRCAT activities were remarkably suppressed by the treatment of quercetin. These results were well consistent with the finding that the treatment of quercetin decreased the expression level of P-gp, MDR1 gene product, in dose-dependent manner, and markedly increased the sensitivity of MDR cells to vincristine or vinblastine. These results suggest that quercetin can decrease the expression of P-gp via inhibition of HSF DNA-binding activity, and might be useful as a chemosensitizer in MDR cells.


Subject(s)
Mice , Animals , Antineoplastic Agents/pharmacology , Arsenites/pharmacology , Carcinoma/drug therapy , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/antagonists & inhibitors , Leukemia, Experimental/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Quercetin/pharmacology , Sodium Compounds/pharmacology , Tumor Cells, Cultured , Vinblastine/pharmacology , Vincristine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL