Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ann Oncol ; 28(8): 1738-1750, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28453615

RESUMO

BACKGROUND: The NHS Cancer Drugs Fund (CDF) was established in 2010 to reduce delays and improve access to cancer drugs, including those that had been previously appraised but not approved by NICE (National Institute for Health and Care Excellence). After 1.3 billion GBP expenditure, a UK parliamentary review in 2016 rationalized the CDF back into NICE. METHODS: This paper analyses the potential value delivered by the CDF according to six value criteria. This includes validated clinical benefits scales, cost-effectiveness criteria as defined by NICE and an assessment of real-world data. The analysis focuses on 29 cancer drugs approved for 47 indications that could be prescribed through the CDF in January 2015. RESULTS: Of the 47 CDF approved indications, only 18 (38%) reported a statistically significant OS benefit, with an overall median survival of 3.1 months (1.4-15.7 months). When assessed according to clinical benefit scales, only 23 (48%) and 9 (18%) of the 47 drug indications met ASCO and ESMO criteria, respectively. NICE had previously rejected 26 (55%) of the CDF approved indications because they did not meet cost-effectiveness thresholds. Four drugs-bevacizumab, cetuximab, everolimus and lapatinib-represented the bulk of CDF applications and were approved for a total of 18 separate indications. Thirteen of these indications were subsequently delisted by the CDF in January 2015 due to insufficient evidence for clinical benefit-data which were unchanged since their initial approval. CONCLUSIONS: We conclude the CDF has not delivered meaningful value to patients or society. There is no empirical evidence to support a 'drug only' ring fenced cancer fund relative to concomitant investments in other cancer domains such as surgery and radiotherapy, or other noncancer medicines. Reimbursement decisions for all drugs and interventions within cancer care should be made through appropriate health technology appraisal processes.


Assuntos
Antineoplásicos/economia , Acessibilidade aos Serviços de Saúde , Medicina Estatal , Análise Custo-Benefício , Custos de Medicamentos , Humanos , Reino Unido
2.
Breast Cancer Res Treat ; 160(1): 51-59, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27620882

RESUMO

PURPOSE: Improved therapies and imaging modalities are needed for the treatment of breast cancer brain metastases (BCBM). ANG1005 is a drug conjugate consisting of paclitaxel covalently linked to Angiopep-2, designed to cross the blood-brain barrier. We conducted a biomarker substudy to evaluate 18F-FLT-PET for response assessment. METHODS: Ten patients with measurable BCBM received ANG1005 at a dose of 550 mg/m2 IV every 21 days. Before and after cycle 1, patients underwent PET imaging with 18F-FLT, a thymidine analog, retention of which reflects cellular proliferation, for comparison with gadolinium-contrast magnetic resonance imaging (Gd-MRI) in brain metastases detection and response assessment. A 20 % change in uptake after one cycle of ANG1005 was deemed significant. RESULTS: Thirty-two target and twenty non-target metastatic brain lesions were analyzed. The median tumor reduction by MRI after cycle 1 was -17.5 % (n = 10 patients, lower, upper quartiles: -25.5, -4.8 %) in target lesion size compared with baseline. Fifteen of twenty-nine target lesions (52 %) and 12/20 nontarget lesions (60 %) showed a ≥20 % decrease post-therapy in FLT-PET SUV change (odds ratio 0.71, 95 % CI: 0.19, 2.61). The median percentage change in SUVmax was -20.9 % (n = 29 lesions; lower, upper quartiles: -42.4, 2.0 %), and the median percentage change in SUV80 was also -20.9 % (n = 29; lower, upper quartiles: -49.0, 0.0 %). Two patients had confirmed partial responses by PET and MRI lasting 6 and 18 cycles, respectively. Seven patients had stable disease, receiving a median of six cycles. CONCLUSIONS: ANG1005 warrants further study in BCBM. Results demonstrated a moderately strong association between MRI and 18F-FLT-PET imaging.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Paclitaxel/análogos & derivados , Peptídeos/uso terapêutico , Adulto , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Biomarcadores , Biomarcadores Tumorais , Neoplasias Encefálicas/diagnóstico , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Terapia Combinada , Feminino , Fluordesoxiglucose F18 , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Paclitaxel/uso terapêutico , Peptídeos/administração & dosagem , Peptídeos/efeitos adversos , Tomografia por Emissão de Pósitrons , Resultado do Tratamento
3.
Nat Cell Biol ; 2(10): 709-17, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11025661

RESUMO

Here we show that p53 protein is physically associated with tubulin in vivo and in vitro, and that it localizes to cellular microtubules. Treatment with vincristine or paclitaxel before DNA-damage or before leptomycin B treatment reduces nuclear accumulation of p53 and expression of mdm2 and p21. Overexpression of dynamitin or microinjection of anti-dynein antibody before DNA damage abrogates nuclear accumulation of p53. Our results indicate that transport of p53 along microtubules is dynein-dependent. The first 25 amino acids of p53 contain the residues that are essential for binding to microtubules. We propose that functional microtubules and the dynein motor protein participate in transport of p53 and facilitate its accumulation in the nucleus after DNA damage.


Assuntos
Transporte Ativo do Núcleo Celular , Dineínas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Complexo Dinactina , Ácidos Graxos Insaturados/farmacologia , Imunofluorescência , Humanos , Proteínas Motores Moleculares , Paclitaxel/farmacologia , Testes de Precipitina , Ligação Proteica , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas , Vincristina/farmacologia
4.
Science ; 275(5298): 343-9, 1997 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-8994024

RESUMO

Since 1990, the National Cancer Institute (NCI) has screened more than 60,000 compounds against a panel of 60 human cancer cell lines. The 50-percent growth-inhibitory concentration (GI50) for any single cell line is simply an index of cytotoxicity or cytostasis, but the patterns of 60 such GI50 values encode unexpectedly rich, detailed information on mechanisms of drug action and drug resistance. Each compound's pattern is like a fingerprint, essentially unique among the many billions of distinguishable possibilities. These activity patterns are being used in conjunction with molecular structural features of the tested agents to explore the NCI's database of more than 460,000 compounds, and they are providing insight into potential target molecules and modulators of activity in the 60 cell lines. For example, the information is being used to search for candidate anticancer drugs that are not dependent on intact p53 suppressor gene function for their activity. It remains to be seen how effective this information-intensive strategy will be at generating new clinically active agents.


Assuntos
Antineoplásicos/farmacologia , Biologia Computacional , Bases de Dados Factuais , Ensaios de Seleção de Medicamentos Antitumorais , Algoritmos , Antineoplásicos/química , Análise por Conglomerados , Redes de Comunicação de Computadores , Genes p53 , Humanos , Estrutura Molecular , Mutação , Software , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/fisiologia
5.
J Clin Invest ; 105(4): 533-9, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10683383

RESUMO

Cell-cycle checkpoint mechanisms, including the p53- and p21-dependent G(2) arrest that follows DNA damage, are often lost during tumorigenesis. We have exploited the ability of DNA-damaging drugs to elicit this checkpoint, and we show here that such treatment allows microtubule drugs, which cause cell death secondary to mitotic arrest, to kill checkpoint-deficient tumor cells while sparing checkpoint-competent cells. Low doses of the DNA-damaging drug doxorubicin cause predominantly G(2) arrest without killing HCT116 cells that harbor wt p53. Doxorubicin treatment prevented mitotic arrest, Bcl-2 phosphorylation, and cell death caused by paclitaxel, epothilones, and vinblastine. In contrast, doxorubicin enhanced cytotoxicity of FR901228, an agent that does not affect microtubules. Low doses of doxorubicin did not arrest p21-deficient clones of HCT116 cells and did not protect these cells from cytotoxicity caused by microtubule drugs, but cells in which p21 expression was restored enjoyed partial protection under these conditions. Moreover, in p53-deficient clones of HCT116 cells doxorubicin did not induce either p53 or p21 and provided no protection against paclitaxel-induced cytotoxicity. Therefore, (a) p53-dependent p21 induction caused by doxorubicin protects from microtubule drug-induced cytotoxicity, and (b) pretreatment with cytostatic doses of DNA-damaging drugs before treatment with microtubule drugs results in selective cytotoxicity to cancer cells with defective p53/p21-dependent checkpoint.


Assuntos
Antineoplásicos/farmacologia , Microtúbulos/efeitos dos fármacos , Mutagênicos/farmacologia , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Doxorrubicina/farmacologia , Interações Medicamentosas , Compostos de Epóxi/farmacologia , Humanos , Paclitaxel/farmacologia , Tiazóis/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Vimblastina/farmacologia
6.
J Clin Invest ; 95(5): 2205-14, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7738186

RESUMO

Identifying new chemotherapeutic agents and characterizing mechanisms of resistance may improve cancer treatment. The Anticancer Drug Screen of the National Cancer Institute uses 60 cell lines to identify new agents. Expression of mdr-1/P-glycoprotein was measured by quantitative PCR. Expression was detected in 39 cell lines; the highest levels were in renal and colon carcinomas. Expression was also detected in all melanomas and central nervous system tumors, but in only one ovarian carcinoma and one leukemia cell line. Using a modified version of the COMPARE program, a high correlation was found between expression of mdr-1 and cellular resistance to a large number of compounds. Evidence that these compounds are P-glycoprotein substrates includes: (a) enhancement of cytotoxicity by verapamil; (b) demonstration of cross-resistance in a multidrug-resistant cell line, (c) ability to antagonize P-glycoprotein, increasing vinblastine accumulation by decreasing efflux; and (d) inhibition of photoaffinity labeling by azidopine. Identification of many heretofore unrecognized compounds as substrates indicates that P-glycoprotein has a broader substrate specificity than previously recognized. This study confirms the validity of this novel approach and provides the basis for similar studies examining a diverse group of gene products, including other resistance mechanisms, putative drug targets, and genes involved in the cell cycle and apoptosis.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Antineoplásicos/toxicidade , Resistência a Múltiplos Medicamentos/genética , Expressão Gênica , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/análise , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Sistema Nervoso Central , Neoplasias do Colo , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Neoplasias Renais , Neoplasias Pulmonares , Melanoma , National Institutes of Health (U.S.) , Neoplasias Ovarianas , Células Tumorais Cultivadas , Estados Unidos , Verapamil/farmacologia
7.
J Clin Invest ; 99(8): 1947-57, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9109439

RESUMO

Drug resistance, a major obstacle to cancer chemotherapy, can be mediated by MDR-1/P-glycoprotein. Deletion of the first 68 residues of MDR-1 in an adriamycin-selected cell line after a 4;7 translocation, t(4q;7q), resulted in a hybrid mRNA containing sequences from both MDR-1 and a novel chromosome 4 gene. Further selection resulted in amplification of a hybrid gene. Expression of the hybrid mRNA was controlled by the chromosome 4 gene, providing a model for overexpression of MDR-1. Additional hybrid mRNAs in other drug-selected cell lines and in patients with refractory leukemia, with MDR-1 juxtaposed 3' to an active gene, establishes random chromosomal rearrangements with overexpression of hybrid MDR-1 mRNAs as a mechanism of acquired drug resistance.


Assuntos
Rearranjo Gênico , Genes MDR , Sequência de Aminoácidos , Antibióticos Antineoplásicos/farmacologia , Sequência de Bases , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 7/genética , Primers do DNA/genética , DNA de Neoplasias/genética , Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Hibridização Genética , Hibridização in Situ Fluorescente , Dados de Sequência Molecular , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Neoplásico/genética , Ativação Transcricional , Translocação Genética , Células Tumorais Cultivadas
8.
J Natl Cancer Inst ; 92(8): 642-7, 2000 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-10772682

RESUMO

BACKGROUND: Cisplatin (cis-diamminedichloroplatinum) is one of the most active agents against a broad range of malignancies, including ovarian cancer. Cisplatin resistance appears to be associated with several molecular alterations, including overexpression of metallothionein, a metal-binding protein. In the present study, we attempted to take advantage of metallothionein overexpression to overcome cisplatin resistance. METHODS: Using a virus-free system (liposomes), we sought to express the suicide gene, thymidine kinase (TK), driven by the promoter of the human metallothionein IIa (hMTIIa) gene using the pMT-TK plasmid. We used cisplatin-resistant human ovarian carcinoma cells as a model. RESULTS: We first analyzed metallothionein expression using a ribonuclease protection assay. In comparison to parental cells, the cisplatin-resistant cells were found to have increased expression of metallothionein messenger RNA (mRNA). Metallothionein overexpression in these cells was not associated with an increased copy number of the hMTIIa gene or with different transfection efficiencies. Furthermore, we showed by reverse transcription-polymerase chain reaction analysis that transfection of the pMT-TK plasmid results in a 56-fold higher expression of thymidine kinase mRNA in cisplatin-resistant cells compared with parental cells, consistent with increased metallothionein promoter-mediated transactivation in the cisplatin-resistant cells. Transfection of resistant cells with pMT-TK or a control plasmid (pCD3-TK) resulted in a marked sensitization to ganciclovir, with a 50% cell growth-inhibitory concentration (IC(50)) of 20 microg/mL and 9 microg/mL, respectively. Transfections of the cisplatin-sensitive cells resulted in no sensitization to ganciclovir with pMT-TK (IC(50) 200 microg/mL) and a high sensitization with pCD3-TK (IC(50) = 6 microg/mL). CONCLUSION: These studies suggest that pMT-TK gene therapy may provide an alternative treatment for cisplatin-refractory ovarian tumors.


Assuntos
Cisplatino/farmacologia , Terapia Genética , Metalotioneína/genética , Neoplasias Ovarianas/genética , Regiões Promotoras Genéticas , Timidina Quinase/genética , Resistencia a Medicamentos Antineoplásicos , Feminino , Ganciclovir/farmacologia , Humanos , Lipossomos/administração & dosagem , Pró-Fármacos/metabolismo , RNA Mensageiro/análise , Simplexvirus/enzimologia , Timidina Quinase/metabolismo , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
9.
Cancer Res ; 57(22): 5086-92, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9371507

RESUMO

The efficacy of all chemotherapeutic agents is limited by the occurrence of drug resistance. For etoposide (VP-16), increased expression of MDR-1 or MRP and alterations in topoisomerase IIalpha have been shown to confer tolerance. To further understand resistance to VP-16, three sublines, designated MCF-7-VP17, ZR-75B-VP13, and MDA-MB-231-VP7, were initially isolated as single clones from parental cells by exposure to VP-16. Subsequently, a population of cells from each subline was exposed to 3-fold higher drug concentrations, allowing stable sublines to be established at higher extracellular drug concentrations. Characterization of the resistant sublines demonstrates the adaptation that occurs with advancing drug concentrations during in vitro selections. Reduced topoisomerase II mRNA and protein levels were observed in the initial isolates. This reduction was accompanied by a decrease in topoisomerase II activity and cellular growth rate and was associated with 6-314-fold resistance to topoisomerase II poisons. With advancing resistance, MRP expression increased and VP-16 accumulation decreased. This adaptation allowed for partial restoration of topoisomerase II activity as a result of increased expression (MCF-7-VP17 and ZR-75B-VP13) or hyperphosphorylation (MDA-MB-231-VP7), with a resultant increase in growth rate. In MDA-MB-231-VP7 cells, hyperphosphorylation coincided with increased casein kinase II mRNA and protein levels, suggesting a role for this kinase in the acquired hyperphosphorylation. In this cell line, hyperphosphorylation mediated the increased activity despite a fall in topoisomerase IIalpha protein levels secondary to an acquired 600-bp deletion in one topoisomerase IIalpha allele, which resulted in reduced protein levels. In all three sublines, high levels of resistance were attained as a result of synergism between the reduced topoisomerase IIalpha levels and MRP overexpression. These studies demonstrate how cellular adaptation to increasing drug pressure occurs and how more than one mechanism can contribute to the resistant phenotype when increasing selecting pressure is applied. Reduced expression of topoisomerase II is sufficient to confer substantial resistance early in the selection process, with synergy from MRP overexpression helping to confer high levels of resistance.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Resistência a Múltiplos Medicamentos/fisiologia , Etoposídeo/metabolismo , Ensaio Tumoral de Célula-Tronco , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , DNA Topoisomerases Tipo II/genética , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos , Etoposídeo/farmacologia , Humanos , Fenótipo , Fosforilação , RNA Mensageiro/metabolismo , Deleção de Sequência , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
10.
Cancer Res ; 61(17): 6328-30, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11522619

RESUMO

The presence of coxsackie and adenovirus receptor (CAR) and alpha(v) integrin on cell surfaces is required for efficient adenovirus infection. Treatment of cells with the histone deacetylase inhibitor FR901228 (depsipeptide) increased CAR and alpha(v) integrin RNA levels in six cancer cell lines. Sodium butyrate and trichostatin A, other histone deacetylase inhibitors, caused similar increases. Cells treated with FR901228 prior to infection had a 4-10-fold increase in transgene expression from a beta-galactosidase-expressing adenoviral vector. These studies suggest that FR901228 increases the efficiency of adenoviral transgene expression and may be useful in cancer gene therapy.


Assuntos
Adenoviridae/genética , Antibacterianos/farmacologia , Antígenos CD/biossíntese , Depsipeptídeos , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Peptídeos Cíclicos , Receptores Virais/biossíntese , Transgenes/efeitos dos fármacos , Antígenos CD/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Expressão Gênica/efeitos dos fármacos , Humanos , Integrina alfaV , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias/virologia , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Receptores Virais/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
11.
Cancer Res ; 60(13): 3425-8, 2000 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910051

RESUMO

Lack of selectivity in the killing of tumor and normal cells is a major obstacle in cancer therapy. By inhibiting normal but not autonomous cell growth, we exploited the differences in cell cycle regulation to achieve a selective protection of nonautonomous cells against paclitaxel and other microtubule-active drugs. Tubulin polymerization, a primary effect of paclitaxel, can be dissociated from Bcl-2 phosphorylation and cytotoxicity in HL-60 cells. Growth arrest prevented paclitaxel-induced Bcl-2 phosphorylation and apoptosis without affecting paclitaxel-induced tubulin polymerization. We abrogated the effects of paclitaxel on MCF-10A immortalized breast cells, while preserving its effects on MCF-7 cancer cells. Unlike MCF-7 cells, MCF-10A cells were arrested by epidermal growth factor withdrawal, precluding paclitaxel-induced Bcl-2 phosphorylation. Furthermore, the inhibition of the epidermal growth factor receptor kinase with low doses of AG1478 arrested growth of MCF-10A but not MCF-7 cells. Pretreatment with AG1478 did not affect paclitaxel-induced Bcl-2/Raf-1 phosphorylation in MCF-7 but abrogated such phosphorylation in MCF-10A. Exploitation of growth factor dependency may allow the protection of normal cells from microtubule-active drugs.


Assuntos
Ciclo Celular/fisiologia , Microtúbulos/efeitos dos fármacos , Paclitaxel/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Tirfostinas/toxicidade , Apoptose/efeitos dos fármacos , Neoplasias da Mama , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Fator de Crescimento Epidérmico/farmacologia , Feminino , Células HL-60 , Humanos , Mitose/efeitos dos fármacos , Fosforilação , Quinazolinas , Acetato de Tetradecanoilforbol/farmacologia , Tubulina (Proteína)/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas
12.
Cancer Res ; 58(15): 3331-8, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9699663

RESUMO

The oncogenic protein Bcl-2 functions as a potent inhibitor of programmed cell death. This survival activity has been shown in some settings to be influenced by the Bcl-2 phosphorylation state. It has been demonstrated that treatment with microtubule-targeted agents results in phosphorylation of both Raf-1 kinase and Bcl-2. The Bcl-2-related family member Bcl-xL also exhibits a death suppressive activity, but its potential for phosphorylation following exposure to drugs that interact with microtubules has not been evaluated. Several tumor cell lines with low or undetectable levels of Bcl-2 protein expression were found to express Bcl-xL. A more slowly migrating Bcl-xL band was observed on immunoblots after cells were treated with microtubule-targeted agents. The appearance of this band was responsive to dose and was absent when the cell lysates were treated with lambda protein phosphatase. Using a Bcl-xL-specific monoclonal antibody, the phosphorylated form of Bcl-xL was immunoprecipitated from cells treated with paclitaxel and metabolically labeled with 32P-labeled inorganic orthophosphate. Herein, we report that Bcl-xL is phosphorylated in malignant cells after incubation with agents that target tubulin, including paclitaxel, vincristine, vinblastine, colchicine, and nocodazole. Moreover, paclitaxel-resistant ovarian carcinoma cell lines that have mutations in tubulin failed to exhibit phosphorylation of Bcl-xL after paclitaxel exposure, but they did demonstrate Bcl-xL phosphorylation in the presence of other tubulin-targeting agents. As observed for Bcl-2, phosphorylation of Bcl-xL was accompanied by phosphorylation of Raf-1. Interestingly, phosphorylation of these three proteins failed to occur or was much less pronounced when cells grown at high density were challenged with drug. Also, reduced Raf-1 expression, observed after treatment of cells with geldanamycin prior to and during incubation with the microtubule-active drugs, correlated with diminished Bcl-xL phosphorylation. Taken together, these results suggest that Bcl-xL, like Bcl-2, is phosphorylated by agents that disrupt microtubule architecture. By analogy with Bcl-2, this phosphorylation may play a critical role in modulating Bcl-xL function and may be an important determinant of microtubule-directed chemotherapeutic efficacy in human tumors.


Assuntos
Microtúbulos/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Benzoquinonas , Contagem de Células , Resistencia a Medicamentos Antineoplásicos , Eletroforese em Gel de Poliacrilamida , Humanos , Lactamas Macrocíclicas , Microtúbulos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/ultraestrutura , Paclitaxel/farmacologia , Fosfoproteínas Fosfatases/metabolismo , Radioisótopos de Fósforo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-raf/biossíntese , Proteínas Proto-Oncogênicas c-raf/metabolismo , Quinonas/farmacologia , Dodecilsulfato de Sódio , Tubulina (Proteína)/efeitos dos fármacos , Células Tumorais Cultivadas , Proteína bcl-X
13.
Cancer Res ; 57(1): 130-5, 1997 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8988053

RESUMO

Recent studies have shown that paclitaxel leads to activation of Raf-1 kinase and have suggested that this activation is essential for bcl-2 phosphorylation and apoptosis. In the present study, we demonstrate that, in addition to paclitaxel, other agents that interact with tubulin and microtubules also induce Raf-1/bcl-2 phosphorylation, whereas DNA-damaging drugs, antimetabolites, and alkylating agents do not. Activation of Raf-1 kinase by paclitaxel is linked to tubulin polymerization; the effect is blunted in paclitaxel-resistant cells, the tubulin of which does not polymerize following the addition of paclitaxel. In contrast, vincristine and vinblastine, drugs to which the paclitaxel-resistant cells retain sensitivity were able to bring about Raf-1 phosphorylation. The requirement for disruption of microtubules in this signaling cascade was strengthened further using paclitaxel analogues by demonstrating a correlation between tubulin polymerization, Raf-1/bcl-2 phosphorylation, and cytotoxicity. Inhibition of RNA or protein synthesis prevents Raf-1 activation and bcl-2 phosphorylation, suggesting that an intermediate protein(s) acts upstream of Raf-1 in this microtubule damage-activating pathway. A model is proposed that envisions a pathway of Raf-1 activation and bcl-2 phosphorylation following disruption of microtubular architecture, serving a role similar to p53 induction following DNA damage.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Microtúbulos/efeitos dos fármacos , Paclitaxel/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tubulina (Proteína)/efeitos dos fármacos , Antineoplásicos/farmacologia , Resistência a Medicamentos , Ativação Enzimática , Feminino , Humanos , Fosforilação , Polímeros/metabolismo , Inibidores de Proteínas Quinases , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
14.
Cancer Res ; 59(1): 8-13, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9892175

RESUMO

Reports of multiple distinct mitoxantrone-resistant sublines without overexpression of P-glycoprotein or the multidrug-resistance associated protein have raised the possibility of the existence of another major transporter conferring drug resistance. In the present study, a cDNA library from mitoxantrone-resistant S1-M1-80 human colon carcinoma cells was screened by differential hybridization. Two cDNAs of different lengths were isolated and designated MXR1 and MXR2. Sequencing revealed a high degree of homology for the cDNAs with Expressed Sequence Tag sequences previously identified as belonging to an ATP binding cassette transporter. Homology to the Drosophila white gene and its homologues was found for the predicted amino acid sequence. Using either cDNA as a probe in a Northern analysis demonstrated high levels of expression in the S1-M1-80 cells and in the human breast cancer subline, MCF-7 AdVp3000. Levels were lower in earlier steps of selection, and in partial revertants. The gene is amplified 10-12-fold in the MCF-7 AdVp3000 cells, but not in the S1-M1-80 cells These studies are consistent with the identification of a new ATP binding cassette transporter, which is overexpressed in mitoxantrone-resistant cells.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Antineoplásicos/farmacologia , DNA Complementar/genética , Resistencia a Medicamentos Antineoplásicos/genética , Mitoxantrona/farmacologia , Sequência de Bases , Clonagem Molecular , DNA Complementar/isolamento & purificação , Regulação Neoplásica da Expressão Gênica , Humanos , Dados de Sequência Molecular , Análise de Sequência de DNA , Células Tumorais Cultivadas
15.
Cancer Res ; 59(23): 5938-46, 1999 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10606239

RESUMO

The mitoxantrone resistance (MXR) gene encodes a recently characterized ATP-binding cassette half-transporter that confers multidrug resistance. We studied resistance to the camptothecins in two sublines expressing high levels of MXR: S1-M1-80 cells derived from parental S1 colon cancer cells and MCF-7 AdVp3,000 isolated from parental MCF-7 breast cancer cells. Both cell lines were 400- to 1,000-fold more resistant to topotecan, 9-amino-20(S)-camptothecin, and the active metabolite of irinotecan, 7-ethyl-10-hydroxycamptothecin (SN-38), than their parental cell lines. The cell lines demonstrated much less resistance to camptothecin and to several camptothecin analogues. Reduced accumulation and energy-dependent efflux of topotecan was demonstrated by confocal microscopy. A significant reduction in cleavable complexes in the resistant cells could be observed after SN-38 treatment but not after camptothecin treatment. In addition to topotecan and SN-38, MXR-overexpressing cells are highly resistant to mitoxantrone and epirubicin. Because these compounds are susceptible to glucuronidation, we examined UDP-glucurono-syltransferase (UGT) activity in parental and resistant cells by TLC. Glucuronides were found at equal levels in both parental and resistant colon cancer cell lines for epirubicin and to a lesser extent for SN-38 and mitoxantrone. Low levels of glucuronidation could also be detected in the resistant breast cancer cells. These results were confirmed by analysis of the UGT1A family mRNAs. We thus conclude that colon and breast cancer cells have a capacity for glucuronidation that could contribute to intrinsic drug resistance in colon cancer cells and may be acquired in breast cancer cells. The lack of selection for higher levels of UGT capacity in the colon cells suggests that high levels of expression of MXR alone are sufficient to confer resistance to the camptothecins.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/toxicidade , Camptotecina/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Resistência a Múltiplos Medicamentos , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Mitoxantrona/toxicidade , Antineoplásicos Fitogênicos/toxicidade , Neoplasias da Mama , Camptotecina/análogos & derivados , Dano ao DNA , Epirubicina/toxicidade , Feminino , Humanos , Irinotecano , Microscopia Confocal , Reação em Cadeia da Polimerase , Topotecan/toxicidade , Células Tumorais Cultivadas
16.
Oncogene ; 20(29): 3806-13, 2001 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-11439344

RESUMO

Paclitaxel (PTX), a microtubule-active agent, blocks cell proliferation by inhibiting mitotic progression leading to mitotic and postmitotic arrest and cell death. Here we demonstrate for the first time that very low concentrations of PTX (3-6 nM) can completely inhibit cell proliferation without arresting cells at mitosis. At these low concentrations that are insufficient to inhibit mitotic progression, PTX induced both p53 and p21 causing G1 and G2 arrest in A549. In contrast, low PTX concentrations failed to induce G1 and G2 arrest in A549/E6 cells, that do not express p53. Furthermore, we observed that the levels of p53 and p21 induced by adriamycin and by low concentrations of PTX in A549 cells were comparable. This observation led us to conclude that low concentrations of PTX can induce p53 and p21 sufficiently to cause G1 and G2. Many other cell lines, including HCT116 cells, do not readily upregulate p53 in response to PTX, and therefore undergo exclusively mitotic and postmitotic arrest after PTX treatment. At low concentrations that do not cause mitotic arrest, PTX did not significantly inhibit proliferation of these cells. In HCT116 cells, loss of p53 (HCT/p53(-/-)) or p21 (HCT/p21(-/-)) affects both Bax and Bcl-2 expression. In cells lacking p53, levels of Bax and p21 were decreased. In cells lacking p21, levels of wt p53 were highly increased to compensate for the loss of p21. This in turn results in upregulation of Bax and downregulation of Bcl-2 resulting in an increase of the apoptotic Bax/Bcl2 ratio consistent with increased sensitivity of these cells to apoptotic stimuli. High levels of p53 and Bax/Bcl-2 ratio can also explain why loss of p21 is rarely found in human cancer.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Ciclinas/metabolismo , Inibidores do Crescimento/farmacologia , Mitose/efeitos dos fármacos , Paclitaxel/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Inibidor de Quinase Dependente de Ciclina p21 , Relação Dose-Resposta a Droga , Fase G1 , Fase G2 , Humanos , Interfase , Mitose/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2
17.
Oncogene ; 19(27): 3078-85, 2000 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-10871860

RESUMO

The efficacy of anticancer therapy is limited by the development of drug resistance. While the role of p53 in the intrinsic sensitivity of human cancer cells to paclitaxel (PTX) remains controversial, its role in acquired paclitaxel resistance has never been addressed. In this study we examined the p53 status of three paclitaxel selected human ovarian carcinoma sublines, resistant to paclitaxel due to acquired beta-tubulin mutations which impair paclitaxel's interaction with tubulin. In contrast to parental cells which have wt p53, in all PTX-resistant sublines p53 was functionally inactive. Two of the resistant sublines expressed high levels of transcriptionally inactive p53 protein, each with a distinct point mutation in codons 236 and 239 of the DNA binding domain. The third subline presented a novel p53 pseudo-null phenotype as a result of markedly decreased wt p53 mRNA expression. Introduction of ectopic wt p53 had no effect on PTX sensitivity in both parental and resistant cells, while it induced p21WAF1/CIP1, demonstrating an intact p53 pathway. While PTX resistance is primarily conferred by the tubulin mutations, the loss of functional p53 observed in all clones, suggests that this loss may facilitate the development of resistance potentially by providing a clonal advantage which promotes the isolation of paclitaxel resistant cells.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Proteínas de Neoplasias/genética , Paclitaxel/farmacologia , Tubulina (Proteína)/genética , Proteína Supressora de Tumor p53/genética , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/genética , Amplificação de Genes , Vetores Genéticos , Humanos , Hibridização in Situ Fluorescente , Mutação , Proteínas de Neoplasias/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
18.
J Clin Oncol ; 19(3): 832-42, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11157037

RESUMO

PURPOSE: PSC 833 (valspodar) is a second-generation P-glycoprotein (Pgp) antagonist developed to reverse multidrug resistance. We conducted a phase I study of a 7-day oral administration of PSC 833 in combination with paclitaxel, administered as a 96-hour continuous infusion. PATIENTS AND METHODS: Fifty patients with advanced cancer were enrolled onto the trial. PSC 833 was administered orally for 7 days, beginning 72 hours before the start of the paclitaxel infusion. Paclitaxel dose reductions were planned because of the pharmacokinetic interactions known to occur with PSC 833. RESULTS: In combination with PSC 833, maximum-tolerated doses were defined as paclitaxel 13.1 mg/m(2)/d continuous intravenous infusion (CIVI) for 4 days without filgrastim, and paclitaxel 17.5 mg/m(2)/d CIVI for 4 days with filgrastim support. Dose-limiting toxicity for the combination was neutropenia. Statistical analysis of cohorts revealed similar mean steady-state concentrations (C(pss)) and areas under the concentration-versus-time curve (AUCs) when patients received paclitaxel doses of 13.1 or 17.5 mg/m(2)/d for 4 days with PSC 833, as when they received a paclitaxel dose of 35 mg/m(2)/d for 4 days without PSC 833. However, the effect of PSC 833 on paclitaxel pharmacokinetics varied greatly among individual patients, although a surrogate assay using CD56+ cells suggested inhibition of Pgp was complete or nearly complete at low concentrations of PSC 833. Responses occurred in three of four patients with non-small-cell lung cancer, and clinical benefit occurred in five of 10 patients with ovarian carcinoma. CONCLUSION: PSC 833 in combination with paclitaxel can be administered safely to patients provided the paclitaxel dose is reduced to compensate for the pharmacokinetic interaction. Surrogate studies with CD56+ cells indicate that the maximum-tolerated dose for PSC 833 gives serum levels much higher than those required to block Pgp. The variability in paclitaxel pharmacokinetics, despite complete inhibition of Pgp in the surrogate assay, suggests that other mechanisms, most likely related to P450, contribute to the pharmacokinetic interaction. Future development of combinations such as this should include strategies to predict pharmacokinetics of the chemotherapeutic agent. This in turn will facilitate dosing to achieve comparable CPss and AUCs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias/tratamento farmacológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Administração Oral , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Antígeno CD56/biossíntese , Ciclosporinas/administração & dosagem , Ciclosporinas/efeitos adversos , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Corantes Fluorescentes/farmacocinética , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Paclitaxel/farmacocinética , Rodaminas/farmacocinética , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
19.
Leukemia ; 13(7): 1028-36, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10400418

RESUMO

Raf-1 activation and Bcl-2 hyperphosphorylation following treatment with paclitaxel (Taxol) or other microtubule-active drugs is associated with mitotic arrest. Here we show that microtubule-active drugs do not activate the mitogen-activated protein kinase (MAPK) pathway in leukemia cells. PD98059, a MEK inhibitor, and SB202190, a p38 MAP kinase inhibitor, do not abrogate Bcl-2 phosphorylation nor apoptosis. Simultaneously with PARP cleavage, paclitaxel induces cleavage of Bcl-2 protein yielding a potentially pro-apoptotic 22 kDa product. In comparison, the stimulation of Raf-1 by phorbol ester (TPA) activates the MAPK pathway, causes MAPK-dependent p21WAF1/CIP1 induction, Rb dephosphorylation and growth arrest without Bcl-2 phosphorylation or apoptosis. Like TPA, cAMP induces p21WAF1/CIP1 but does not cause Bcl-2 phosphorylation. MEKK1 and Ras, upstream activators of JNK and ERK MAPK, also fail to induce Bcl-2 hyperphosphorylation. Although Lck tyrosine kinase has been recently implicated in Raf-1 activation during mitotic arrest, microtubule-active drugs induce Raf-1/Bcl-2 hyperphosphorylation and apoptosis in a Lck-deficient Jurkat cells. Therefore, microtubule-active drugs induce apoptosis which is associated with Raf-1 and Bcl-2 phosphorylation and Bcl-2 cleavage but is independent of the MAPK pathway. In contrast, TPA-activated MAPK pathway causes p21WAF1/CIP1-dependent growth arrest without apoptosis.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Leucemia Mieloide/tratamento farmacológico , MAP Quinase Quinase Quinase 1 , Microtúbulos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Apoptose/efeitos dos fármacos , Genes ras , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Paclitaxel/uso terapêutico , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Acetato de Tetradecanoilforbol/uso terapêutico , Células Tumorais Cultivadas
20.
Leukemia ; 15(10): 1537-43, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11587211

RESUMO

The Bcr-Abl fusion protein drives leukemogenesis and can render leukemia cells resistant to conventional chemotherapy. Geldanamycin (GA), a drug which destabilizes Hsp90-associated proteins, depletes cells of Bcr-Abl, an Hsp90 client, but not of Abl. Both HL60 cells transfected with Bcr-Abl and naturally Ph1-positive K562 leukemia cells are resistant to most cytotoxic drugs, but were found to be sensitive to GA. Furthermore, GA sensitized Bcr-Abl-expressing cells to doxorubicin (DOX) and paclitaxel (PTX). In contrast, in parental HL60 cells, 90 nM GA inhibited PARP cleavage, nuclear fragmentation, and cell death caused by 500 ng/ml DOX. Like GA, STI 571 (an inhibitor of the Abl kinase) sensitized Bcr-Abl-expressing cells to DOX. Unlike GA, STI 571 did not antagonize the cytotoxic effects of DOX in parental HL60 cells. These results indicate that sensitization of Bcr-Abl-expressing cells, but not desensitization of HL60 cells, depends on inhibition of Bcr-Abl. Thus, GA differentially affects leukemia cells depending on their Bcr-Abl expression and selectively increases apoptosis in Bcr-Abl-expressing cells.


Assuntos
Proteínas de Fusão bcr-abl/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Leucemia/patologia , Quinonas/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzoquinonas , Doxorrubicina/farmacologia , Interações Medicamentosas , Resistência a Medicamentos , Proteínas de Fusão bcr-abl/biossíntese , Humanos , Lactamas Macrocíclicas , Leucemia/metabolismo , Paclitaxel/farmacologia , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA