Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Oral Oncol ; 69: 38-45, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28559019

RESUMO

OBJECTIVE: Src family kinase (SFK) activation circumvents epidermal growth factor receptor (EGFR) targeting in head and neck squamous cell carcinoma (HNSCC); dual SFK-EGFR targeting could overcome cetuximab resistance. PATIENTS AND METHODS: We conducted a Simon two-stage, phase II trial of the SFK inhibitor, dasatinib, and cetuximab in biomarker-unselected patients with cetuximab-resistant, recurrent/metastatic HNSCC. Pre- and post-treatment serum levels of interleukin-6 (IL6) were measured by ELISA. HNSCC cell lines were assessed for viability and effects of IL6 modulation following dasatinib-cetuximab treatment. RESULTS: In the first stage, 13 patients were evaluable for response: 7 had progressive and 6 had stable disease (SD). Enrollment was halted for futility, and biomarker analysis initiated. Low serum IL6 levels were associated with SD (raw p=0.028, adjusted p=0.14) and improved overall survival (p=0.010). The IL6 classifier was validated in a separate trial of the same combination, but was unable to segregate survival risk in a clinical trial of cetuximab and bevacizumab suggesting serum IL6 may be specific for the dasatinib-cetuximab combination. Enhanced in vitro HNSCC cell death was observed with dasatinib-cetuximab versus single agent treatment; addition of IL6-containing media abrogated this effect. CONCLUSION: Clinical benefit and overall survival from the dasatinib-cetuximab combination were improved among patients with low serum IL6. Preclinical studies support IL6 as a modifier of dasatinib-cetuximab response. In the setting of clinical cetuximab resistance, serum IL6 is a candidate predictive marker specific for combined dasatinib-cetuximab. The trial was modified and redesigned as a biomarker-enriched Phase II study enrolling patients with undetectable IL6.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Cetuximab/administração & dosagem , Dasatinibe/administração & dosagem , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Interleucina-6/sangue , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carcinoma de Células Escamosas/sangue , Resistencia a Medicamentos Antineoplásicos , Feminino , Neoplasias de Cabeça e Pescoço/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma de Células Escamosas de Cabeça e Pescoço , Análise de Sobrevida
2.
Ann Oncol ; 27(8): 1594-600, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27177865

RESUMO

BACKGROUND: We previously reported the safety of concurrent cetuximab, an antibody against epidermal growth factor receptor (EGFR), pemetrexed, and radiation therapy (RT) in patients with locally advanced squamous cell carcinoma of the head and neck (SCCHN). In this non-comparative phase II randomized trial, we evaluated this non-platinum combination with or without bevacizumab, an inhibitor of vascular endothelial growth factor (VEGF). PATIENTS AND METHODS: Patients with previously untreated stage III-IVB SCCHN were randomized to receive: conventionally fractionated radiation (70 Gy), concurrent cetuximab, and concurrent pemetrexed (arm A); or the identical regimen plus concurrent bevacizumab followed by bevacizumab maintenance for 24 weeks (arm B). The primary end point was 2-year progression-free survival (PFS), with each arm compared with historical control. Exploratory analyses included the relationship of established prognostic factors to PFS and quality of life (QoL). RESULTS: Seventy-eight patients were randomized: 66 oropharynx (42 HPV-positive, 15 HPV-negative, 9 unknown) and 12 larynx; 38 (49%) had heavy tobacco exposure. Two-year PFS was 79% [90% confidence interval (CI) 0.69-0.92; P < 0.0001] for arm A and 75% (90% CI 0.64-0.88; P < 0.0001) for arm B, both higher than historical control. No differences in PFS were observed for stage, tobacco history, HPV status, or type of center (community versus academic). A significantly increased rate of hemorrhage occurred in arm B. SCCHN-specific QoL declined acutely, with marked improvement but residual symptom burden 1 year post-treatment. CONCLUSIONS: RT with a concurrent non-platinum regimen of cetuximab and pemetrexed is feasible in academic and community settings, demonstrating expected toxicities and promising efficacy. Adding bevacizumab increased toxicity without apparent improvement in efficacy, countering the hypothesis that dual EGFR-VEGF targeting would overcome radiation resistance, and enhance clinical benefit. Further development of cetuximab, pemetrexed, and RT will require additional prospective study in defined, high-risk populations where treatment intensification is justified.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Cetuximab/administração & dosagem , Receptores ErbB/genética , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Pemetrexede/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/genética , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Bevacizumab/administração & dosagem , Bevacizumab/efeitos adversos , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/radioterapia , Cetuximab/efeitos adversos , Intervalo Livre de Doença , Receptores ErbB/antagonistas & inibidores , Feminino , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Masculino , Terapia de Alvo Molecular , Estadiamento de Neoplasias , Pemetrexede/efeitos adversos , Qualidade de Vida , Carcinoma de Células Escamosas de Cabeça e Pescoço , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
3.
Ann Oncol ; 24(1): 220-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22898037

RESUMO

BACKGROUND: We evaluated combined targeting with cetuximab, an anti-epidermal growth factor receptor (EGFR) monoclonal antibody, and bevacizumab, an anti-vascular endothelial growth factor (VEGF) monoclonal antibody, in squamous cell carcinoma of the head and neck (SCCHN). PATIENTS AND METHODS: The combination was studied in human endothelial cells and head and neck and lung cancer xenograft model systems. Patients with recurrent or metastatic SCCHN were treated with weekly cetuximab and bevacizumab, 15 mg/kg on day 1 given intravenously every 21 days, until disease progression. Analysis of tumor biomarkers and related serum cytokines was performed. RESULTS: Cetuximab plus bevacizumab enhanced growth inhibition both in vitro and in vivo, and resulted in potent reduction in tumor vascularization. In the clinical trial, 46 eligible patients were enrolled. The objective response rate was 16% and the disease control rate 73%. The median progression-free survival and overall survival were 2.8 and 7.5 months, respectively. Grade 3-4 adverse events were expected and occurred in less than 10% of patients. transforming growth factor alpha, placenta-derived growth factor, EGFR, VEGFR2 increased and VEGF decreased after treatment but did not correlate with treatment efficacy. CONCLUSIONS: Cetuximab and bevacizumab are supported by preclinical observations and are well tolerated and active in previously treated patients with SCCHN.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Metástase Neoplásica , Recidiva Local de Neoplasia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados/administração & dosagem , Bevacizumab , Biomarcadores Tumorais , Carcinoma de Células Escamosas/patologia , Cetuximab , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Pessoa de Meia-Idade
4.
Ann Oncol ; 22(11): 2482-2488, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21363880

RESUMO

BACKGROUND: We studied the combination of pemetrexed, a multi-targeted antifolate, and cetuximab, an mAb against the epidermal growth factor receptor, with radiotherapy in poor prognosis head and neck cancer. PATIENTS AND METHODS: Patients received pemetrexed on days 1, 22, and 43 on a dose-escalation scheme with starting level (0) 350 mg/m(2) (level -1, 200 mg/m(2); level +1, 500 mg/m(2)) with concurrent radiotherapy (2 Gy/day) and cetuximab in two separate cohorts, not previously irradiated (A) and previously irradiated (B), who received 70 and 60-66 Gy, respectively. Genetic polymorphisms of thymidylate synthase and methylenetetrahydrofolate reductase were evaluated. RESULTS: Thirty-two patients were enrolled. The maximum tolerated dose of pemetrexed was 500 mg/m(2) in cohort A and 350 mg/m(2) in cohort B. Prophylactic antibiotics were required. In cohort A, two dose-limiting toxicities (DLTs) occurred (febrile neutropenia), one each at levels 0 and +1. In cohort B, two DLTs occurred at level +1 (febrile neutropenia; death from perforated duodenal ulcer and sepsis). Grade 3 mucositis was common. No association of gene polymorphisms with toxicity or efficacy was evident. CONCLUSION: The addition of pemetrexed 500 mg/m(2) to cetuximab and radiotherapy is recommended for further study in not previously irradiated patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Adulto , Idoso , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Cetuximab , Terapia Combinada , Relação Dose-Resposta a Droga , Feminino , Glutamatos/administração & dosagem , Glutamatos/efeitos adversos , Guanina/administração & dosagem , Guanina/efeitos adversos , Guanina/análogos & derivados , Neoplasias de Cabeça e Pescoço/genética , Humanos , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/radioterapia , Pemetrexede , Polimorfismo Genético , Carcinoma de Células Escamosas de Cabeça e Pescoço , Timidilato Sintase/genética
5.
Cancer Chemother Pharmacol ; 68(4): 863-70, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21286719

RESUMO

PURPOSE: Germline genetic variations may partly explain the clinical observation that normal tissue tolerance to radiochemotherapy varies by individual. Our objective was to evaluate the association between single-nucleotide polymorphisms (SNPs) in radiation/platinum pathways and serious treatment-related toxicity in subjects with esophageal adenocarcinoma who received cisplatin-based preoperative radiochemotherapy. METHODS: In a multicenter clinical trial (E1201), 81 eligible treatment-naïve subjects with resectable esophageal adenocarcinoma received cisplatin-based chemotherapy concurrent with radiotherapy, with planned subsequent surgical resection. Toxicity endpoints were defined as grade ≥3 radiation-related or myelosuppressive events probably or definitely related to therapy, occurring during or up to 6 weeks following the completion of radiochemotherapy. SNPs were analyzed in 60 subjects in pathways related to nucleotide/base excision- or double stranded break repair, or platinum influx, efflux, or detoxification. RESULTS: Grade ≥3 radiation-related toxicity (mostly dysphagia) and myelosuppression occurred in 18 and 33% of subjects, respectively. The variant alleles of the XRCC2 5' flanking SNP (detected in 28% of subjects) and of GST-Pi Ile-105-Val (detected in 65% of subjects) were each associated with higher odds of serious radiation-related toxicity compared to the major allele homozygote (47% vs. 9%, and 31% vs. 0%, respectively; P = 0.005). No SNP was associated with myelosuppression. CONCLUSIONS: This novel finding in a well-characterized cohort with robust endpoint data supports further investigation of XRCC2 and GST-Pi as potential predictors of radiation toxicity.


Assuntos
Adenocarcinoma/terapia , Cisplatino/administração & dosagem , Proteínas de Ligação a DNA/genética , Neoplasias Esofágicas/terapia , Glutationa S-Transferase pi/genética , Adenocarcinoma/genética , Adulto , Idoso , Antineoplásicos/administração & dosagem , Terapia Combinada , Neoplasias Esofágicas/genética , Feminino , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Lesões por Radiação
6.
Ann Oncol ; 21(11): 2278-2283, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20430907

RESUMO

PURPOSE: RECIST have limitations when applied to potentially curable locally advanced squamous cell carcinoma of the head and neck (SCCHN). [¹8F]fluorodeoxyglucose-positron emission tomography (PET) scan may be useful in assessing treatment response and predicting patient outcome. PATIENTS AND METHODS: We studied patients with previously untreated stages III-IVb SCCHN treated with primary concurrent chemoradiotherapy on five prospective clinical trials. Response was assessed by clinical exam, computed tomography (CT), and PET portions of combined PET-CT scan ∼8 weeks after completion of chemoradiotherapy. RESULTS: Fifty-three patients were analyzed. Complete response (CR) was demonstrated in 42 patients (79%) by clinical exam, 15 (28%) by CT, and 27 (51%) by PET. CR as assessed by PET, but not as assessed by clinical exam or CT using RECIST, correlated significantly with progression-free status (PFS) (P < 0.0001). The 2-year PFS for patients with CR and without CR by PET was 93% and 48%, respectively (P = 0.0002). CONCLUSIONS: A negative PET scan on combined PET-CT after chemoradiotherapy is a powerful predictor of outcome in patients receiving curative chemoradiotherapy for SCCHN. PET-CT is indicated for response evaluation in this setting to improve the accuracy of post-treatment assessment by CT.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/diagnóstico , Neoplasias de Cabeça e Pescoço/terapia , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X , Adulto , Idoso , Carcinoma de Células Escamosas/patologia , Terapia Combinada , Feminino , Fluordesoxiglucose F18 , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Estudos Prospectivos , Compostos Radiofarmacêuticos , Dosagem Radioterapêutica , Taxa de Sobrevida , Resultado do Tratamento , Adulto Jovem
7.
Ann Oncol ; 21(1): 145-51, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19602566

RESUMO

BACKGROUND: Patients treated with chemoradiotherapy (CRT) for head and neck cancers often require feeding tubes (FTs) due to toxicity. We sought to identify factors associated with a prolonged FT requirement. PATIENTS AND METHODS: We retrospectively reviewed 80 patients treated with CRT for head and neck cancers. The pharyngeal constrictors (PCs), supraglottic larynx (SGL), and glottic larynx (GL) were contoured and the mean radiation doses and the volumes of each receiving >40, 50, 60, and 70 Gy (V40, V50, V60, and V70) were determined. RESULTS: A total of 33 of 80 patients required a FT either before or during the course of CRT. Fifteen patients required the FT for > or = 6 months. On univariate analysis, significant factors associated with a prolonged FT requirement were mean PC dose, PC-V60, PC-V70, SGL dose, SGL-V70, and advanced T3-T4 disease. Multivariate analyses found both PC-V70 and T3-T4 disease as significant factors .The proportions of patients requiring a FT > or = 6 months were 8% and 28% for treatment plans with PC-V70 <30% and > or = 30%, respectively. CONCLUSIONS: Increased radiation dose to the PCs is associated with a higher risk of a prolonged FT need. Dose sparing of the PC muscles may reduce this risk.


Assuntos
Nutrição Enteral , Neoplasias de Cabeça e Pescoço/radioterapia , Faringe/efeitos da radiação , Lesões por Radiação/complicações , Radioterapia/efeitos adversos , Adulto , Idoso , Antineoplásicos/efeitos adversos , Terapia Combinada , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso/efeitos da radiação , Estadiamento de Neoplasias , Dosagem Radioterapêutica , Estudos Retrospectivos , Tempo
9.
Nucleic Acids Res ; 25(19): 3868-74, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9380510

RESUMO

p53 is thought to function in the maintenance of genomic stability by modulating transcription and interacting with cellular proteins to influence the cell cycle, DNA repair and apoptosis. p53 mutations occur in >50% of human cancers, and cells which lack wild type p53 accumulate karyotypic abnormalities such as amplifications, deletions, inversions and translocations. We propose that p53 hinders these promiscuous recombinational events by interacting with cellular recombination and repair machinery. We recently reported that p53 can directly bind in vivo to human Rad51 (hRad51) protein and in vitro to its bacterial homologue RecA. We used GST-fusion and his-tagged protein systems to further investigate the physical interaction between p53 and hRad51, homologue of the yeast Rad51 protein that is involved in recombination and DNA double strand repair. The hRad51 binds to wild-type p53 and to a lesser extent, point mutants 135Y, 249S and 273H. This binding is not mediated by a DNA or RNA intermediate. Mapping studies using a panel of p53 deletion mutants indicate that hRad51 could bind to two regions of p53; one between amino acids 94 and 160 and a second between 264 and 315. Addition of anti-p53 antibody PAb421 (epitope 372-381 amino acids) inhibited the interaction with hRad51. In contrast, p53 interacts with the region between aa 125 and 220 of hRad51, which is highly conserved among Rad51 related proteins from bacteria to human. In Escherichia coli ecA protein, this region is required for homo-oligomerization, suggesting that p53 might disrupt the interaction between RecA and Rad51 subunits, thus inhibiting biochemical functions of Rad51 like proteins. These data are consistent with the hypothesis that p53 interaction with hRAD51 may influence DNA recombination and repair and that additional modifications of p53 by mutation and protein binding may affect this interaction.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sítios de Ligação , Sequência Conservada , Reparo do DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Técnicas In Vitro , Modelos Moleculares , Mapeamento de Peptídeos , Mutação Puntual , Ligação Proteica , Conformação Proteica , Rad51 Recombinase , Recombinases Rec A/química , Recombinases Rec A/genética , Recombinases Rec A/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Recombinação Genética , Deleção de Sequência , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética
10.
Cancer Res ; 55(24): 6012-6, 1995 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8521383

RESUMO

The p53 tumor suppressor gene product is a transcriptional transactivator and a potent apoptotic inducer. The fact that many of the DNA tumor virus oncoproteins bind to p53 and affect these p53 functions indicates that this interaction is an important step in oncogenic transformation. We and others have recently demonstrated that the hepatitis B virus oncoprotein, HBx, can form a complex with p53 and inhibit its DNA consensus sequence binding and transcriptional transactivator activity. Using a microinjection technique, we report here that HBx efficiently blocks p53-mediated apoptosis and describe the results of studies exploring two possible mechanisms of HBx action. First, inhibition of apoptosis may be a consequence of the failure of p53, in the presence of HBx, to upregulate genes, such as p21WAF1, Bax, or Fas, that are involved in the apoptotic pathway. Data consistent with this hypothesis include HBx reduction of p53-mediated p21WAF1 expression. Alternatively, HBx could affect p53 binding to the TFIIH transcription-nucleotide excision repair complex as HBx binds to the COOH terminus of p53 and inhibits its binding to XPB or XPD. Binding of p53 to these constituents of the core TFIIH is a process that may be involved in apoptosis. Because the HBx gene is frequently integrated into the genome of hepatocellular carcinoma cells, inhibition of p53-mediated apoptosis by HBx may provide a clonal selective advantage for hepatocytes expressing this integrated viral gene during the early stages of human liver carcinogenesis.


Assuntos
Apoptose , DNA Helicases , Antígenos da Hepatite B/metabolismo , Transativadores/fisiologia , Fatores de Transcrição , Proteína Supressora de Tumor p53/fisiologia , Células Cultivadas , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Hepatite B/patologia , Vírus da Hepatite B/genética , Humanos , Ligação Proteica , Proteínas/metabolismo , Rad51 Recombinase , Proteínas Virais Reguladoras e Acessórias , Proteína Grupo D do Xeroderma Pigmentoso
11.
Endocrinology ; 130(5): 2617-24, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1572285

RESUMO

Recent studies have demonstrated the presence of estrogen receptor (ER) in both normal human osteoblast-like and osteoblast-like osteosarcoma cells. The number of ER in cultured osteoblastic cells is very low (200-500 sites/cell). This has complicated characterization of the biological role of estrogens in bone cells. To study the responsiveness of bone cells to estrogens, we established osteoblast-like cell lines expressing higher ER levels. ROS 17/2.8, an osteoblastic cell line, was stably transfected with the cDNA encoding for the mouse ER. After a selection period, positive clones were isolated and evaluated for the presence of ER by both Northern blot analysis and ligand binding assays. Using these techniques, we detected a significant increase in the level of both ER transcript and binding compared to that in wild-type cells. The levels of expressed ER protein were similar to those reported in normal human osteoblast-like cells in primary culture (approximately 2000 sites/cell). To test whether the exogenously inserted ER was responsive, both wild-type and ER stably transfected cells were transiently transfected with a reporter construct containing an estrogen-responsive element linked to a truncated thymidine kinase promoter and a chloramphenicol acetyltransferase (CAT) reporter gene. Exposure of the cells to increased concentrations of estradiol induced a slight increase in CAT activity in wild-type cells (approximately 1.5-fold) at maximal stimulation; however, it provoked a clear concentration-dependent increase in CAT activity in the ER stably transfected cells, with a maximal stimulation of approximately 10-fold. This event was receptor mediated, since ICI 164,384, an ER antagonist, blocked the enhancement of estradiol-induced CAT activity, and it was specific, since other steroid hormones did not stimulate CAT activity. Finally, we evaluated the ability of ER to modulate an endogenous estrogen-responsive gene by measuring the activity of the enzyme alkaline phosphatase. In addition, diethylstilbestrol, a synthetic estrogen agonist, increased the activity of both the CAT reporter gene and the endogenous alkaline phosphatase enzyme. In summary, we have established osteoblast-like cells expressing high levels of an exogenously inserted ER, which has characteristics similar to those of the endogenous ER in terms of its Kd. Finally, the exogenous ER regulates both exogenously inserted construct (VITERECAT) and endogenous properties of the cells (enzymatic activity and proliferation).


Assuntos
Estradiol/farmacologia , Osteoblastos/fisiologia , Receptores de Estrogênio/fisiologia , Transfecção , Fosfatase Alcalina/metabolismo , Análise de Variância , Animais , Sequência de Bases , Northern Blotting , Linhagem Celular , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Dexametasona/farmacologia , Dietilestilbestrol/farmacologia , Relação Dose-Resposta a Droga , Vetores Genéticos , Globinas/genética , Cinética , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Osteoblastos/efeitos dos fármacos , Osteossarcoma , Progesterona/farmacologia , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/genética , Mapeamento por Restrição
12.
Mol Pharmacol ; 40(5): 806-11, 1991 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1944245

RESUMO

Racemic (Rac) 4'- and 5-deoxyindenestrol A (4'-DIA and 5-DIA), monohydroxyl analogs of the diethylstilbestrol (DES) oxidative metabolite indenestrol A (IA), were synthesized, and their enantiomers were resolved and isolated. Each compound was then tested for estrogen receptor (ER) binding affinity, uterotropic activity, and nuclear ER levels, to further define the stereochemical preference of the ER and to structually evaluate the function of each IA hydroxyl group for binding and biological activity. Competitive binding to cytosolic ER determined the relative binding affinity of racemic mixtures of 4'- and 5-DIA as 1.3 and 3.7, respectively, compared with that of DES, 286. The ER exhibited a binding preference for the S-enantiomer of both compounds, with relative binding affinities of 4'-DIA-R, 0.2; 4'-DIA-S, 1.8; 5-DIA-R, 0.9; and 5-DIA S, 5.6. 4'-DIA-Rac produced 3 times the in vivo stimulation of 5-DIA-Rac in the uterotropic bioassay (with mouse uterine doubling doses of 302.4 and 800 micrograms/kg, respectively). Nuclear ER levels measured 1 hr after in vivo treatment with either 160 micrograms/kg 4'-DIA or 80 micrograms/kg 5-DIA showed a maximum binding level of 2 (4'-DIA) and 1.5 (5-DIA) times saline control, with these doses producing levels nearly equal to that caused by a 10 micrograms/kg dose of IA. Metabolic studies were carried out by treating mice with [3H]4'- and [3H]5-DIA-Rac, to determine the differential binding affinity and biological stimulation of 4'-DIA and 5-DIA. The in vivo metabolism of the [3H]DIA compounds showed formation of [3H]IA-Rac in urine extracts, as analyzed by chiral high performance liquid chromatography. Furthermore, in vitro incubation of unlabeled 4'- and 5-DIA-Rac with mouse liver microsomes showed stereospecific metabolism, with IA-S primarily formed from 4'-DIA-Rac and IA-R from 5-DIA-Rac. Metabolism of 4'-DIA-Rac to the more active IA S-enantiomer and of 5-DIA-Rac to the less active IA R-enantiomer contributes to the different biological activities, because the ER exhibits a chiral preference for these compounds. The higher binding affinity of 5-DIA indicates that the phenyl ring hydroxyl group is required for high affinity binding; however, both hydroxyl groups are needed for subsequent biological activity. These data further suggest that the ER demonstrates stereochemical ligand binding and that IA binds in an orientation relative to 17 beta-estradiol in which the IA phenyl ring corresponds to the estradiol A-ring.


Assuntos
Receptores de Estrogênio/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Feminino , Indenos/metabolismo , Indenos/farmacologia , Camundongos , Camundongos Endogâmicos , Conformação Molecular , Estereoisomerismo , Útero/efeitos dos fármacos
13.
Endocrinology ; 129(4): 2000-10, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1915080

RESUMO

The antiestrogen ICI 164,384 (ICI) binds the estrogen receptor (ER) with approximately 20% the affinity of estradiol, but without the partial agonistic effects caused by tamoxifen. Investigations into the mechanism of ICI action have used ER molecules expressed in vitro to examine the binding of ER to ICI and the capacity of ICI-ER complexes to dimerize and bind to the estrogen response element (ERE). Our objectives were to study the biological effects, cellular distribution, and ERE-binding capacity of native uterine ICI-ER complexes after ip injection of 1 mg/kg ICI into 10-day castrate adult female mice. Synthesis of DNA and progesterone receptor were measured as end points of agonistic activity. ICI failed to stimulate either DNA or progesterone receptor synthesis above control levels, and pretreatment with ICI for 0.5 h reduced the stimulatory effect of estradiol by 75%. Measurement of uterine nuclear ER and cytosolic levels by exchange binding assay indicated a reduction in total ER levels within 0.5 h after ICI treatment, which remained below 20% for 24 h. Cycloheximide treatment did not block the ICI effect. Western blot analysis, immunohistochemistry, and steroid autoradiography confirmed the loss of ER protein. The ICI effect on ER was also demonstrable in vitro in the mouse TM4 estrogen-responsive cell line. ICI dramatically reduced ER levels to 5% of the control value by 4 h. Northern analysis indicated that ICI did not affect ER message levels, suggesting that the observed reduction in ER did not occur at the level of transcription. Gel shift assays indicated a low, but detectable, amount of ICI-ER binding to the vitellogenin A2 (VitA2) ERE. These results suggest that, although the ICI-ER complex binds weakly to DNA, ICI may cause its antagonistic effect by producing a rapid disappearance of the ER from the target tissue, resulting in an insufficient amount of ER to bind the native ligand and elicit agonist responses.


Assuntos
Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Receptores de Estrogênio/metabolismo , Útero/metabolismo , Animais , Western Blotting , Linhagem Celular , Cicloeximida/farmacologia , DNA/biossíntese , Estradiol/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos ICR , Ovariectomia , Alcamidas Poli-Insaturadas , Inibidores de Proteases/farmacologia , Receptores de Progesterona/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA