Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Breast Cancer Res Treat ; 96(2): 131-46, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16261397

RESUMO

We have previously demonstrated that oestrogen receptor alpha (ERalpha) modulates epidermal growth factor receptor (EGFR)/mitogen-activated protein kinase (MAPK) signalling efficiency in a tamoxifen-resistant MCF-7 breast cancer cell line (Tam-R). In the present study we have investigated whether this cross-talk between EGFR/MAPK and ERalpha signalling pathways is bidirectional by examining the effects of EGFR/MAPK activity on ER functionality in the same cell line. Elevated expression levels of phosphorylated serine 118 (S118) ERalpha were observed in the Tam-R compared to the parental wild type MCF-7 cell line (WT-MCF-7) under basal growth conditions. Phosphorylation of ERalpha at S118 was regulated by the EGFR/MAPK pathway in Tam-R cells being increased in response to amphiregulin (AR) and inhibited by the selective EGFR tyrosine kinase inhibitor, gefitinib and the MEK1/2 inhibitor, PD184352. Recruitment of the co-activators p68 RNA helicase and SRC1 to ERalpha, oestrogen response element (ERE) activity and Tam-R cell growth were similarly EGFR/MAPK-regulated. Chromatin immunoprecipitation (ChIP) studies revealed that in Tam-R cells the ERalpha assembled on the AR gene promoter and this was associated with elevated basal expression of AR mRNA. Furthermore, AR mRNA expression was under the regulation of the EGFR/MAPK and ERalpha signalling pathways. Neutralising antibodies to AR inhibited EGFR/ERK1/2 activity, reduced S118 ERalpha phosphorylation and reduced AR mRNA expression in TAM-R cells. These findings suggest that ERalpha function in Tam-R cells is maintained as a consequence of EGFR/MAPK-mediated phosphorylation at serine residue 118 resulting in the generation of a self-propogating autocrine growth-regulatory loop through the ERalpha-mediated production of AR.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Moduladores de Receptor Estrogênico/farmacologia , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Tamoxifeno/farmacologia , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Endocr Relat Cancer ; 13 Suppl 1: S77-88, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17259561

RESUMO

Breast cancer inhibition by antihormones is rarely complete, and our studies using responsive models reveal the remarkable flexibility of breast cancer cells in recruiting alternative signalling to limit maximal anti-tumour effects of oestrogen receptor alpha (ER) blockade. The recruited mechanism involves antihormone-induced expression of oestrogen-repressed signalling genes. For example, epidermal growth factor receptor gene (EGFR) is induced by antioestrogens and maintains residual kinase and ER phosphorylation, cell survival genes, and thereby allows incomplete antihormone response and emergence of resistance. Microarrays are revealing the breadth of antihormone-induced genes that may attenuate growth inhibition, including NFkappaB, Bag1, 14-3-3zeta and tyrosine kinases, such as HER2 and Lyn. Three concepts are emerging: first, some genes are induced exclusively by antioestrogens, while others extend to oestrogen deprivation; secondly, some are transiently induced, while others persist into resistance; finally, some confer additional adverse features when tumour cells are in an appropriate context. Among the latter is CD59 whose antioestrogen induction may permit evasion of immune surveillance in vivo. Also, induction of pro-invasive genes (including NFkappaB, RhoE and delta-catenin) may underlie our findings that antioestrogens can markedly stimulate migratory behaviour when tumour intercellular contacts are compromised. Based on our promising studies selectively inhibiting EGFR (gefitinib), NFkappaB (parthenolide) or CD59 (neutralising antibody) together with antioestrogens, we propose that co-targeting strategies could markedly improve anti-tumour activity (notably enhancing cell kill) during the antihormone-responsive phase. Furthermore, subverting those induced signalling genes that are retained into resistance (e.g. EGFR, NFkappaB, HER2) may prove valuable in this state. Alongside future deciphering and targeting of genes underlying antioestrogen-promoted invasiveness, embracing of intelligent combination strategies could significantly extend patient survival.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Carcinoma/tratamento farmacológico , Carcinoma/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Estrogênios/farmacologia , Animais , Neoplasias da Mama/genética , Carcinoma/genética , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
3.
Endocrinology ; 144(11): 5105-17, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12960029

RESUMO

Although many estrogen receptor-positive breast cancers initially respond to antihormones, responses are commonly incomplete with resistance ultimately emerging. Delineation of signaling mechanisms underlying these phenomena would allow development of therapies to improve antihormone response and compromise resistance. This in vitro investigation in MCF-7 breast cancer cells examines whether epidermal growth factor receptor (EGFR) signaling limits antiproliferative and proapoptotic activity of antihormones and ultimately supports development of resistance. It addresses whether the anti-EGFR agent gefitinib (ZD1839/Iressa; TKI: 1 mum) combined with the antihormones 4-hydroxytamoxifen (TAM: 0.1 mum) or fulvestrant (Faslodex; 0.1 mum) enhances growth inhibition and prevents resistance. TAM significantly suppressed MCF-7 growth over wk 2-5, reducing proliferation detected by immunocytochemistry and fluorescence-activated cell sorter cell cycle analysis. A modest apoptotic increase was observed by fluorescence-activated cell sorter and fluorescence microscopy, with incomplete bcl-2 suppression. EGFR induction occurred during TAM response, as measured by immunocytochemistry and Western blotting, with EGFR-positive, highly proliferative resistant growth subsequently emerging. Although TKI alone was ineffective on growth, TAM plus TKI cotreatment exhibited superior antigrowth activity vs. TAM, with no viable cells by wk 12. Cotreatment was more effective in inhibiting proliferation, promoting apoptosis, and eliminating bcl-2. Cotreatment blocked EGFR induction, markedly depleted ERK1/2 MAPK and protein kinase B phosphorylation, and prevented emergence of EGFR-positive resistance. Faslodex plus TKI cotreatment was also a superior antitumor strategy. Thus, increased EGFR evolves during treatment with antihormones, limiting their efficacy and promoting resistance. Gefitinib addition to antihormonal therapy could prove more effective in treating estrogen receptor-positive breast cancer and may combat development of resistance.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Fator de Crescimento Epidérmico/antagonistas & inibidores , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Proteínas Serina-Treonina Quinases , Quinazolinas/farmacologia , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Combinação de Medicamentos , Resistência a Medicamentos/efeitos dos fármacos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Feminino , Fulvestranto , Gefitinibe , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos
4.
Ann N Y Acad Sci ; 963: 104-15, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12095935

RESUMO

An increasing body of evidence demonstrates that growth factor networks are highly interactive with estrogen receptor signaling in the control of breast cancer growth. As such, tumor responses to antihormones are likely to be a composite of the estrogen receptor and growth factor inhibitory activity of these agents. The modulation of growth factor networks during endocrine response is examined, and in vitro and clinical evidence is presented that epidermal growth factor receptor signaling, maintained in either an estrogen receptor-dependent or a receptor-independent manner, is critical to antihormone-resistant breast cancer cell growth. The considerable potential of the epidermal growth factor receptor-selective tyrosine kinase inhibitor Iressa (ZD 1839) to efficiently treat, and perhaps even prevent, endocrine-resistant breast cancer is highlighted.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Receptores ErbB/metabolismo , Estradiol/análogos & derivados , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Divisão Celular/efeitos dos fármacos , Glândulas Endócrinas , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/farmacologia , Feminino , Fulvestranto , Gefitinibe , Humanos , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacologia , Receptor ErbB-2/fisiologia , Transdução de Sinais/fisiologia , Tamoxifeno/farmacologia , Trastuzumab , Células Tumorais Cultivadas
5.
Endocr Relat Cancer ; 8(3): 175-82, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11566608

RESUMO

There is an increasing body of evidence demonstrating that growth factor networks are highly interactive with oestrogen receptor (ER) signalling in the control of breast cancer growth. As such, tumour responses to anti- hormones are likely to be a composite of the ER and growth factor inhibitory activity of these agents. The current article examines the modulation of growth factor networks during endocrine response, and presents in vitro and clinical evidence that epidermal growth factor receptor signalling, maintained in either an ER-dependent or -independent manner, is critical to anti- hormonal-resistant breast cancer cell growth. The considerable potential of the epidermal growth factor receptor-selective tyrosine kinase inhibitor, ZD 1839 (Iressa; AstraZeneca) to efficiently treat, and perhaps even prevent, endocrine-resistant breast cancer is highlighted.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Antineoplásicos/uso terapêutico , Antagonistas de Estrogênios/uso terapêutico , Feminino , Humanos , Receptor ErbB-2/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
6.
Endocrinology ; 142(7): 2776-88, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11415996

RESUMO

This paper describes the establishment of an antiestrogen-resistant MCF7 breast cancer cell subline (FASMCF) by continuous culture of the estrogen-responsive parental line in steroid-depleted, ICI 182,780 (Faslodex; 10(-7) M)-supplemented medium. After a 3-month period of growth suppression, cells began to proliferate in ICI 182,780 at rates similar to those of untreated wild-type cells. Immunocytochemistry showed these cells to have reduced estrogen receptor and an absence of progesterone receptor proteins. RT-PCR and transient transfection studies with estrogen response element-reporter constructs confirmed that ICI 182,780-suppressed estrogen response element-mediated signaling. FASMCF cells show increased dependence upon epidermal growth factor receptor (EgfR)/mitogen-activated protein kinase (MAPK)-mediated signaling. Thus, EgfR protein and messenger RNA, growth responses to transforming growth factor-alpha, and extracellular signal-regulated kinase 1/2 MAPK activation levels are all increased. Unlike wild-type cells, FASMCF cells are highly sensitive to growth inhibition by an EgfR-specific tyrosine-kinase inhibitor (TKI), ZD1839 (Iressa), and an inhibitor of the activation of MEK1 (MAPKK), PD098059. Short-term ( approximately 3 weeks) withdrawal of cells from antiestrogen had no effect on growth or phenotype, whereas longer withdrawal (>10 weeks) appeared to partially reverse the cellular phenotype with increasing estrogen receptor and decreasing EgfR levels. In subsequent studies FASMCF cells were maintained in TKI, where their growth was again suppressed and secondary TKI resistance failed to develop within the 3-month period in which initial ICI 182,780 resistance arose. Furthermore, wild-type cells similarly maintained in combination ICI 182,780 and TKI treatment conditions remained growth arrested (>6 months), with notable cell loss through both reduced rates of cellular proliferation and increased cell death.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/fisiopatologia , Receptores ErbB/fisiologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/farmacologia , Transdução de Sinais , Neoplasias da Mama/patologia , Técnicas Citológicas , Resistência a Medicamentos , Inibidores Enzimáticos/farmacologia , Feminino , Fulvestranto , Gefitinibe , Humanos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos
7.
Eur J Cancer ; 36 Suppl 4: S34-5, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11056309

RESUMO

Selective oestrogen receptor downregulators (SERDs) are a class of highly effective steroidal antitumour agents that reduce cellular levels of the oestrogen receptor (ER). In this study, we compared the efficacy by which three novel molecular approaches: (1) antisense oligonucleotides; (2) antisense RNA; and (3) dominant negative mutants are able to act as SERDs. Using transient and, where appropriate, stable gene transfection experiments we found that constitutive overexpression of ER antisense RNA and a hormone-binding domain compromised dominant-negative ER mutant (DNER-1), were most effective at downregulating ER expression and/or activity in vitro.


Assuntos
Antineoplásicos/uso terapêutico , Estradiol/análogos & derivados , Estradiol/uso terapêutico , Antagonistas de Estrogênios/uso terapêutico , Receptores de Estrogênio/genética , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Northern Blotting , Divisão Celular , Células Cultivadas , Fulvestranto , Genes Reporter , Humanos , Oligonucleotídeos Antissenso/metabolismo
8.
Endocr Relat Cancer ; 6(3): 373-87, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10516852

RESUMO

Multiple lines of evidence implicate steroid hormone and growth factor cross-talk as a modulator of endocrine response in breast cancer and that aberrations in growth factor signaling pathways are a common element in the endocrine resistant phenotype. Delineation of these relationships is thus an important diagnostic goal in cancer research, while the targeting of aberrant growth factor signaling holds the promise of improving therapeutic response rates.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/fisiopatologia , Estrogênios/fisiologia , Substâncias de Crescimento/fisiologia , Receptor Cross-Talk/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Neoplasias da Mama/patologia , Feminino , Humanos , Receptores de Estrogênio/fisiologia , Transdução de Sinais/fisiologia
9.
J Pathol ; 188(2): 126-32, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10398154

RESUMO

An immunocytochemical assay for the p53-regulated protein product of the WAF1/Cip1 gene, p21(WAF1) (p21), was developed and applied to archival primary breast tumour material from 91 patients whose subsequent recurrent disease was treated with assessable courses of endocrine therapy. Nuclear localization of p21 protein was observed in 76 (82.4 per cent) cases. Status cut-offs were established and 29 (31.9 per cent) were deemed negative, 39 (42.9 per cent) weakly positive, and 23 (25.3 per cent) strongly positive. p21 status was inversely correlated with p53 protein (p=0.047) but did not relate to oestrogen receptor (ER) status, response to endocrine therapy, or time to further disease progression (TTP). Highly p21-positive patients had a significantly improved overall survival time (p=0. 020). Co-assessment of p21 and p53 subgroups revealed p21+/p53- patients to have good survival characteristics, whilst p21-/p53+ patients did poorly (p=0.008). The p21-/p53- patients overall did intermediately well, but Ki67-defined cellular proliferation analysis of these revealed two subclasses: those with high proliferation and poor survival times resembling the p21-/p53+ phenotype, and those with less proliferative tumours with good survival, similar to the p21+/p53- group. The significance of these results is discussed in the light of recent research concerning the role of p21 and p53 in breast cancer aetiology.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/química , Ciclinas/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Núcleo Celular/química , Inibidor de Quinase Dependente de Ciclina p21 , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/análise , Pessoa de Meia-Idade , Receptores de Estrogênio/análise , Análise de Sobrevida , Proteína Supressora de Tumor p53/análise
10.
Oncogene ; 17(8): 1053-9, 1998 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-9747885

RESUMO

The EMS1 and CCND1 genes at chromosome 11q13 are amplified in about 15% of primary breast cancers but appear to confer different phenotypes in ER positive and ER negative tumours. Since there are no published data on EMS1 expression in large series of breast cancers we examined the relationship of EMS1 expression with EMS1 gene copy number and expression of mRNAs for cyclin D1 and ER. In a subset of 129 patients, where matched tumour RNA and DNA was available, EMS1 mRNA overexpression was associated predominantly with gene amplification (P = 0.0061), whereas cyclin D1 mRNA overexpression was not (P = 0.3142). In a more extensive series of 351 breast cancers, there was no correlation between cyclin D1 and EMS1 expression in the EMS1 and cyclin D1 overexpressors (P = 0.3503). Although an association between EMS1 mRNA expression and ER positivity was evident (P = 0.0232), when the samples were divided into quartiles of EMS1 or cyclin D1 mRNA expression, the increase in the proportion of ER positive tumours in the ascending EMS1 mRNA quartiles was not statistically significant (P = 0.0951). In marked contrast there was a significant stepwise increase in ER positivity in ascending quartiles of cyclin D1 mRNA (P = 0.030). A potential explanation for this difference was provided by the observation that in ER positive breast cancer cells oestradiol treatment resulted in increased cyclin D1 gene expression but was without effect on EMS1. The relationship between EMS1 expression and clinical outcome was examined in a subset of 234 patients with median follow-up of 74 months. High EMS1 expression was associated with age > 50 years (P = 0.0001), postmenopausal status (P = 0.0008), lymph node negativity (P = 0.019) and an apparent trend for worse prognosis in the ER negative subgroup. These data demonstrate that overexpression of EMS1 mRNA is largely due to EMS1 gene amplification, is independent of cyclin D1 and ER expression and, in contrast to cyclin D1, is not regulated by oestrogen. Independent overexpression of these genes may confer different phenotypes and disease outcomes in breast cancer as has been inferred from recent studies of EMS1 and CCND1 gene amplification.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Ciclina D1/biossíntese , Regulação Neoplásica da Expressão Gênica , Proteínas dos Microfilamentos , Proteínas de Neoplasias/genética , Receptores de Estrogênio/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/mortalidade , Cromossomos Humanos Par 11/genética , Cortactina , Ciclina D1/genética , Humanos , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese
11.
Biochem Pharmacol ; 56(3): 335-44, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9744571

RESUMO

The 2-nitroimidazoles have been used clinically to radiosensitize resistant hypoxic cells, but a dose-limiting peripheral neuropathy has restricted their therapeutic effectiveness. A model compound, 1-methyl-2-nitroimidazole (INO2), was used to investigate the possible role of oxidative stress in this normal tissue toxicity. Chinese hamster ovary (CHO) cells were 10-15 times more resistant to 20 mM INO2 under aerobic than hypoxic conditions. In comparison, a pair of transformed rat embryo fibroblasts (ER17-1wtp53 and ER12L5mtpP53), differing in their p53 genotype, were approximately 3- to 4-fold more sensitive than Chinese hamster ovary cells to INO2 under aerobic conditions, but had the same sensitivity as Chinese hamster ovary cells under hypoxic conditions. These results are consistent with an earlier hypothesis that the mechanism of aerobic toxicity is different from that of hypoxic toxicity (nitroreduction) and show that neither toxicity is dependent on cellular p53 status. There was an increase in the production of reactive oxygen intermediates and a decrease in the antioxidant glutathione following aerobic exposure to INO2, which correlated with cell survival in all three cell lines. No evidence of reductive adducts of the 2-nitroimidazole 2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl)acetam ide (EF5) was found by immunofluorescent techniques in aerobic cells. Differing activities of the antioxidant enzymes superoxide dismutase and catalase could be correlated with INO2 aerobic cytotoxicity. DNA strand breaks, as measured by the comet assay, paralleled the appearance of INO2 aerobic cytotoxicity in all three cell lines. Taken together, these results strongly support the conclusion that the aerobic toxicity of IN02 is due to active oxygen species created by futile redox cycling of the parent compound.


Assuntos
Nitroimidazóis/toxicidade , Estresse Oxidativo , Aerobiose , Animais , Células CHO , Catalase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Dano ao DNA , Glutationa/análise , Ratos , Espécies Reativas de Oxigênio , Superóxido Dismutase/metabolismo
12.
N Engl J Med ; 339(7): 417-23, 1998 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-9700174

RESUMO

BACKGROUND: Deferiprone is an orally active iron-chelating agent that is being evaluated as a treatment for iron overload in thalassemia major. Studies in an animal model showed that prolonged treatment is associated with a decline in the effectiveness of deferiprone and exacerbation of hepatic fibrosis. METHODS: Hepatic iron stores were determined yearly by chemical analysis of liver-biopsy specimens, magnetic susceptometry, or both. Three hepatopathologists who were unaware of the patients' clinical status, the time at which the specimens were obtained, and the iron content of the specimens examined 72 biopsy specimens from 19 patients treated with deferiprone for more than one year. For comparison, 48 liver-biopsy specimens obtained from 20 patients treated with parenteral deferoxamine for more than one year were similarly reviewed. RESULTS: Of the 19 patients treated with deferiprone, 18 had received the drug continuously for a mean (+/-SE) of 4.6+/-0.3 years. At the final analysis, 7 of the 18 had hepatic iron concentrations of at least 80 micromol per gram of liver, wet weight (the value above which there is an increased risk of cardiac disease and early death in patients with thalassemia major). Of 19 patients in whom multiple biopsies were performed over a period of more than one year, 14 could be evaluated for progression of hepatic fibrosis; of the 20 deferoxamine-treated patients, 12 could be evaluated for progression. Five deferiprone-treated patients had progression of fibrosis, as compared with none of those given deferoxamine (P=0.04). By the life-table method, we estimated that the median time to progression of fibrosis was 3.2 years in deferiprone-treated patients. After adjustment for the initial hepatic iron concentration, the estimated odds of progression of fibrosis increased by a factor of 5.8 (95 percent confidence interval, 1.1 to 29.6) with each additional year of deferiprone treatment. CONCLUSIONS: Deferiprone does not adequately control body iron burden in patients with thalassemia and may worsen hepatic fibrosis.


Assuntos
Quelantes de Ferro/uso terapêutico , Sobrecarga de Ferro/tratamento farmacológico , Piridonas/uso terapêutico , Talassemia beta/tratamento farmacológico , Adolescente , Adulto , Biópsia , Criança , Deferiprona , Desferroxamina/uso terapêutico , Progressão da Doença , Feminino , Humanos , Ferro/análise , Quelantes de Ferro/efeitos adversos , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/patologia , Fígado/química , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/etiologia , Cirrose Hepática/patologia , Modelos Logísticos , Masculino , Piridonas/efeitos adversos , Falha de Tratamento , Talassemia beta/complicações , Talassemia beta/patologia
13.
Br J Cancer ; 77(10): 1653-6, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9635843

RESUMO

Northern hybridization analyses of the oestrogen-inducible mRNAs pLIV1 and pS2 were compared with oestrogen receptor (ER) immunocytochemistry assessments in 40 untreated primary or early recurrent breast tumours. Significant associations were observed between pLIV1/ER (P < 0.03), pS2/ER (P < 0.001) and pLIV1/pS2 (P < 0.04) status. After disease recurrence, patients were treated with assessable courses of endocrine therapies. Positive pLIV1, pS2 and ER statuses in primary disease were consequently found to be predictive of endocrine responsiveness in the secondary lesions (P < 0.03, P < 0.02, P < 0.005 respectively). However, despite these associations, a number of pLIV1- and/or pS2-positive tumours failed to respond to therapy.


Assuntos
Neoplasias da Mama/genética , Estrogênios/genética , Regulação da Expressão Gênica , Neoplasias Hormônio-Dependentes/genética , Proteínas/genética , RNA Mensageiro/genética , Adulto , Idoso , Estrogênios/uso terapêutico , Feminino , Humanos , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Receptores de Estrogênio/análise , Fator Trefoil-1 , Proteínas Supressoras de Tumor
14.
Am J Trop Med Hyg ; 58(3): 358-64, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9546419

RESUMO

While the parenteral iron-chelating agent desferrioxamine B has anti-malarial activity in humans, the usefulness of an orally active chelator for this indication has not been investigated previously in vivo. We conducted a prospective, double-blind, placebo-controlled, cross-over trial of deferiprone (L1; CP20; 1,2-dimethyl-3-hydroxypyridin-4-one) in 25 adult Zambians with asymptomatic Plasmodium falciparum parasitemia. Deferiprone was administered daily for three or four days in divided doses of 75 or 100 mg/kg of body weight, dosages that are effective for treating iron overload. No reduction in asexual intra-erythrocytic parasites was observed during or after deferiprone treatment. The mean peak plasma concentration of deferiprone (108.9 +/- 24.9 micromol/L) achieved was within the range demonstrated to inhibit the growth of P. falciparum in vitro, but the systemic exposure as determined by the 24-hr plasma concentration-time curve would not be predicted inhibit growth in vivo. No evidence of deferiprone-associated hematological toxicity was noted in this short-term study of these subjects, all of whom had clinical evidence of normal body iron stores. Because of the risk of neutropenia and other adverse effects with higher doses or prolonged use of the chelator, additional trials of deferiprone as a sole anti-malarial agent would not seem to be justified. In contrast, further efforts are needed to develop other orally active iron-chelating agents specifically for their anti-malarial action.


Assuntos
Quelantes de Ferro/uso terapêutico , Malária Falciparum/tratamento farmacológico , Parasitemia/tratamento farmacológico , Piridonas/uso terapêutico , Administração Oral , Adulto , Área Sob a Curva , Estudos Cross-Over , Deferiprona , Relação Dose-Resposta a Droga , Método Duplo-Cego , Humanos , Quelantes de Ferro/administração & dosagem , Quelantes de Ferro/farmacocinética , Malária Falciparum/metabolismo , Masculino , Parasitemia/metabolismo , Estudos Prospectivos , Piridonas/administração & dosagem , Piridonas/farmacocinética
15.
Br J Cancer ; 76(2): 180-8, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9231917

RESUMO

The time course and characteristics of the selective hypoxic cytotoxicity of the 2-nitroimidazole model compound 1-methyl-2-nitroimidazole (INO2) were analysed during prolonged time periods (up to 5 days post treatment). When control populations were seeded at the same cell density as drug-treated cells, they entered confluency at day 3 and underwent apoptosis at day 5, which appeared to be mediated by an autocrine mechanism. In subsequent studies of drug-treated cells, the seeding density of treated cells was adjusted to avoid this cell confluency effect. Treatment with a low INO2 concentration (2.5 mM) resulted in apoptotic DNA fragmentation (ladders), which was observed 4-5 days after an acute 6-h hypoxic drug exposure. In contrast, at a high INO2 concentration (40 mM) for 2 h, which was equitoxic to the low concentration, no characteristic DNA ladders were observed. Fluorescence microscopy revealed apoptotic bodies and pyknotic nuclei 5 days following hypoxic 2.5 mM INO2 exposure, whereas 40 mM INO2 hypoxic treatment produced cellular ghosts devoid of DNA 5 days after exposure, consistent with the DNA ladder results. However, characteristic apoptotic morphology was previously observed immediately after the acute hypoxic exposure of 40 mM INO2. Cell cycle analysis and DNA fragmentation as measured by the TdT assay suggested that dose-dependent differences in the apoptotic response occur post exposure after an equitoxic acute hypoxic exposure to either the low or the high INO2 concentration. This dose-dependent differential in response may be attributed to the degree of initial DNA damage as measured by the comet assay.


Assuntos
Apoptose/efeitos dos fármacos , Células CHO/efeitos dos fármacos , Nitroimidazóis/toxicidade , Animais , Células CHO/patologia , Células CHO/ultraestrutura , Ciclo Celular , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Cricetinae , Dano ao DNA/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida
16.
Clin Cancer Res ; 3(11): 2165-72, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9815611

RESUMO

We describe the development and use of a sensitive reverse transcription-PCR (RT-PCR) procedure to detect novel estrogen-regulated gene expression in small clinical breast cancer samples, in which such study would be extremely difficult by any other molecular or immunocytochemical means. Assay optimization for pLIV1, estrogen receptors (ERs), progesterone receptors, and pS2 gene products was carried out on 50 primary breast cancers for which comparative Northern analysis and immunocytochemical data were available. Using 27 amplification cycles and a 0.5 microM primer concentration, varying expressions of the gene products were recorded simultaneously with a constant densitometric signal for a coamplified endogenous control gene (alpha-actin). Good concordances were subsequently observed between pLIV1 status generated by RT-PCR and both Northern analysis (P = 0.002) and ER status by immunocytochemistry (P = 0.0244). Agreement was also noted between ER (P = 0.002), progesterone receptor (P = 0.0005), and pS2 (P = 0. 0023) RT-PCR and immunocytochemical methodologies. The RT-PCR assays were then applied to 10 needle core trucut biopsies in which similar relationships were obtained. Our results justify the future use of this RT-PCR methodology to examine new estrogen-regulated genes in small breast cancer samples, and it is envisaged that this technology will prove invaluable in many future breast cancer studies.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Proteínas/análise , Receptores de Estrogênio/análise , Receptores de Progesterona/análise , Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Estrogênios/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Proteínas/genética , RNA Mensageiro/análise , Receptores de Estrogênio/genética , Receptores de Progesterona/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transcrição Gênica , Fator Trefoil-1 , Proteínas Supressoras de Tumor
17.
Clin Cancer Res ; 2(6): 923-8, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9816251

RESUMO

The CCND1 gene, encoding the cell cycle regulatory protein cyclin D1, maps to chromosome 11q13, a locus that is amplified in about 13% of breast cancers. Because several studies have indicated a relationship between 11q13 amplification and markers of phenotype including estrogen receptor (ER) status, we tested the relationship between CCND1 and ER gene expression in 364 primary breast cancers using Northern blot analysis. Seventy-three % of samples were positive for ER mRNA, and cyclin D1 mRNA levels in the ER-positive group were significantly higher than those in the ER-negative group (P = 0.0001). When the samples were divided into quartiles of cyclin D1 expression, 58% of samples were ER positive in the lowest quartile and 87% in the highest quartile. The tumors expressing the highest levels of cyclin D1 (7%) were all ER positive. Furthermore, ER mRNA levels in the half with lower cyclin D1 mRNA were significantly less than in the half with higher cyclin D1 levels (P = 0.0001). Using simple regression analysis, there was a significant positive correlation between cyclin D1 and ER mRNA levels in the total population (P = 0.0001). This study demonstrates that cyclin D1 mRNA and ER mRNA are positively correlated in primary breast cancer, but the functional relationship between these genes remains to be elucidated.


Assuntos
Neoplasias da Mama/metabolismo , Ciclina D1/genética , RNA Mensageiro/análise , Receptores de Estrogênio/genética , Feminino , Amplificação de Genes , Humanos
19.
Eur J Cancer ; 32A(3): 413-6, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8814683

RESUMO

Expression of oestrogen receptor (ER), epidermal growth factor receptor (EGFR) and transforming growth factor-alpha (TGF alpha) proteins was assessed by immunocytochemistry on primary breast cancer specimens obtained before and following short-term (7-day) presurgical exposure to pure anti-oestrogen (7 alpha- [9- (4,4,5,5,5-pentafluoropentylsulphinyl) nonyl] estra-1,3,5, (10)-triene-3,17 beta-diol, ICI 182780) treatment and compared with no-treatment controls. Paired needle-core and mastectomy samples were obtained from 21 patients. Effects of ICI 182780 (10(-7)M) on MCF7 breast cancer cell ER, EGFR and TGF alpha expression were also examined over 14 days. ER protein was significantly suppressed by ICI 182780 in vivo (P = 0.009) and comparative analysis of short term ICI 182780 effects in vitro, using ER-positive MCF7 cells, gave largely equivalent results. EGFR and TGF alpha protein levels were unaltered by treatment. ICI 182780 suppresses ER without a concomitant rise in either EGFR or TGF alpha.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Neoplasias da Mama/metabolismo , Receptores ErbB/metabolismo , Estradiol/farmacologia , Feminino , Secções Congeladas , Fulvestranto , Humanos , Imuno-Histoquímica , Pós-Menopausa , Receptores de Estrogênio/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
20.
Breast Cancer Res Treat ; 41(1): 31-41, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8932874

RESUMO

Effects of the pure antiestrogen ICI182780 and tamoxifen on ER-protein, ER-mRNA, and estrogen-regulated mRNA expression were analysed using matched pretreatment core-cut biopsies and post-treatment mastectomy samples from 43 ER positive human breast cancers. Sixteen controls received either no preoperative treatment (n = 9) (7 days) or placebo (n = 7) (median 21 days) prior to primary surgery. Nineteen patients received ICI182780 6 mg/day (n = 10) or 18 mg/day (n = 9) for 7 days. Eight patients were given preoperative tamoxifen (4 x 40 mg-day 1, 20 mg/day thereafter, median 21 days). ER-protein expression was assessed on pre and post treatment samples by immunocytochemistry. ER, pS2, pLIV1, and actin-mRNA expression was determined by northern analysis on post-treatment samples only. ER-mRNA levels were similar to controls following ICI182780 or tamoxifen treatment. However ER-protein levels were significantly suppressed by ICI182780, particularly at the higher dosage (p = 0.0013). Tamoxifen had no significant effect on ER-protein levels. The ER-mRNA and ER-protein contents of control tumors were linearly related (Spearman r = 0.719, p = 0.006). A similar relationship between pretreatment protein and post ICI182780 treatment mRNA levels was observed (r = 0.652, p = 0.005). However, comparison of post ICI182780 treatment protein and mRNA results shows a loss of linearity through a reduction in protein without concurrent loss of mRNA (r = 0.28, p = 0.257). pS2 mRNA hybridization was lower in ICI182780 treated samples than controls (Mann-Whitney p = 0.035) but was unaffected by tamoxifen. pLIV1 mRNA hybridization was uninfluenced by either treatment. Short term exposure of breast tumors to ICI182780 appears to produce a greater inhibition of estrogen-induced transcriptional events than tamoxifen. These effects appear to occur without a concurrent reduction in ER mRNA levels.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Antagonistas de Estrogênios/uso terapêutico , Estrogênios/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/genética , Proteínas , Receptores de Estrogênio/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Estradiol/análogos & derivados , Estradiol/uso terapêutico , Estrogênios/biossíntese , Fulvestranto , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Proteínas de Neoplasias/genética , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , RNA Mensageiro/análise , Tamoxifeno/uso terapêutico , Fator Trefoil-1 , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA