Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pathol ; 217(1): 32-41, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18825690

RESUMEN

The AP-2gamma transcription factor encoded by the TFAP2C gene is a member of a family of homologous DNA binding proteins that play essential roles during vertebrate embryogenesis but show a restricted pattern of expression in the adult. Elevated expression of the AP-2alpha and AP-2gamma family members has been associated with a number of neoplasms, particularly breast cancer. Here we present an exploratory immunohistochemical study of an archival primary breast tumour series (n = 75) with parallel clinicopathological data using a new, well-characterized antibody to AP-2gamma. Heterogeneous, exclusively nuclear expression of AP-2gamma was found in the epithelial and myoepithelial compartments of normal breast and within tumour epithelial cells. In the breast cancer series, the most notable association was a correlation between elevated levels of AP-2gamma and shortened patient survival (p = 0.0009*). This relationship was also conserved in ER-positive and ErbB2-negative patients; sub-groups generally considered to have a relatively good prognosis. When patient data for survival and duration of treatment response on anti-hormone therapy were examined by multivariate analysis, AP-2gamma was revealed in this study to be an independent predictor of outcome for both survival (p = 0.005) and response to anti-hormone therapy (p = 0.046). Studies using in vitro models confirmed that while tamoxifen response is associated with lower levels of AP-2gamma, acquisition of resistance to this and other anti-hormone measures (eg faslodex or oestrogen deprivation) is associated with high levels of nuclear AP-2gamma. Together these data suggest that elevated tumour AP-2gamma expression can contribute to the failure of cells to growth arrest following anti-hormone treatment and lead to sustained growth and poorer patient outcome.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Factor de Transcripción AP-2/metabolismo , Adulto , Anciano , Anticuerpos Monoclonales/inmunología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Femenino , Humanos , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Pronóstico , Análisis de Supervivencia , Tamoxifeno/uso terapéutico , Factor de Transcripción AP-2/inmunología , Resultado del Tratamiento , Células Tumorales Cultivadas
2.
J Clin Pathol ; 60(11): 1216-21, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17965220

RESUMEN

BACKGROUND: Although in vitro breast cancer models have demonstrated a role for protein kinase C (PKC) alpha and delta isoforms in endocrine insensitivity and resistance respectively, there is currently little clinical evidence to support these observations. AIMS: To define the pattern of PKC alpha and delta expression using breast cancer cell lines, with and without endocrine resistance, and also breast cancer samples, where expression can be correlated with clinicopathological and endocrine therapy outcome data. METHODS: PKC isoform expression was examined in tamoxifen responsive, oestrogen receptor positive (ER(+)), ER(+) acquired tamoxifen resistant (TAM-R) and oestrogen receptor negative (ER(-)) cell lines by western blotting and immunocytochemical analysis. PKC isoform expression was then examined by immunohistochemistry in archival breast cancer specimens from primary breast cancer patients with known clinical outcome in relation to endocrine response and survival on therapy. RESULTS: ER(+) breast cancer cell lines expressed considerable PKC-delta but barely detectable levels of PKC-alpha, whereas ER(-) cell lines expressed PKC-alpha but little PKC-delta. ER(+) acquired TAM-R cell lines expressed substantial levels of both PKC-alpha and delta. In clinical samples, high PKC-delta expression correlated to endocrine responsiveness whereas PKC-alpha expression correlated to ER negativity. PKC-delta was an independent predictor of duration of response to therapy. Patients showing a PKC-delta(+)/PKC-alpha(-) phenotype had a six times longer endocrine response than patients with the PKC-delta(+)/ PKC-alpha(+) phenotype (equating to tamoxifen resistance in vitro). CONCLUSIONS: Levels of PKC-alpha and delta expression appear to be indicative of response to anti-oestrogen therapy and could be useful in predicting a patient's suitability for endocrine therapy.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-delta/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Femenino , Humanos , Metástasis Linfática , Receptores de Estrógenos/metabolismo , Análisis de Supervivencia , Tamoxifeno/uso terapéutico , Resultado del Tratamiento , Células Tumorales Cultivadas
3.
Rev Endocr Metab Disord ; 8(3): 241-53, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17486454

RESUMEN

Growth factors provide powerful mitogenic and survival signals to breast cancer cells and it is therefore not surprising that they are able to subvert inhibitory responses to anti-hormonal drugs. In this review we discuss several mechanisms by which this may be achieved and expand our observations to encompass recently emerging anti-growth factor treatments. The information presented is underpinned by inhibitor studies that show the targeting of such mechanisms in advance of anti-hormone or anti-growth factor resistance development is able to substantially delay this event, thus pointing the way forward to intelligent combination therapies relevant to the future management of breast cancer.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Transducción de Señal/fisiología , Receptores ErbB/fisiología , Femenino , Humanos , Receptores de Estrógenos/fisiología
4.
Eur J Cancer ; 43(1): 64-70, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17064888

RESUMEN

Fulvestrant (Faslodex) reduces markers of hormone sensitivity and proliferation in postmenopausal women. This Phase II double-blind, randomised, multicentre study compared the effects of a single 250mg intramuscular dose of fulvestrant and placebo 14-21 days prior to surgery of curative intent on the oestrogen receptor (ER), progesterone receptor and Ki67 levels in 66 premenopausal women with ER-positive primary breast cancer. There were no statistically significant differences between fulvestrant and placebo with respect to any of the three markers analysed. The most common adverse events in both groups were nausea, headache and pyrexia. Fulvestrant 250mg had no effects on markers of hormone-sensitivity and proliferation in premenopausal women with primary breast cancer when measured at 14-21 days after injection. These findings suggest that a higher fulvestrant dose may be required in this patient population. Further clinical trials are necessary to evaluate the efficacy of fulvestrant in premenopausal women.


Asunto(s)
Antineoplásicos Hormonales/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Estradiol/análogos & derivados , Receptores de Estrógenos/metabolismo , Adulto , Antineoplásicos Hormonales/efectos adversos , Antineoplásicos Hormonales/sangre , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Estradiol/administración & dosificación , Estradiol/efectos adversos , Estradiol/sangre , Femenino , Fase Folicular , Fulvestrant , Humanos , Antígeno Ki-67/metabolismo , Fase Luteínica , Persona de Mediana Edad , Receptores de Progesterona/metabolismo
5.
Br J Cancer ; 95(2): 172-80, 2006 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-16819546

RESUMEN

Resistance to antiepidermal growth factor (EGFR) strategies is an emerging clinical problem. Using human colorectal cancer (CRC) cells, we evaluated the involvement of the insulin receptor isoform-A (InsR-A) in de novo resistance to gefitinib, an EGFR tyrosine kinase inhibitor. Challenging the EGFR positive LoVo cells with gefitinib (1 microM) resulted in a small ( approximately 18%) inhibition of cell growth and although a modest reduction in phospho (p)EGFR Tyr845 was seen, pEGFR at residues -Tyr1068 and -Tyr1173 were unchanged. LoVo cells produced unprocessed pro-IGF-1R protein, substantial levels of IGF-II mRNA and mature InsR protein, consisting mainly of the InsR-A isoform. Insulin and IGF-II promoted cell growth and pEGFR Tyr845, Tyr1068 and Tyr1173 activity and conversely, the insulin-like growth factor-1 receptor (IGF-1R)/InsR inhibitor ABDP (1 muM) inhibited growth and reduced pEGFR activity at all three tyrosine residues. pInsR and pAkt levels were increased after gefitinib treatment. Blocking of pInsR with ABDP enabled gefitinib to markedly reduce pEGFR Tyr845, Tyr1068 and Tyr1173. Short-term gefitinib/ABDP dual treatment was more effective than either agent alone and chronic exposure to this combination resulted in total cell loss after 9 weeks, preventing acquisition of resistance to ABDP. LoVo cells with acquired resistance to ABDP were acutely sensitive to gefitinib. We concluded that InsR-A reduces sensitivity to gefitinib in LoVo CRC cells, thus its co-targeting alongside EGFR can improve the anti-tumour effect of gefitinib.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos , Quinazolinas/farmacología , Receptor de Insulina/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Difosfonatos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Gefitinib , Humanos , Insulina/farmacología , Factor II del Crecimiento Similar a la Insulina/farmacología , Fosforilación , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/biosíntesis , ARN Mensajero/biosíntesis , Receptor IGF Tipo 1/biosíntesis , Receptor de Insulina/biosíntesis , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
6.
Breast Cancer Res Treat ; 96(2): 131-46, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16261397

RESUMEN

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.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Moduladores de los Receptores de Estrógeno/farmacología , Receptores de Estrógenos/metabolismo , Transducción de Señal , Tamoxifeno/farmacología , Western Blotting , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Endocr Relat Cancer ; 13 Suppl 1: S77-88, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17259561

RESUMEN

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.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/metabolismo , Resistencia a Antineoplásicos/genética , Estrógenos/farmacología , Animales , Neoplasias de la Mama/genética , Carcinoma/genética , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Endocr Relat Cancer ; 12 Suppl 1: S1-7, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16113086

RESUMEN

Anti-hormones (notably tamoxifen), chemotherapy and modern radiotherapeutic approaches are invaluable in the management of breast cancer, and collectively have contributed substantially to the improved survival in this disease. Moreover, there is promise that these successes will continue with the emergence of other endocrine agents (for example, aromatase inhibitors and pure anti-oestrogens). However, de novo and acquired resistance comprises a significant problem with all treatment approaches examined to date. This Workshop aimed to evaluate the contribution made by growth factor signalling pathways in the various resistant states, primarily focusing on resistance to anti-hormonal strategies and spanning experimental models and, where possible, clinical breast cancer data. The successes and limitations of therapeutic targeting of these pathways with various signal transduction inhibitors (STIs) were evaluated in model systems and from emerging clinical trials (including epidermal growth factor receptor inhibitors such as gefitinib). It was concluded that growth factor signalling is an important contributor in the development of endocrine resistance in breast cancer and that use of STIs provides a promising therapeutic strategy for this disease. However, the cancer cell is clearly able to harness alternative growth factor signalling pathways for growth and cell survival in the presence of STI monotherapy and, as a consequence, the efficacy of STIs is likely to be limited by the acquisition of resistance. A number of strategies were proposed from studies in model systems that appeared to enhance anti-tumour actions of existing STI monotherapy, notably including combination therapies targeting multiple pathways. With the increased availability of diverse STIs and improved drug delivery, there is much hope that the more complex therapeutic strategies proposed may ultimately be achievable in clinical practice.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de Crecimiento/uso terapéutico , Antagonistas de Hormonas/uso terapéutico , Transducción de Señal , Resistencia a Antineoplásicos , Quimioterapia Combinada , Femenino , Humanos , Transducción de Señal/efectos de los fármacos
9.
Endocr Relat Cancer ; 12 Suppl 1: S135-44, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16113090

RESUMEN

Studies of cell models and profiling of clinical breast cancer material to reveal the mechanisms of resistance to anti-oestrogen therapy, and to tamoxifen in particular, have reported that this phenomenon can be associated with increased expression and signalling through erbB Type 1 growth factor receptors, notably the epidermal growth factor receptor (EGFR) and HER2. Further molecular studies have revealed an intricate interlinking between such growth factor receptor pathways and oestrogen receptor (ER) signalling. Inhibition of receptor tyrosine kinase activity involved in the EGFR signalling cascade forms the basis for the use of EGFR specific tyrosine kinase inhibitors exemplified by gefitinib (ZD1839, Iressa) and erlotinib (OSI-774, Tarceva). Such agents have proved promising in pre-clinical studies and are currently in clinical trials in breast cancer, where gefitinib has been studied more extensively to date. Here, we present an overview of the current development of gefitinib in clinical breast cancer. This includes results from our clinical breast cancer trial 1839IL/0057 that demonstrate the efficacy of gefitinib within ER-positive, tamoxifen-resistant patients with locally advanced/metastatic disease, where parallel decreases in EGFR signal transduction and the Ki67 (MIB1) proliferation marker can be detected as predicted from model system studies. We also consider trials examining combination treatment with gefitinib and anti-hormonal strategies that will begin to address the clinically important question of whether gefitinib can delay/prevent onset of anti-hormone resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Neoplasias de la Mama/enzimología , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib , Femenino , Gefitinib , Humanos
10.
Endocr Relat Cancer ; 12 Suppl 1: S173-82, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16113094

RESUMEN

Aberrant signalling through the epidermal growth factor receptor (EGFR) is associated with increased cancer cell proliferation, reduced apoptosis, invasion and angiogenesis. Over-expression of the EGFR is seen in a variety of tumours and is a rational target for antitumour strategies. Among the classes of agent targeting the EGFR are small-molecule inhibitors, which include gefitinib (IRESSA), which acts by preventing EGFR phosphorylation and downstream signal transduction. De novo and acquired resistance, however, have been reported to gefitinib and here we describe evidence which indicates that the type II receptor tyrosine kinases (RTKs) insulin-like growth factor-I receptor (IGF-IR) and/or insulin receptor (InsR) play important roles in the mediation of responses to gefitinib in the de novo- and acquired-resistance phenotypes in several cancer types. Moreover, combination strategies that additionally target the IGF-IR/InsR can enhance the antitumour effects of gefitinib.


Asunto(s)
Antineoplásicos/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Antineoplásicos/farmacología , Proliferación Celular , Resistencia a Antineoplásicos , Gefitinib , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos
11.
Endocr Relat Cancer ; 12 Suppl 1: S29-36, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16113097

RESUMEN

De novo insensitivity and acquired resistance to the selective oestrogen receptor modulator tamoxifen and the pure anti-oestrogen fulvestrant (faslodex) severely limit their effectiveness in breast cancer patients. This is a major clinical problem, since each year upward of 1 million women are dispensed anti-oestrogenic drugs. In order to investigate the phenomenon of anti-oestrogen resistance and to rapidly screen drugs that target the resistance mechanism(s), we have previously established several in vitro breast cancer models that have acquired resistance to anti-hormones. Such cells commonly develop an ability to proliferate after approximately 3 months of exposure to 4-hydroxytamoxifen or fulvestrant, despite an initial endocrine-responsive (i.e. growth-suppressive) phase. The current paper explores the role that growth factor signalling plays in the transition of oestrogen receptor-positive endocrine-responsive breast cancer cells to anti-oestrogen resistance or insensitivity and how we might, in the future, most effectively use anti-growth factor therapies to treat or delay endocrine-resistant states.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Antagonistas de Estrógenos/uso terapéutico , Inhibidores de Crecimiento/uso terapéutico , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Quimioterapia Combinada , Femenino , Sustancias de Crecimiento/metabolismo , Humanos , Fosforilación , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Endocrinology ; 146(11): 4609-18, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16037379

RESUMEN

There is considerable evidence that the epidermal growth factor receptor (EGFR) and IGF-I receptor (IGF-IR) cross-talk in breast cancer cells. In the present study, we have examined whether EGFR/IGF-IR cross-talk exists in EGFR-positive tamoxifen-resistant variants of MCF-7 (Tam-R) and T47D (T47D-R) breast cancer cell lines. Although Tam-R cells expressed reduced IGF-IR protein levels compared with their wild-type MCF-7 counterparts, phosphorylated IGF-IR protein levels were equivalent in the two cell lines under basal growth conditions, possibly as a consequence of increased IGF-II expression in Tam-R cells. IGF-II activated both IGF-IR and EGFR in Tam-R cells, whereas only activation of IGF-IR was observed in wild-type cells. In contrast, epidermal growth factor rapidly induced EGFR, but not IGF-IR, phosphorylation in Tam-R cells. IGF-II promoted direct association of c-SRC with IGF-IR, phosphorylated c-SRC, and increased EGFR phosphorylation at tyrosine 845, a c-SRC-dependent phosphorylation site. Pretreatment with either AG1024 (IGF-IR-specific inhibitor) or an IGF-II neutralizing antibody inhibited basal IGF-IR, c-SRC, and EGFR phosphorylation, and AG1024 significantly reduced Tam-R basal cell growth. The c-SRC inhibitor SU6656 also inhibited growth, reduced basal and IGF-II-induced c-SRC and EGFR phosphorylation, and blocked EGFR activation by TGFalpha. Similarly, in T47D-R cells, AG1024 and SU6656 inhibited basal and IGF-II-induced phosphorylation of c-SRC and EGFR, and SU6656 reduced TGFalpha-induced EGFR activity. These results suggest the existence of a unidirectional IGF-IR/EGFR cross-talk mechanism whereby IGF-II, acting through the IGF-IR, regulates basal and ligand-activated EGFR signaling and cell proliferation in a c-SRC-dependent manner in Tam-R cells. This cross-talk between IGF-IR and EGFR is not unique to Tam-R cells because this mechanism is also active in a tamoxifen-resistant T47D-R cell line.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/fisiopatología , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Tamoxifeno/farmacología , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/efectos de los fármacos , Femenino , Humanos , Factor II del Crecimiento Similar a la Insulina/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptor Cross-Talk , Receptor IGF Tipo 1/antagonistas & inhibidores , Factor de Crecimiento Transformador alfa/farmacología
13.
J Steroid Biochem Mol Biol ; 93(2-5): 257-62, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15860268

RESUMEN

Recent evidence demonstrates that growth factor networks are highly interactive with the estrogen receptor (ER) in the control of breast cancer growth and development. As such, tumor responses to anti-hormones are likely to be a composite of the ER and growth factor inhibitory activity of these agents, with alterations/aberrations in growth factor signalling providing a mechanism for the development of anti-hormone resistance. In this light, the current article focuses on illustrating the relationship between growth factor signalling and anti-hormone failure in our in-house tumor models of breast cancer and describes how we are now beginning to successfully target their actions to improve the effects of anti-hormonal drugs and to block aggressive disease progression.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Sustancias de Crecimiento/metabolismo , Resistencia a Antineoplásicos , Quimioterapia Combinada , Receptores ErbB/metabolismo , Femenino , Antagonistas de Hormonas/uso terapéutico , Humanos , Invasividad Neoplásica , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Tamoxifeno/uso terapéutico , Insuficiencia del Tratamiento
14.
Endocr Relat Cancer ; 11(4): 623-41, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15613443

RESUMEN

There is an increasing body of evidence demonstrating that elevated growth signaling in breast cancer cells can promote forms of endocrine resistance in either an estrogen receptor-dependent or -independent manner. The current article reviews what is known about such growth factor signaling networks and resistance to estrogen withdrawal and considers the many novel therapeutic opportunities that stem from this knowledge.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estrógenos/metabolismo , Sustancias de Crecimiento/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Femenino , Humanos , Receptores de Estrógenos/metabolismo , Transducción de Señal
15.
Endocr Relat Cancer ; 11(4): 793-814, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15613453

RESUMEN

De novo and acquired resistance to the anti-tumour drug gefitinib (ZD1839; Iressa), a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) has been reported. We have determined whether signalling through the IGF-I receptor (IGF-1R) pathway plays a role in the gefitinib-acquired resistance phenotype. Continuous exposure of EGFR-positive MCF-7-derived tamoxifen resistant breast cancer cells (TAM-R) to 1 microM gefitinib resulted in a sustained growth inhibition (90%) for 4 months before the surviving cells resumed proliferation. A stable gefitinib-resistant subline (TAM/TKI-R) was established after a further 2 months and this showed no detectable basal phosphorylated EGFR activity. Compared with the parental TAM-R cells, the TAM/ TKI-R cells demonstrated (a) elevated levels of activated IGF-1R, AKT and protein kinase C (PKC)delta, (b) an increased sensitivity to growth inhibition by the IGF-1R TKI AG1024 and (c) an increased migratory capacity that was reduced by AG1024 treatment. Similarly, the EGFR-positive androgen-independent human prostate cancer cell line DU145 was also continuously challenged with 1 microM gefitinib and, although substantial growth inhibition (60%) was seen initially, a gefitinib-resistant variant (DU145/TKI-R) developed after 3 months. Like their breast cancer counterparts, the DU145/TKI-R cells showed increases in the levels of components of the IGF-1R signalling pathway and an elevated sensitivity to growth inhibition by AG1024 compared with the parent DU145 cell line. Additionally, DU145/TKI-R cell migration was also decreased by this inhibitor. We have therefore concluded that in breast and prostate cancer cells acquired resistance to gefitinib is associated with increased signalling via the IGF-1R pathway, which also plays a role in the invasive capacity of the gefitinib-resistant phenotype.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Quinazolinas/farmacología , Receptor IGF Tipo 1/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , ADN Complementario/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinib , Regulación Neoplásica de la Expresión Génica , Sustancias de Crecimiento/genética , Sustancias de Crecimiento/metabolismo , Sustancias de Crecimiento/farmacología , Humanos , Receptor IGF Tipo 1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
16.
Endocrinology ; 144(11): 5105-17, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12960029

RESUMEN

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.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Proteínas Serina-Treonina Quinasas , Quinazolinas/farmacología , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Combinación de Medicamentos , Resistencia a Medicamentos/efectos de los fármacos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Receptores ErbB/fisiología , Femenino , Fulvestrant , Gefitinib , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/efectos de los fármacos
17.
Ann N Y Acad Sci ; 963: 104-15, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12095935

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos/fisiología , Receptores ErbB/metabolismo , Estradiol/análogos & derivados , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , División Celular/efectos de los fármacos , Glándulas Endocrinas , Estradiol/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Femenino , Fulvestrant , Gefitinib , Humanos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/farmacología , Receptor ErbB-2/fisiología , Transducción de Señal/fisiología , Tamoxifeno/farmacología , Trastuzumab , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA