Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Ann Oncol ; 24(3): 784-91, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23104723

RESUMO

BACKGROUND: Type 1 insulin-like growth factor receptor (IGF-1R) mediates resistance to chemotherapy and targeted agents. This study assessed the safety, pharmacokinetics (PK), and tolerability of humanized IGF-1R antibody AVE1642 with other cancer treatments. PATIENTS: Patients with advanced solid tumors received three weekly AVE1642 dosed at 6 mg/kg, chosen following previous study, with 75 (cohort A) or 100 mg/m(2) (B) docetaxel, 1250 mg/m(2) gemcitabine/100 mg erlotinib (C1), or 60 mg/m(2) doxorubicin (D1). Blood samples were assayed for PK, IGFs, and IGF-BP3. RESULTS: Fifty-eight patients received 317 AVE1642 infusions. The commonest adverse events were diarrhea (37/58 patients), asthenia (34/58), nausea (30/58), and stomatitis (21/58). Dose-limiting toxic effects in cohorts C1 (diarrhea) and D1 (neutropenia) prompted addition of cohorts C2 (1000 mg/m(2) gemcitabine/75 mg erlotinib) and D2 (50 mg/m(2) doxorubicin). Grade 3-4 hyperglycemia (three cases) accompanied steroid premedication for docetaxel administration. No PK interactions were detected. There were three partial responses in cohorts B (melanoma) and C (leiomyosarcoma, two cases) and 22 stabilizations ≥12 weeks, giving a control rate of 25/57 (44%). On treatment IGF-II rose by 68 ± 25 ng/ml in patients discontinuing treatment <12 weeks, and fell by 55.5 ± 21 ng/ml with disease control (P < 0.001). CONCLUSION: AVE1642 was tolerable with 75-100 mg/m(2) docetaxel and 1000 mg/m(2) gemcitabine/75 mg erlotinib, achieving durable disease control in 44%, with an association between IGF-II and response.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leiomiossarcoma/tratamento farmacológico , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias de Tecidos Moles/tratamento farmacológico , Adulto , Idoso , Anticorpos Monoclonais Humanizados/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Diarreia/induzido quimicamente , Docetaxel , Doxorrubicina/administração & dosagem , Cloridrato de Erlotinib , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Quinazolinas/administração & dosagem , Receptor IGF Tipo 1/imunologia , Taxoides/administração & dosagem , Resultado do Tratamento , Gencitabina
2.
BMC Cancer ; 12: 590, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23231599

RESUMO

BACKGROUND: Sorafenib is an orally available kinase inhibitor with activity at Raf, PDGFß and VEGF receptors that is licensed for the treatment of advanced renal cell carcinoma (RCC) and hepatocellular carcinoma (HCC). Current evidence-based post-nephrectomy management of individuals with localized RCC consists of surveillance-based follow up. The SORCE trial is designed to investigate whether treatment with adjuvant sorafenib can reduce recurrence rates in this cohort. CASE PRESENTATION: Here we report an idiosyncratic reaction to sorafenib resulting in fatal hepatotoxicity and associated renal failure in a 62 year-old man treated with sorafenib within the SORCE trial. CONCLUSION: This is the first reported case of sorafenib exposure associated fatal toxicity in the adjuvant setting and highlights the unpredictable adverse effects of novel adjuvant therapies.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Antineoplásicos/efeitos adversos , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Quimioterapia Adjuvante , Evolução Fatal , Humanos , Masculino , Pessoa de Meia-Idade , Niacinamida/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , Sorafenibe
3.
Oncogene ; 26(45): 6499-508, 2007 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-17486080

RESUMO

Clear cell renal cell cancer (CC-RCC) is a highly chemoresistant tumor characterized by frequent inactivation of the von Hippel-Lindau (VHL) gene. The prognosis is reportedly worse in patients whose tumors express immunoreactive type I insulin-like growth factor receptor (IGF1R), a key mediator of tumor cell survival. We aimed to investigate how IGF1R expression is regulated, and found that IGF1R protein levels were unaffected by hypoxia, but were higher in CC-RCC cells harboring mutant inactive VHL than in isogenic cells expressing wild-type (WT) VHL. IGF1R mRNA and promoter activities were significantly lower in CC-RCC cells expressing WT VHL, consistent with a transcriptional effect. In Sp1-null Drosophila Schneider cells, IGF1R promoter activity was dependent on exogenous Sp1, and was suppressed by full-length VHL protein (pVHL) but only partially by truncated VHL lacking the Sp1-binding motif. pVHL also reduced the stability of IGF1R mRNA via sequestration of HuR protein. Finally, IGF1R mRNA levels were significantly higher in CC-RCC biopsies than benign kidney, confirming the clinical relevance of these findings. Thus, we have identified a new hypoxia-independent role for VHL in suppressing IGF1R transcription and mRNA stability. VHL inactivation leads to IGF1R upregulation, contributing to renal tumorigenesis and potentially also to chemoresistance.


Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Receptor IGF Tipo 1/metabolismo , Regulação para Cima , Proteína Supressora de Tumor Von Hippel-Lindau/fisiologia , Humanos , Rim/metabolismo , RNA Mensageiro/metabolismo , Fator de Transcrição Sp1/fisiologia , Transcrição Gênica , Células Tumorais Cultivadas
4.
Oncogene ; 25(50): 6574-81, 2006 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-16715137

RESUMO

The type 1 insulin-like growth factor receptor (IGF1R) is overexpressed by malignant melanomas compared with benign naevi, and mediates proliferation, motility and protection from apoptosis. However, the utility of IGF1R targeting as anti-cancer therapy may be limited by activating mutations in downstream signaling intermediates. We previously showed that IGF1R knockdown blocked survival of prostate cancer cells in which Akt activation was deregulated by PTEN loss. The current study investigated effects of IGF1R targeting in cells harboring activating RAS-RAF mutations, found in 70-80% of human melanomas. We assembled a panel of eight human melanoma cell lines: two expressing wild-type (WT) B-RAF and N-RAS, two with activating N-RAS mutations and four harboring V600E B-RAF. We also generated isogenic cell populations overexpressing WT or V600E B-RAF. Cells expressing V600E B-RAF were relatively resistant to apoptosis. However, IGF1R gene silencing was capable of inducing significant inhibition of survival, enhancement of apoptosis, and approximately two-fold sensitization to cisplatin and temozolomide. These effects were independent of mutation status and were associated with reduced activation of Akt and also, unexpectedly, of ERKs. These results support development of IGF1R targeting as therapy for melanoma, regardless of the presence of activating mutations in the RAS-RAF pathway.


Assuntos
Melanoma/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor IGF Tipo 1/metabolismo , Antineoplásicos/uso terapêutico , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inativação Gênica , Humanos , Melanócitos/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , Mutação Puntual , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Transfecção , Células Tumorais Cultivadas
5.
Endocr Relat Cancer ; 13 Suppl 1: S33-43, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17259557

RESUMO

The type 1 IGF receptor (IGF1R) is a transmembrane tyrosine kinase that is frequently overexpressed by tumours, and mediates proliferation and apoptosis protection. IGF signalling also influences hypoxia signalling, protease secretion, tumour cell motility and adhesion, and thus can affect the propensity for invasion and metastasis. Therefore, the IGF1R is now an attractive anti-cancer treatment target. This review outlines the effects of IGF1R activation in tumour cells, and will describe the strategies that are available to block IGF signalling, both as investigational tools and as novel anti-cancer therapeutics. Design of specific IGF1R inhibitors has been problematic due to close homology with the insulin receptor, but recently it has proved possible to design selective IGF1R inhibitors. These compounds and IGF1R antibodies are showing promise in preclinical models of human cancer, and several agents are now in early phase clinical trials. Both classes of agents affect insulin receptor signalling, either by direct kinase inhibition or antibody-induced insulin receptor downregulation. This effect may lead to clinical toxicity, but could be therapeutically beneficial in blocking signalling via variant insulin receptors capable of a mitogenic response to IGF-II. Specificity for IGF1R targeting can be achieved by antisense and siRNA-mediated IGF1R downregulation; these approaches have undoubted utility as research tools, and may in future generate nucleic-acid-based therapeutics. It will be important to use data from preclinical and early clinical trials to establish the molecular correlates of sensitivity to IGF1R blockade, and the optimum means of combining this new approach with standard treatment modalities.


Assuntos
Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/fisiologia , Transdução de Sinais , Animais , Anticorpos Monoclonais/farmacologia , Citotoxinas/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/fisiologia , Neoplasias/tratamento farmacológico , Oligonucleotídeos Antissenso/farmacologia , Interferência de RNA , Radiossensibilizantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
6.
Cancer Res ; 50(8): 2511-7, 1990 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-2156621

RESUMO

We showed previously that insulin-like growth factor I (IGF-I) is detectable in small cell lung cancer (SCLC) tumor biopsies and cell lines and that recombinant human IGF-I stimulates DNA synthesis in SCLC cells. Here we report further studies on the role of IGF-I in 2 SCLC cell lines: HC12, classic; and ICR-SC17, variant. Immunoreactive IGF-I was detected in medium conditioned by HC12 but not ICR-SC17. Both HC12 and ICR-SC17 bound IGF-I with 100-fold greater affinity than insulin. Scatchard analysis revealed two classes of IGF-I binding site of high (Kd 0.1 nM, n = 2,300) and lower (Kd 3 nM, n = 28,000) affinity. In both cell lines [3H]thymidine incorporation was enhanced by recombinant human IGF-I, 100-1000 ng/ml. ICR-SC17 also showed growth enhancement as measured by increase in cell numbers. There was no response in HC12, probably due to endogenous IGF-I production. 125I-IGF-I binding and basal and IGF-I-stimulated mitogenesis were inhibited by monoclonal antibodies to IGF-I (SM1.20B, SM1.25) or the type I IGF receptor alpha IR3 but not an isotypic control monoclonal antibody. Antiproliferative effects were manifest in [3H]thymidine incorporation assays in serum-free conditions and growth of serum-supplemented liquid cultures. We also tested fresh or newly cultured tumor cells obtained by fine needle aspiration of metastases in three previously untreated and four relapsed patients with SCLC. IGF-I binding sites were demonstrable on fresh SCLC cells, and specific binding was inhibited by SM1.20B. All seven samples showed stimulation of [3H]thymidine incorporation in the presence of recombinant human IGF-I, 100-500 ng/ml. As in cultured cells, basal and IGF-I-stimulated DNA synthesis was inhibited by monoclonal antibodies SM1.20B, SM1.25, and alpha IR3 but not the isotypic control. These results confirm the findings of previous studies and suggest that IGF-I can function as an autocrine growth factor in SCLC in vitro and possibly also in vivo.


Assuntos
Carcinoma de Células Pequenas/patologia , Fator de Crescimento Insulin-Like I/farmacologia , Neoplasias Pulmonares/patologia , Somatomedinas/farmacologia , Células Tumorais Cultivadas/citologia , Anticorpos Monoclonais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Replicação do DNA/efeitos dos fármacos , Imunofluorescência , Humanos , Fator de Crescimento Insulin-Like I/análise , Fator de Crescimento Insulin-Like I/fisiologia , Cinética , Radioimunoensaio , Ensaio Radioligante , Receptores de Superfície Celular/metabolismo , Receptores de Somatomedina , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos
7.
Oncogene ; 20(30): 4029-40, 2001 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-11494131

RESUMO

The type 1 insulin-like growth factor receptor (IGF1R) is required for growth, tumorigenicity and protection from apoptosis. IGF1R overexpression is associated with radioresistance in breast cancer. We used antisense (AS) RNA to downregulate IGF1R expression in mouse melanoma cells. Cells expressing AS-IGF1R transcripts were more radiosensitive in vitro and in vivo than controls. Also they showed reduced radiation-induced p53 accumulation and p53 serine 18 phosphorylation, and radioresistant DNA synthesis. These changes were reminiscent of the cellular phenotype of the human genetic disorder ataxia-telangiectasia (A-T), caused by mutations in the ATM gene. Cellular Atm protein levels were lower in AS-IGF1R-transfected cells than in control cells, although there was no difference in Atm expression at the transcriptional level. AS-IGF1R cells had detectable basal Atm kinase activity, but failed to induce kinase activity after irradiation. This suggests that IGF1R signalling can modulate the function of Atm, and supports the concept of targeted IGF1R downregulation as a potential treatment for malignant melanoma and other radioresistant tumours.


Assuntos
Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/genética , Proteínas de Neoplasias/metabolismo , Tolerância a Radiação/genética , Receptor IGF Tipo 1/biossíntese , Animais , Apoptose , Ataxia Telangiectasia/patologia , Ativação Enzimática , Feminino , Humanos , Melanoma Experimental/enzimologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/fisiologia , Transfecção , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/efeitos da radiação , Células Tumorais Cultivadas/transplante
8.
Clin Cancer Res ; 5(3): 513-20, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10100701

RESUMO

Tumor cells and associated stromal cells secrete matrix metalloproteinases (MMPs), contributing to invasion, angiogenesis, and metastasis. Batimastat (BB-94) is a broad-spectrum MMP inhibitor that causes resolution of ascites and/or tumor growth delay in animal models of breast, ovarian, and colorectal cancer. We recruited 18 patients with cytologically positive malignant pleural effusions into a Phase I study of intrapleural BB-94. Three patients received single doses of BB-94 at each dose level: 15, 30, 60, 105, 135, and 300 mg/m2. Two patients were retreated with a second course at 60 and 105 mg/m2. BB-94 was detectable in plasma 1 h after intrapleural administration, and peak levels of 20-200 ng/ml occurred after 4 h to 1 week. BB-94 persisted in the plasma for up to 12 weeks, at levels exceeding the IC50s for target MMPs. Peak values were higher, and persistence in the plasma was longer after higher doses of BB-94. The treatment was well tolerated. Toxic effects included low-grade fever for 24-48 h (6 of 18 patients, 33%) and reversible asymptomatic elevation of liver enzymes (8 patients, 44%). Toxicity seemed unrelated to BB-94 dose or plasma levels. Sixteen patients evaluable for response required significantly fewer pleural aspirations in the 3 months after BB-94 compared with the 3 months before. Seven patients (44%) required no further pleural aspiration until death/last follow-up. After 1 month, patients treated with 60-300 mg/m2 BB-94 had significantly better dyspnea scores, indicating improved exercise tolerance, compared with baseline scores the day after BB-94. The maximum tolerated intrapleural dose remains to be defined, but it is clear that intrapleural BB-94 is well tolerated, with evidence of local activity.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Inibidores de Metaloproteinases de Matriz , Fenilalanina/análogos & derivados , Derrame Pleural Maligno/tratamento farmacológico , Tiofenos/uso terapêutico , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Feminino , Humanos , Masculino , Metaloproteinase 1 da Matriz , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde , Fenilalanina/efeitos adversos , Fenilalanina/farmacocinética , Fenilalanina/uso terapêutico , Derrame Pleural Maligno/metabolismo , Tiofenos/efeitos adversos , Tiofenos/farmacocinética
9.
FEBS Lett ; 372(2-3): 222-8, 1995 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-7556673

RESUMO

The cytochrome P450 enzyme aromatase (P450arom) is an important target in breast cancer treatment. We have designed a 20-base pyrimidine oligodeoxynucleotide (ODN) which forms a sequence-specific triple helix (triplex) with a purine-rich tract in the P450arom coding sequence. The psoralen-linked ODN (Pso20T) formed photo-induced cross-linked products with target double-stranded DNA. Cross-linked adducts formed in vitro between ODNs and P450arom expression constructs were used to transfect COS and human MCF-7 breast cancer cells. Levels of aromatase transcripts and enzyme activity were significantly lower in cultures transfected with Pso20T-treated cDNA relative to controls. Pso20T had a lesser inhibitory effect on aromatase expression from a mutant P450arom construct, consistent with predicted effects of the mutations on triplex formation. These results are compatible with triplex-mediated interruption of transcription within intact cells.


Assuntos
Aromatase/biossíntese , Ficusina/metabolismo , Oligonucleotídeos/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Sequência de Bases , Sítios de Ligação , Humanos , Dados de Sequência Molecular , Células Tumorais Cultivadas
10.
Eur J Cancer ; 29A(10): 1450-3, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-7691115

RESUMO

We report the effect of substance P analogue, [D-Arg1, D-Phe5, D-Trp7,9, Leu11] substance P (D-Phe5SP), on the growth of human small cell lung cancer (SCLC) xenografts HC12 and ICR-SC112. Daily intraperitoneal (ip) administration (500 micrograms/day for 3 weeks) had no effect on HC12 growth rate. When administered by continuous 14-day subcutaneous (sc) infusion by osmotic minipump implanted adjacent to the tumour, D-Phe5SP 2.1 micrograms/day, caused significant inhibition (P < 0.05) of the growth of HC12 and ICR-SC112 on day 7 and day 14 compared with phosphate buffered saline (PBS)-treated controls. HC12 and ICR-SC112 tumour volume remained at 53-67% of control for 14-21 days postinfusion. D-Phe5SP 1 mg/day did not inhibit tumour growth, but dense fibrous capsules developed at the minipump outlet. Animals treated by sc infusion (but not ip) of PBS or D-Phe5SP failed to gain weight, and some groups lost weight. D-Phe5SP-treated animals had lower white blood counts than controls (not significant). These data suggest a potential clinical role for D-Phe5SP in the treatment of SCLC.


Assuntos
Carcinoma de Células Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Recombinantes , Substância P/análogos & derivados , Animais , Carcinoma de Células Pequenas/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores do Crescimento/uso terapêutico , Humanos , Injeções Subcutâneas , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Substância P/administração & dosagem , Substância P/uso terapêutico , Fatores de Tempo , Transplante Heterólogo
11.
J Endocrinol ; 138(3): 545-54, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7506292

RESUMO

Proteolytic modification of circulating insulin-like growth factor binding protein-3 (IGFBP-3) has been described in a number of conditions. Using Western ligand blotting and SDS-PAGE analysis of fragmentation patterns of 125I-labelled IGFBP-3 and 125-labelled IGFBP-1, we have examined conditioned media from cultured human cell lines for the presence of proteolytic activity and compared this with the action of circulating proteases and with characterized enzymes including cathepsin D, kallikrein, plasmin and tissue plasminogen activator. 125I-Labelled IGFBP-3 was incubated with serum from pregnant women, patients following heart surgery and patients with cancer of the breast, lung or head/neck. Following separation of the preincubated samples by SDS-PAGE, a distinct pattern of degradation fragments was observed which was similar in all cases. This proteolytic activity was inhibited in the presence of EDTA, phenanthroline and 4(-2-aminoethyl)-benzenesulphonylfluoride, HCl. These proteases had no detectable effect on IGFBP-1. Serum-free conditioned medium from a human dermal fibroblast cell line, a rabdomyosarcoma, a cervical, a bladder, a chorio- and two-tongue squamous cell carcinoma cell lines all contained proteolytic activity which fragmented IGFBP-3. The pattern of fragments was similar in all cell lines but different from that produced by the circulating proteases. Six out of nine cell lines produced protease(s) which degraded IGFBP-1 in addition to IGFBP-3. Whilst all the characterized enzymes tested also fragmented IGFBP-3 and plasmin cleaved IGFBP-1, none of these acted in the same way as either circulating or cell line-derived proteolytic activity. The activity associated with the characterized enzymes and cell lines was inhibited in the presence of serum from normal healthy subjects. These results demonstrate that the serum of pregnant women, post-operative patients and patients with cancer contain circulating proteases which cause fragmentation of IGFBP-3 but have little effect on IGFBP-1. Cell-derived proteases were shown to act on IGFBP-3 and IGFBP-1 in a number of instances but are not active in the presence of circulating inhibitors. These proteases may play an important role in regulating the availability of IGFs to normal and neoplastic tissues.


Assuntos
Proteínas de Transporte/efeitos dos fármacos , Peptídeo Hidrolases/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Proteínas de Transporte/sangue , Proteínas de Transporte/metabolismo , Meios de Cultura , Eletroforese em Gel de Poliacrilamida , Feminino , Glicosilação , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Peptídeo Hidrolases/sangue , Período Pós-Operatório , Proteínas Recombinantes/efeitos dos fármacos , Células Tumorais Cultivadas
12.
J Steroid Biochem Mol Biol ; 56(1-6 Spec No): 145-50, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8603035

RESUMO

The existence of aromatase activity in human breast carcinomas has been established for about 20 years but the clinical and biological importance of this remains unclear. A number of studies in clinical material suggest that aromatase activity may be a prerequisite of response to aromatase inhibitors and that aromatase activity may be enhanced in those tumours relapsing during treatment with one such inhibitor, aminoglutethimide. These results would carry more significance, however, if it was demonstrable that the growth of breast carcinomas is affected by the conversion of androgens to oestrogens by intratumoural aromatase. We have tried to address this by establishing model systems with aromatase-transfected MCF7 breast cancer cells. We have demonstrated that these cells can be stimulated mitogenically with androgen and that this proliferation is suppressible with aromatase inhibitors. Similarly the growth of aromatase transfected cells but not wild type cells as xenografts is supported by androstenedione and inhibitable by both the steroidal aromatase inhibitor, 4-hydroxyandrostenedione and non-steroidal inhibitor, CGS 20267. Work with the former of these, which is a suicide inhibitor allowed us to demonstrate that growth can proceed with aromatase activity approximating to the highest level seen in breast carcinomas indicating that at least at this extreme level the intratumoural conversion of androgens to oestrogens may indeed be able to support tumour growth. Further work with this mode system should allow us to define the minimal amount of intratumoural activity which can support tumour growth.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Aromatase/fisiologia , Neoplasias da Mama/enzimologia , Carcinoma/enzimologia , Inibidores Enzimáticos/uso terapêutico , Estrogênios , Proteínas de Neoplasias/fisiologia , Neoplasias Hormônio-Dependentes/enzimologia , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Androgênios/metabolismo , Androgênios/farmacologia , Androstenodiona/análogos & derivados , Androstenodiona/farmacologia , Androstenodiona/uso terapêutico , Animais , Antineoplásicos Hormonais/farmacologia , Inibidores da Aromatase , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Carcinoma/tratamento farmacológico , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/farmacologia , Estrogênios/biossíntese , Feminino , Humanos , Letrozol , Masculino , Camundongos , Proteínas de Neoplasias/antagonistas & inibidores , Recidiva Local de Neoplasia/enzimologia , Transplante de Neoplasias , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/patologia , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Triazóis/farmacologia , Triazóis/uso terapêutico , Células Tumorais Cultivadas
13.
Cancer Chemother Pharmacol ; 9(3): 179-81, 1982.
Artigo em Inglês | MEDLINE | ID: mdl-6891627

RESUMO

Hypomagnesaemia is recognised as a feature of the renal tubular defect produced by cis-platinum therapy for cancer. It may be sufficiently severe to cause tetany and grand mal fits. Attempts to correct established hypomagnesaemia whilst continuing cis-platinum therapy have not proved satisfactory. We have therefore investigated the prophylactic addition of 3 g magnesium sulphate to the high-dose platinum regimen with which metastatic malignant teratoma is treated in this unit. Serum magnesium levels have been measured in eight patients treated in this way and compared with those recorded for eight matched patients previously treated without routine magnesium supplements. Magnesium levels fell into the range frequently associated with clinical manifestation in five of the eight unsupplemented patients and only one of those given magnesium prophylactically. Mean serum magnesium levels were significantly higher in the supplemented group when compared using the paired t-test (P less than 0.01). Routine supplementation with intravenous magnesium sulphate is a simple and effective way of preventing symptomatic hypomagnesaemia associated with cis-platinum therapy.


Assuntos
Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Sulfato de Magnésio/uso terapêutico , Magnésio/sangue , Adolescente , Adulto , Idoso , Antineoplásicos/administração & dosagem , Cisplatino/administração & dosagem , Quimioterapia Combinada , Feminino , Humanos , Rim/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Neoplasias Ovarianas/tratamento farmacológico , Estudos Retrospectivos , Teratoma/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico
14.
Oncogene ; 33(45): 5262-73, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24186206

RESUMO

Inhibition of type 1 insulin-like growth factor receptor (IGF-1R) enhances tumor cell sensitivity to ionizing radiation. It is not clear how this effect is mediated, nor whether this approach can be applied effectively in the clinic. We previously showed that IGF-1R depletion delays repair of radiation-induced DNA double-strand breaks (DSBs), unlikely to be explained entirely by reduction in homologous recombination (HR) repair. The current study tested the hypothesis that IGF-1R inhibition induces a repair defect that involves non-homologous end joining (NHEJ). IGF-1R inhibitor AZ12253801 blocked cell survival and radiosensitized IGF-1R-overexpressing murine fibroblasts but not isogenic IGF-1R-null cells, supporting specificity for IGF-1R. IGF-1R inhibition enhanced radiosensitivity in DU145, PC3 and 22Rv1 prostate cancer cells, comparable to effects of Ataxia Telangiectasia Mutated inhibition. AZ12253801-treated DU145 cells showed delayed resolution of γH2AX foci, apparent within 1 h of irradiation and persisting for 24 h. In contrast, IGF-1R inhibition did not influence radiosensitivity or γH2AX focus resolution in LNCaP-LN3 cells, suggesting that radiosensitization tracks with the ability of IGF-1R to influence DSB repair. To differentiate effects on repair from growth and cell-survival responses, we tested AZ12253801 in DU145 cells at sub-SF50 concentrations that had no early (⩽48 h) effects on cell cycle distribution or apoptosis induction. Irradiated cultures contained abnormal mitoses, and after 5 days IGF-1R-inhibited cells showed enhanced radiation-induced polyploidy and nuclear fragmentation, consistent with the consequences of entry into mitosis with incompletely repaired DNA. AZ12253801 radiosensitized DNA-dependent protein kinase (DNA-PK)-proficient but not DNA-PK-deficient glioblastoma cells, and did not radiosensitize DNA-PK-inhibited DU145 cells, suggesting that in the context of DSB repair, IGF-1R functions in the same pathway as DNA-PK. Finally, IGF-1R inhibition attenuated repair by both NHEJ and HR in HEK293 reporter assays. These data indicate that IGF-1R influences DSB repair by both major DSB repair pathways, findings that may inform clinical application of this approach.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Receptor IGF Tipo 1/genética , Reparo de DNA por Recombinação/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/efeitos da radiação , Western Blotting , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos da radiação , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Células HEK293 , Histonas/efeitos dos fármacos , Histonas/metabolismo , Histonas/efeitos da radiação , Recombinação Homóloga/efeitos dos fármacos , Recombinação Homóloga/genética , Recombinação Homóloga/efeitos da radiação , Humanos , Isoxazóis/farmacologia , Camundongos Knockout , Morfolinas/farmacologia , Pirimidinas/farmacologia , Pironas/farmacologia , Quinolinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Reparo de DNA por Recombinação/efeitos dos fármacos , Reparo de DNA por Recombinação/efeitos da radiação , Tiazóis/farmacologia
15.
Endocrinology ; 151(4): 1418-27, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20179263

RESUMO

Insulin receptor (IR) overexpression is common in cancers, with expression of the A isoform (IR-A, exon 11-) predominating over the B isoform. The IR-A signals a proliferative, antiapoptotic response to IGF-II, which itself can be secreted by tumors to establish an autocrine proliferative loop. Therefore, IGF-II signaling via the IR-A could mediate resistance to type 1 IGF receptor (IGF-IR) inhibitory drugs that are currently in development. This study addressed the role of the IR-A, using a small interfering RNA-based approach in SW480 human colon adenocarcinoma cells that coexpress the IGF-IR. Clonogenic survival was inhibited by depletion of the IGF-IR but not the IR-A, and dual receptor depletion had no greater effect than IGF-IR knockdown alone, suggesting that the IR-A could not compensate for IGF-IR loss. IGF-IR knockdown also resulted in a decrease in viability, whereas IR-A depletion resulted in increased viability. Consistent with this, upon IR-A depletion, we found a concomitant enhancement of IGF-IR activation by IGF-I and IGF-II, reduced formation of IGF-IR:IR-A hybrid receptors and increased IGF-IR homodimer formation. Together, these results suggest that IGF bioactivity is mediated more effectively by the IGF-IR than by the IR-A or receptor hybrids and that signaling via the IGF-IR is dominant to the IR-A in colon cancer cells that express both receptors.


Assuntos
Inativação Gênica/fisiologia , Multimerização Proteica/genética , Receptor de Insulina/genética , Western Blotting , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Imunoprecipitação , Indanos , Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like II/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
17.
Ann Oncol ; 13(3): 349-56, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11996463

RESUMO

The purpose of this review is to examine recent evidence that investigates the role of the insulin-like growth factor (IGF) system in colorectal cancer. We concentrate on the evidence that makes the case for the investigation of strategies that might be used to disrupt the IGF system in prevention and treatment. Even though the weight of evidence suggests that components of the IGF system may be appropriate targets, there are a lack of studies that make a systematic characterisation of all the system components in human colorectal cancer. It is anticipated that this information, and the new therapeutic molecules which follow, will impact on the prevention and treatment of patients with this disease.


Assuntos
Neoplasias Colorretais/prevenção & controle , Somatomedinas/efeitos dos fármacos , Animais , Neoplasias Colorretais/fisiopatologia , Humanos , Camundongos , Somatomedinas/fisiologia
18.
Horm Metab Res ; 35(11-12): 843-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14710367

RESUMO

The type 1 insulin-like growth factor receptor (IGF1R) is a promising anticancer treatment target, being frequently overexpressed by tumours, and mediating proliferation, motility and apoptosis protection. Design of specific kinase inhibitors is problematic because of homology between the IGF1R and insulin receptor. This obstacle can be circumvented using sequence-specific molecular agents including antisense, triplex and ribozymes. Recent studies indicate that profound sequence-specific IGF1R gene silencing can be induced by small interfering RNAs that mediate RNA interference in mammalian cells. IGF1R downregulation blocks tumour growth and metastasis, and enhances sensitivity to cytotoxic drugs and irradiation. In murine melanoma cells, radiosensitisation is associated with impaired activation of Atm, which is required for initiation of cell cycle checkpoints and DNA repair pathways after double-strand DNA breaks. Furthermore, tumour cells killed in vivo following IGF1R downregulation can provoke an immune response, protecting against tumour rechallenge. After years of studying the role of the IGF system in tumour biology, novel agents for IGF1R targeting will soon be available for clinical testing. This review summarises the development of molecular agents, and considers factors that will influence clinical activity, including the requirement of established tumours for IGF signalling, and the efficacy and toxicity of IGF1R inhibitors.


Assuntos
Antineoplásicos , Regulação da Expressão Gênica/genética , Neoplasias/tratamento farmacológico , Receptor IGF Tipo 1/antagonistas & inibidores , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , RNA Interferente Pequeno , Receptor IGF Tipo 1/genética
19.
Postgrad Med J ; 61(714): 331-2, 1985 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-4022864

RESUMO

Emboli from left atrial myxomas are a recognized cause of cerebral ischaemia. The myxomatous embolus may infiltrate the cerebral arterial wall, resulting in aneurysmal dilatation with a risk of rupture. Myxoma-associated cerebral haemorrhage has been described in patients with progressive neurological dysfunction. We report a new presentation. Our patient, a previously healthy 20 year old girl, developed acute intracerebral haemorrhage for which craniotomy was performed. Histology of evacuated haematoma revealed an intravascular fragment of myxoma. Echocardiography demonstrated a left atrial myxoma which later was uneventfully excised. Early diagnosis of embolic cardiac myxoma requires awareness of the diversity of clinical manifestations, and careful histological analysis of surgical specimens.


Assuntos
Hemorragia Cerebral/etiologia , Neoplasias Cardíacas/complicações , Mixoma/complicações , Adulto , Feminino , Átrios do Coração , Neoplasias Cardíacas/patologia , Humanos , Mixoma/patologia
20.
BJU Int ; 91(3): 271-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12581018

RESUMO

OBJECTIVE: To assess the effect of the downregulation of type 1 insulin-like growth factor receptor (IGF1R) on the chemosensitivity of prostate cancer cells. IGF1R is overexpressed by prostate cancer compared with benign prostatic epithelium and IGF1R expression commonly persists in androgen-independent metastatic disease at levels comparable to those in the primary. MATERIALS AND METHODS: Human androgen-independent DU145 prostate cancer cells were transfected with IGF1R antisense oligonucleotides or antisense RNA. Transfected cultures were treated with cisplatin, mitoxantrone, paclitaxel or vehicle control, and survival measured using a clonogenic assay. RESULTS: Both antisense strategies suppressed IGF1R protein levels to 30-50% of those in control cultures. This was associated with 1.5-2-fold enhancement of sensitivity to cisplatin, mitoxantrone and paclitaxel, and an increase in cisplatin-induced apoptosis. CONCLUSION: This approach has potential for development as a clinical treatment for advanced prostate cancer and other chemoresistant tumours.


Assuntos
Oligonucleotídeos Antissenso/uso terapêutico , Neoplasias da Próstata/metabolismo , Receptor IGF Tipo 1/metabolismo , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Cisplatino/uso terapêutico , Regulação para Baixo , Humanos , Masculino , Mitoxantrona/uso terapêutico , Oligonucleotídeos Antissenso/genética , Paclitaxel/uso terapêutico , Neoplasias da Próstata/terapia , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA