Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
IUBMB Life ; 75(1): 40-54, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35499745

RESUMO

The hypoxic tumour microenvironment (hTME), arising from inadequate and chaotic vascularity, can present a major obstacle for the treatment of solid tumours. Hypoxic tumour cells compromise responses to treatment since they can generate resistance to radiotherapy, chemotherapy and immunotherapy. The hTME impairs the delivery of a range of anti-cancer drugs, creates routes for metastasis and exerts selection pressures for aggressive phenotypes; these changes potentially occur within an immunosuppressed environment. Therapeutic strategies aimed at the hTME include targeting the molecular changes associated with hypoxia. An alternative approach is to exploit the prevailing lack of oxygen as a principle for the selective activation of prodrugs to target cellular components within the hTME. This review focuses on the design concepts and rationale for the use of unidirectional Hypoxia-Activated Prodrugs (uHAPs) to target the hTME as exemplified by the uHAPs AQ4N and OCT1002. These agents undergo irreversible reduction in a hypoxic environment to active forms that target DNA topoisomerase IIα (TOP2A). This nuclear enzyme is essential for cell division and is a recognised chemotherapeutic target. An activated uHAP interacts with the enzyme-DNA complex to induce DNA damage, cell cycle arrest and tumour cell death. uHAPs are designed to overcome the shortcomings of conventional HAPs and offer unique pharmacodynamic properties for effective targeting of TOP2A in the hTME. uHAP therapy in combination with standard of care treatments has the potential to enhance outcomes by co-addressing the therapeutic challenge presented by the hTME.


Assuntos
Neoplasias , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Microambiente Tumoral , Hipóxia Celular , Neoplasias/tratamento farmacológico , Neoplasias/genética , Hipóxia/tratamento farmacológico , DNA Topoisomerases/farmacologia
2.
Prostate ; 77(15): 1539-1547, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28944496

RESUMO

BACKGROUND: OCT1002 is a unidirectional hypoxia-activated prodrug (uHAP) OCT1002 that can target hypoxic tumor cells. Hypoxia is a common feature in prostate tumors and is known to drive disease progression and metastasis. It is, therefore, a rational therapeutic strategy to directly target hypoxic tumor cells in an attempt to improve treatment for this disease. Here we tested OCT1002 alone and in combination with standard-of-care agents in hypoxic models of castrate-resistant prostate cancer (CRPC). METHODS: The effect of OCT1002 on tumor growth and vasculature was measured using murine PC3 xenograft and dorsal skin fold (DSF) window chamber models. The effects of abiraterone, docetaxel, and cabazitaxel, both singly and in combination with OCT1002, were also compared. RESULTS: The hypoxia-targeting ability of OCT1002 effectively controls PC3 tumor growth. The effect was evident for at least 42 days after exposure to a single dose (30 mg/kg) and was comparable to, or better than, drugs currently used in the clinic. In DSF experiments OCT1002 caused vascular collapse in the PC3 tumors and inhibited the revascularization seen in controls. In this model OCT1002 also enhanced the anti-tumor effects of abiraterone, cabazitaxel, and docetaxel; an effect which was accompanied by a more prolonged reduction in tumor vasculature density. CONCLUSIONS: These studies provide the first evidence that OCT1002 can be an effective agent in treating hypoxic, castrate-resistant prostate tumors, either singly or in combination with established chemotherapeutics for prostate cancer.


Assuntos
Antraquinonas/farmacologia , Etilenodiaminas/farmacologia , Pró-Fármacos/farmacologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Animais , Antraquinonas/farmacocinética , Processos de Crescimento Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Etilenodiaminas/farmacocinética , Humanos , Masculino , Camundongos , Camundongos Nus , Pró-Fármacos/farmacocinética , Neoplasias de Próstata Resistentes à Castração/irrigação sanguínea , Neoplasias de Próstata Resistentes à Castração/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Clin Cancer Res ; 23(7): 1797-1808, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27697998

RESUMO

Purpose: To understand the role of hypoxia in prostate tumor progression and to evaluate the ability of the novel unidirectional hypoxia-activated prodrug OCT1002 to enhance the antitumor effect of bicalutamide.Experimental Design: The effect of OCT1002 on prostate cancer cells (LNCaP, 22Rv1, and PC3) was measured in normoxia and hypoxia in vitroIn vivo, tumor growth and lung metastases were measured in mice treated with bicalutamide, OCT1002, or a combination. Dorsal skin fold chambers were used to image tumor vasculature in vivo Longitudinal gene expression changes in tumors were analyzed using PCR.Results: Reduction of OCT1002 to its active form (OCT1001) decreased prostate cancer cell viability. In LNCaP-luc spheroids, OCT1002 caused increased apoptosis and decreased clonogenicity. In vivo, treatment with OCT1002 alone, or with bicalutamide, showed significantly greater tumor growth control and reduced lung metastases compared with controls. Reestablishment of the tumor microvasculature following bicalutamide-induced vascular collapse is inhibited by OCT1002. Significantly, the upregulation of RUNX2 and its targets caused by bicalutamide alone was blocked by OCT1002.Conclusions: OCT1002 selectively targets hypoxic tumor cells and enhances the antitumor efficacy of bicalutamide. Furthermore, bicalutamide caused changes in gene expression, which indicated progression to a more malignant genotype; OCT1002 blocked these effects, emphasizing that more attention should be attached to understanding genetic changes that may occur during treatment. Early targeting of hypoxic cells with OCT1002 can provide a means of inhibiting prostate tumor growth and malignant progression. This is of importance for the design and refinement of existing androgen-deprivation regimens in the clinic. Clin Cancer Res; 23(7); 1797-808. ©2016 AACR.


Assuntos
Antraquinonas/administração & dosagem , Etilenodiaminas/administração & dosagem , Proteínas de Neoplasias/genética , Pró-Fármacos/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Anilidas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Nitrilas/administração & dosagem , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Compostos de Tosil/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Gene ; 577(2): 109-18, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26657039

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neural disorder that causes death of the motor neurons in the brain and spinal cord; this affects the voluntary muscles and gradually leads to paralysis of the whole body. Most ALS cases are sporadic, though about 5-10% are familial. ALS is caused by multiple factors including mutation in any one of a number of specific genes, one of the most frequently affected is superoxide dismutase (SOD) 1. Alterations in SOD 1 have been linked with several variants of familial ALS. SOD 1 is a powerful antioxidant enzyme that protects cells from the damaging effects of superoxide radicals. The enzyme binds both copper and zinc ions that are directly involved in the deactivation of toxic superoxide radicals. Mutated SOD1 gene can acquire both gain and loss of function mutations. The most commonly identified mutations in SOD1 that affect protein activity are D90A, A4V and G93A. Deleterious mutations have been shown to modify SOD1 activity, which leads to the accumulation of highly toxic hydroxyl radicals. Accumulation of these free radicals causes degradation of both nuclear and mitochondrial DNA and protein misfolding, features which can be used as pathological indicators associated with ALS. Numerous clinical trials have been carried out over last few years with limited success. In some patients advanced techniques like gene and stem cell therapy have been trialed. However no definitive treatment option can provide a cure and currently ALS is managed by drugs and other supportive therapies. Consequently there is a need to identify new approaches for treatment of this ultimately fatal disease.


Assuntos
Esclerose Lateral Amiotrófica/genética , Mutação , Superóxido Dismutase/genética , Sequência de Aminoácidos , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/terapia , Animais , Terapia Genética , Humanos , Dados de Sequência Molecular , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/química , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
5.
J Cell Physiol ; 231(2): 473-82, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26189652

RESUMO

Aberrant expression of the transcription factor RUNX2 in prostate cancer has a number of important consequences including increased resistance to apoptosis, invasion and metastasis to bone. We previously demonstrated that hypoxia up-regulated RUNX2 in tumour cells, which in turn up-regulated the anti-apoptotic factor Bcl-2. Here, we investigate the impact of nitric oxide (NO) on RUNX2 and Bcl-2 expression in prostate cancer and further, how RUNX2 over-expression can impact tumour growth, angiogenesis and oxygenation in vivo. The effect of NO levels on RUNX2 and thus Bcl-2 expression was examined in prostate cancer cells in vitro using methods including gene and protein expression analyses, nitrite quantitation, protein-DNA interaction assays (ChIP) and viability assays (XTT). The effect of RUNX2 over-expression on tumour physiology (growth, oxygenation and angiogenesis) was also assessed in vivo using LNCaP xenografts. A low (but not high) concentration of NO (10 µM) induced expression of RUNX2 and Bcl-2, conferring resistance to docetaxel. These effects were induced via the ERK and PI3K pathways and were dependent on intact AP-1 binding sites in the RUNX2 promoter. RUNX2 over-expression in LNCaP tumours in vivo decreased the time to tumour presentation and increased tumour growth. Moreover, these tumours exhibited improved tumour angiogenesis and oxygenation. Low levels of NO increase expression of RUNX2 and Bcl-2 in LNCaP prostate tumour cells, and in vivo up-regulation of RUNX2 created tumours with a more malignant phenotype. Collectively, our data reveals the importance of NO-regulation of key factors in prostate cancer disease progression.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Óxido Nítrico/metabolismo , Neoplasias da Próstata/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Subunidade alfa 1 de Fator de Ligação ao Core/antagonistas & inibidores , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , RNA Interferente Pequeno/genética , Regulação para Cima
6.
Br J Radiol ; 88(1056): 20150405, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26462717

RESUMO

Most radiotherapy (RT) involves the use of high doses (>50 Gy) to treat malignant disease. However, low to intermediate doses (approximately 3-50 Gy) can provide effective control of a number of benign conditions, ranging from inflammatory/proliferative disorders (e.g. Dupuytren's disease, heterotopic ossification, keloid scarring, pigmented villonodular synovitis) to benign tumours (e.g. glomus tumours or juvenile nasopharyngeal angiofibromas). Current use in UK RT departments is very variable. This review identifies those benign diseases for which RT provides good control of symptoms with, for the most part, minimal side effects. However, exposure to radiation has the potential to cause a radiation-induced cancer (RIC) many years after treatment. The evidence for the magnitude of this risk comes from many disparate sources and is constrained by the small number of long-term studies in relevant clinical cohorts. This review considers the types of evidence available, i.e. theoretical models, phantom studies, epidemiological studies, long-term follow-up of cancer patients and those treated for benign disease, although many of the latter data pertain to treatments that are no longer used. Informative studies are summarized and considered in relation to the potential for development of a RIC in a range of key tissues (skin, brain etc.). Overall, the evidence suggests that the risks of cancer following RT for benign disease for currently advised protocols are small, especially in older patients. However, the balance of risk vs benefit needs to be considered in younger adults and especially if RT is being considered in adolescents or children.


Assuntos
Neoplasias Induzidas por Radiação/etiologia , Dosagem Radioterapêutica , Humanos , Risco
7.
Int J Cancer ; 134(5): 1102-11, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23959905

RESUMO

Bladder cancer patients suffer significant treatment failure, including high rates of recurrence and poor outcomes for advanced disease. If mechanisms to improve tumour cell treatment sensitivity could be identified and/or if tumour response could be predicted, it should be possible to improve local-control and survival. Previously, we have shown that radiation-induced DNA damage, measured by alkaline Comet assay (ACA), correlates bladder cancer cell radiosensitivity in vitro. In this study we first show that modified-ACA measures of cisplatin and mitomycin-C-induced damage also correlate bladder cancer cell chemosensitivity in vitro, with essentially the same rank order for chemosensitivity as for radiosensitivity. Furthermore, ACA studies of radiation-induced damage in different cell-DNA substrates (nuclei, nucleoids and intact parent cells) suggest that it is a feature retained in the prepared nucleoids that is responsible for the relative damage sensitivity of bladder cancer cells, suggestive of differences in the organisation of DNA within resistant vs. sensitive cells. Second, we show that ACA analysis of biopsies from bladder tumours reveal that reduced DNA damage sensitivity associates with poorer treatment outcomes, notably that tumours with a reduced damage response show a significant association with local recurrence of non-invasive disease and that reduced damage response was a better predictor of recurrence than the presence of high-risk histology in this cohort. In conclusion, this study demonstrates that mechanisms governing treatment-induced DNA damage are both central to and predictive of bladder cancer cell treatment sensitivity and exemplifies a link between DNA damage resistance and both treatment response and tumour aggression.


Assuntos
Ensaio Cometa/métodos , Dano ao DNA , Neoplasias da Bexiga Urinária/tratamento farmacológico , Linhagem Celular Tumoral , Cisplatino/farmacologia , Humanos , Mitomicina/farmacologia , Resultado do Tratamento , Neoplasias da Bexiga Urinária/genética
8.
Int J Cancer ; 132(6): 1323-32, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22915157

RESUMO

Androgen withdrawal induces hypoxia in androgen-sensitive tissue; this is important as in the tumour microenvironment, hypoxia is known to drive malignant progression. Our study examined the time-dependent effect of androgen deprivation therapy (ADT) on tumour oxygenation and investigated the role of ADT-induced hypoxia on malignant progression in prostate tumours. LNCaP xenografted tumours were treated with anti-androgens and tumour oxygenation measured. Dorsal skin fold (DSF) chambers were used to image tumour vasculature in vivo. Quantitative PCR (QPCR) identified differential gene expression following treatment with bicalutamide. Bicalutamide-treated and vehicle-only-treated tumours were re-established in vitro, and invasion and sensitivity to docetaxel were measured. Tumour growth delay was calculated following treatment with bicalutamide combined with the bioreductive drug AQ4N. Tumour oxygenation measurements showed a precipitate decrease following initiation of ADT. A clinically relevant dose of bicalutamide (2 mg/kg/day) decreased tumour oxygenation by 45% within 24 hr, reaching a nadir of 0.09% oxygen (0.67 ± 0.06 mmHg) by Day 7; this persisted until Day 14 when it increased up to Day 28. Using DSF chambers, LNCaP tumours treated with bicalutamide showed loss of small vessels at Days 7 and 14 with revascularisation occurring by Day 21. QPCR showed changes in gene expression consistent with the vascular changes and malignant progression. Cells from bicalutamide-treated tumours were more malignant than vehicle-treated controls. Combining bicalutamide with AQ4N (50 mg/kg, single dose) caused greater tumour growth delay than bicalutamide alone. Our study shows that bicalutamide-induced hypoxia selects for cells that show malignant progression; targeting hypoxic cells may provide greater clinical benefit.


Assuntos
Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Antraquinonas/administração & dosagem , Hipóxia Celular , Nitrilas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Compostos de Tosil/farmacologia , Animais , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/metabolismo , Transdução de Sinais , Fatores de Tempo
9.
PLoS One ; 7(11): e49364, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23145163

RESUMO

The alkaline single cell gel electrophoresis (comet) assay can be combined with fluorescent in situ hybridisation (FISH) methodology in order to investigate the localisation of specific gene domains within an individual cell. The number and position of the fluorescent signal(s) provides information about the relative damage and subsequent repair that is occurring in the targeted gene domain(s). In this study, we have optimised the comet-FISH assay to detect and compare DNA damage and repair in the p53 and hTERT gene regions of bladder cancer cell-lines RT4 and RT112, normal fibroblasts and Cockayne Syndrome (CS) fibroblasts following γ-radiation. Cells were exposed to 5Gy γ-radiation and repair followed for up to 60 minutes. At each repair time-point, the number and location of p53 and hTERT hybridisation spots was recorded in addition to standard comet measurements. In bladder cancer cell-lines and normal fibroblasts, the p53 gene region was found to be rapidly repaired relative to the hTERT gene region and the overall genome, a phenomenon that appeared to be independent of hTERT transcriptional activity. However, in the CS fibroblasts, which are defective in transcription coupled repair (TCR), this rapid repair of the p53 gene region was not observed when compared to both the hTERT gene region and the overall genome, proving the assay can detect variations in DNA repair in the same gene. In conclusion, we propose that the comet-FISH assay is a sensitive and rapid method for detecting differences in DNA damage and repair between different gene regions in individual cells in response to radiation. We suggest this increases its potential for measuring radiosensitivity in cells and may therefore have value in a clinical setting.


Assuntos
Dano ao DNA , Reparo do DNA/efeitos da radiação , Genes p53 , Telomerase/genética , Linhagem Celular , Ensaio Cometa , Humanos , Hibridização in Situ Fluorescente , Radiação Ionizante
10.
Mol Carcinog ; 49(2): 190-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19908239

RESUMO

CYP1B1 mRNA is expressed constitutively in all normal extrahepatic human tissues, though the protein is usually undetectable. In contrast, CYP1B1 protein is expressed at high levels in tumors. In this study CYP1B1 mRNA and protein expression was measured in a panel of cell lines indicating that CYP1B1 regulation is altered in tumor cell lines in vitro. Interrogation of ONCOMINE revealed that CYP1B1 mRNA is not significantly overexpressed in tumors compared to normal tissues, suggesting CYP1B1 is subject to posttranscriptional control. Analysis of the CYP1B1 mRNA revealed a complex 5' untranslated region (UTR) containing a small upstream open-reading frame (uORF). These features are present in mRNAs subject to translational control so the effect of the 5'UTR was tested using in vitro translation in CHO-K1 cells. The 5'UTR significantly inhibited luciferase reporter gene translation, and mutation of the uORF start codon abolished the inhibitory effect. The 5'UTR also interacted with the microRNA-27b recognition element in the CYP1B1 mRNA 3'UTR to almost completely inhibit translation. CYP1B1 is subject to a high degree of translational control, which may explain the absence of protein expression in normal cells. Alterations in translational control during malignant transformation may help to explain the tumor-specific expression of CYP1B1 protein.


Assuntos
Regiões 5' não Traduzidas , Sistema Enzimático do Citocromo P-450/genética , Biossíntese de Proteínas , Proteínas/metabolismo , RNA Mensageiro/genética , Animais , Hidrocarboneto de Aril Hidroxilases , Sequência de Bases , Western Blotting , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Citocromo P-450 CYP1B1 , Primers do DNA , Humanos , Fases de Leitura Aberta , Reação em Cadeia da Polimerase , Homologia de Sequência do Ácido Nucleico
11.
Mol Carcinog ; 48(2): 110-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18618592

RESUMO

Cytochrome P450 1B1 (CYP1B1) mRNA is constitutively expressed in most normal extra-hepatic tissues; however the protein is not detectable in these tissues but is expressed in a wide variety of tumors. CYP1B1 is responsible for the activation of a number of carcinogens present in tobacco smoke and food. A surgical model of rat esophageal tumorigenesis, promoted by gastric or duodenal reflux was used to determine CYP1B1 expression in premalignant esophageal tissue. Immunohistochemistry was performed using a modified amplified fluorescein tyramide protocol. CYP1B1 was not observed in normal esophageal mucosa, submucosa, or muscularis mucosa. Animals exposed to gastric reflux developed mild hyperplasia. Varying degrees of hyperplasia were observed in the duodenal reflux group. All regions of hyperplasia showed moderate or strong CYP1B1 immunoreactivity. Duodenal reflux induced a small number of premalignant changes: immunoreactivity was absent from the epithelium of squamous dysplasia (0/10), Barrett's esophagus (0/7), and majority of dysplastic Barrett's esophagus (1/4). Moderate or strong immunoreactivity was observed in the majority (7/8) of squamous cell carcinomas (SCCs) in situ. Immunoreactivity was also observed in the lamina propria and submucosa in association with inflammation, regardless of the severity of inflammation. The expression of CYP1B1 in hyperplasia, SCCs in situ, or in association with inflammation may increase the production of carcinogenic metabolites, which may promote esophageal tumorigenesis.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Refluxo Duodenogástrico/complicações , Neoplasias Esofágicas/enzimologia , Refluxo Gastroesofágico/complicações , Animais , Especificidade de Anticorpos , Hidrocarboneto de Aril Hidroxilases/imunologia , Western Blotting , Citocromo P-450 CYP1B1 , Refluxo Duodenogástrico/enzimologia , Neoplasias Esofágicas/etiologia , Feminino , Refluxo Gastroesofágico/enzimologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
12.
Oncol Res ; 17(3): 93-101, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18669161

RESUMO

The contribution of endothelial cell growth to angiogenesis has been widely studied; however, the involvement of pericytes is less well documented, especially in human tumors. In this study we aimed to quantify and assess the prognostic significance of pericyte coverage, the extent of hypoxia, and microvessel density (MVD) in normal bladder mucosa and urothelial carcinoma. Antibody to alpha-smooth muscle actin was used to assess the distribution of pericytes (mural/smooth muscle cells) in the microvessels of normal human bladder (n = 4) mucosa and in urothelial carcinoma (n = 47) samples; this was quantitated using microvessel pericyte index (MPI). The MVD was measured using two different methods (n = 47) and hypoxia was assessed using glucose transporter-1 (Glut-1) staining (n = 30). There was a 70% reduction in MPI in urothelial carcinomas compared to normal bladder mucosa (p < 0.0012); MPI did not correlate with tumor stage or grade. Ta and T1 superficial tumors were divided into two groups with a MPI of <15% or >15%. Progression-free survival was significantly shorter for tumors with MPI >15% (p = 0.0036). MVD had no prognostic value using either evaluation method. Glut-1 immunoreactivity was not prognostic in superficial urothelial carcinoma samples. Tumors with a higher MPI showed a greater Glut-1 immunoreactivity (p = 0.0051). Microvessels in urothelial carcinoma have a considerable loss of pericyte coverage compared to normal bladder mucosa. The data from this preliminary study indicate that progression-free survival was shorter in patients whose superficial tumors had higher pericyte coverage of the microvessels. This may be due to increased levels of hypoxia, as demonstrated by a significant increase in Glut-1 staining.


Assuntos
Carcinoma/patologia , Neovascularização Patológica/patologia , Pericitos/patologia , Neoplasias da Bexiga Urinária/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Permeabilidade Capilar , Carcinoma/irrigação sanguínea , Carcinoma/fisiopatologia , Contagem de Células , Feminino , Transportador de Glucose Tipo 1/análise , Humanos , Hipóxia/etiologia , Hipóxia/patologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/irrigação sanguínea , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/fisiopatologia , Estadiamento de Neoplasias , Neovascularização Patológica/fisiopatologia , Prognóstico , Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/fisiopatologia
13.
Int J Cancer ; 123(4): 760-8, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18512241

RESUMO

Hypoxia confers resistance to common cancer therapies, however, it has also has been shown to result in genetic alterations which may allow a survival advantage and increase the tumorigenic properties of cancer cells. Additionally, it may exert a selection pressure, allowing expansion of tumor cells with a more aggressive phenotype. To further assess the role of hypoxia in malignant progression in prostate cancer we exposed human androgen dependent prostate cancer cells (LNCaP) to cycles of chronic hypoxia and isolated a subline, LNCaP-H1. This article describes the partial characterization of this cell line. The LNCaP-H1 subline showed altered growth characteristics and exhibited androgen independent growth both in vitro and in vivo. Furthermore, these cells were resistant to mitochondrial-mediated apoptosis, probably since the endogenous levels of Bax was lower and Bcl-2 higher than in the parental LNCaP cells. Microarray analysis revealed that a complex array of pathways had differential gene expression between the 2 cell lines, with LNCaP-H1 cells exhibiting a genetic profile which suggests that they may be more likely metastasize to distant organs, especially bone. This was supported by an in vitro invasion assay, and an in vivo metastasis study. This study shows that hypoxia can select for androgen independent prostate cancer cells which have a survival advantage and are more likely to invade and metastasize.


Assuntos
Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Androgênios/metabolismo , Animais , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Expressão Gênica , Genótipo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Mitocôndrias/fisiologia , Transplante de Neoplasias , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Fenótipo , Neoplasias da Próstata/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Androgênicos/biossíntese , Receptores Androgênicos/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transplante Heterólogo
14.
Clin Cancer Res ; 14(5): 1502-9, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18316575

RESUMO

PURPOSE: A number of cytotoxic chemotherapy agents tested at low concentrations show antiangiogenic properties with limited cytotoxicity, e.g., cyclophosphamide, tirapazamine, and mitoxantrone. AQ4N is a bioreductive alkylaminoanthraquinone that is cytotoxic when reduced to AQ4; hence, it can be used to target hypoxic tumor cells. AQ4N is structurally similar to mitoxantrone and was evaluated for antiangiogenic properties without the need for bioreduction. EXPERIMENTAL DESIGN: The effect of AQ4N and fumagillin on human microvascular endothelial cells (HMEC-1) was measured using a variety of in vitro assays, i.e., 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, wound scrape, tubule formation, rat aortic ring, and invasion assays. Low-dose AQ4N (20 mg/kg) was also given in vivo to mice bearing a tumor in a dorsal skin flap. RESULTS: AQ4N (10(-11) to 10(-5) mol/L) had no effect on HMEC-1 viability. AQ4N (10(-9) to 10(-5)mol/L) caused a sigmoidal dose-dependent inhibition of endothelial cell migration in the wound scrape model. Fumagillin showed a similar response over a lower dose range (10(-13) to 10(-9) mol/L); however, the maximal inhibition was less (25% versus 43% for AQ4N). AQ4N inhibited HMEC-1 cell contacts on Matrigel (10(-8) to 10(-5) mol/L), HMEC-1 cell invasion, and sprouting in rat aorta explants. Immunofluorescence staining with tubulin, vimentim, dynein, and phalloidin revealed that AQ4N caused disruption to the cell cytoskeleton. When AQ4N (20 mg/kg) was given in vivo for 5 days, microvessels disappeared in LNCaP tumors grown in a dorsal skin flap. CONCLUSIONS: This combination of assays has shown that AQ4N possesses antiangiogenic effects in normoxic conditions, which could potentially contribute to antitumor activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Antraquinonas/farmacologia , Endotélio Vascular/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Pele/irrigação sanguínea , Animais , Antineoplásicos/farmacologia , Aorta/efeitos dos fármacos , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Neoplasias do Colo/tratamento farmacológico , Cicloexanos/farmacologia , Citoesqueleto/metabolismo , Combinação de Medicamentos , Ácidos Graxos Insaturados/farmacologia , Humanos , Laminina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microcirculação , Neovascularização Fisiológica , Neoplasias da Próstata/tratamento farmacológico , Proteoglicanas/metabolismo , Ratos , Ratos Wistar , Sesquiterpenos/farmacologia , Retalhos Cirúrgicos , Cicatrização/efeitos dos fármacos
15.
Mutagenesis ; 23(3): 183-90, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18256034

RESUMO

The comet assay has been widely used to measure a range of cellular responses to DNA damage and has found applications in genotoxicity studies, bio-monitoring, ecological testing and in the study of human disease. This review discusses how the comet assay has been applied to the study of DNA damage and repair associated with cancer. The potential of the assay as a tool for predicting an individual's tumour sensitivity to radiation and to various chemotherapeutic drugs is examined, as well as outlining the usefulness of the assay in assessing oxidative stress within tumours. In addition, we review the use of the comet assay in investigations of the DNA-damaging effect of anti-neoplastic drugs and radiation used during cancer therapy. The advantages and limitations of the comet assay in carrying out all these studies are outlined, and the suitability of the comet assay for use in the clinical management of cancer is discussed.


Assuntos
Ensaio Cometa , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Neoplasias/terapia , Tolerância a Radiação , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Prognóstico , Resultado do Tratamento
16.
Eur Urol ; 50(2): 290-301, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16455181

RESUMO

OBJECTIVES: Clinical studies have shown that more than 70% of primary bladder tumours arise in the area around the ureteric orifice. In this study a genomic approach was taken to explore the molecular mechanisms that may influence this phenomenon. METHODS: RNA was isolated from each individual normal ureteric orifice and the dome biopsy from 33 male patients. Equal amounts of the pooled ureteric orifice and dome mRNAs were labelled with Cy3 and Cy5, respectively before hybridising to the gene chip (UniGEM 2.0, Incyte Genomics Inc., Wilmington, Delaware, USA). RESULTS: Significant changes (more than a twofold difference) in gene expression were observed in 3.1% (312) of the 10,176 gene array: 211 genes upregulated and 101 downregulated. Analysis of Cdc25B, TK1, PKM, and PDGFra with RT-PCR supported the reliability of the microarray result. Seladin-1 was the most upregulated gene in the ureteric orifice: 8.3-fold on the microarray and 11.4-fold by real time PCR. CONCLUSIONS: Overall, this study suggests significant altered gene expression between these two anatomically distinct areas of the normal human bladder. Of particular note is Seladin-1, whose significance in cancer is yet to be clarified. Further studies of the genes discovered by this work will help clarify which of these differences influence primary bladder carcinogenesis.


Assuntos
Perfilação da Expressão Gênica , Expressão Gênica , Urotélio , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Regulação Neoplásica da Expressão Gênica , Biblioteca Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Neoplásico/análise , RNA Neoplásico/genética , Neoplasias da Bexiga Urinária/genética
17.
Cardiovasc Res ; 69(1): 207-17, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16165110

RESUMO

OBJECTIVE: Pericytes, located in close proximity to the underlying endothelium, form an integral component of the microvasculature. These cells are intimately involved in angiogenesis, which is of fundamental importance in many physiological and pathological processes. We evaluated the influence of pericyte-conditioned medium (PCM) on endothelial cell growth characteristics and modulation of endothelial gene expression. METHODS: Migration and tubule formation assays were performed in vitro to determine the effect of PCM on endothelial growth characteristics. cDNA microarray analysis was used to identify alterations in gene expression following exposure of human microvascular endothelial cells (HMEC-1) to PCM. Overexpression of PAI-1 using recombinant protein or transient transfection, and inhibition using an inhibitory antibody against PAI-1, were used to determine whether up- or down-regulation of this gene was responsible for the changes in endothelial cell characteristics observed in response to PCM exposure. RESULTS: We have shown that PCM exerts a dramatic inhibitory influence on endothelial cell migration in vitro. In addition, endothelial cells cultured on Matrigel and exposed to PCM were found to generate significantly fewer angiogenic branches. Microarray analysis of endothelial cells exposed to PCM identified PAI-1 as the gene showing the greatest level of differential expression (3.4-fold induction). Studies using an inhibitory antibody to PAI-1 suggest that induction of this protein by PCM is pivotal to the observed inhibitory influence on the migratory and angiogenic potential of HMEC-1. We further investigated this by overexpressing PAI-1, which was shown to have a potent inhibitory influence on EC migration and angiogenic branching, although the concentration of PAI-1 was clearly important. CONCLUSION: Collectively, these findings suggest that PCM contains a bioactive element(s) that controls both endothelial cell migration and tubule formation in vitro and that these responses may be partially controlled by increased endothelial cell expression of PAI-1.


Assuntos
Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Neovascularização Fisiológica , Pericitos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Northern Blotting/métodos , Western Blotting/métodos , Movimento Celular , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Inibidor 1 de Ativador de Plasminogênio/metabolismo
18.
J Gene Med ; 7(7): 851-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15712360

RESUMO

BACKGROUND: AQ4N is metabolised in hypoxic cells by cytochrome P450s (CYPs) to the cytotoxin AQ4. Most solid tumours are known to contain regions of hypoxia whereas levels of CYPs have been found to vary considerably. Enhancement of CYP levels may be obtained using gene-directed enzyme prodrug therapy (GDEPT). We have therefore examined the potential of a CYP2B6-mediated GDEPT strategy to enhance the anti-tumour effect of the combination of AQ4N with radiation or cyclophosphamide (CPA). METHODS: In vitro and in vivo transient transfection of human CYP2B6 +/- CYP reductase (CYPRED) was investigated in RIF-1 mouse tumours. Efficacy in vitro was assessed using the alkaline comet assay (ACA). In vivo, the time to reach 4x the treatment volume (quadrupling time; VQT) was used as the end point. RESULTS: When CYP2B6 was transfected into RIF-1 cells and treated with AQ4N under hypoxic conditions there was a significant increase in DNA damage (measured by the ACA) compared with non-transfected cells. In vivo, a single intra-tumoural injection of a CYP2B6 vector construct significantly enhanced tumour growth delay in combination with AQ4N (100 mg/kg) and 10 Gy X-rays. AQ4N (100 mg/kg) and CPA (100 mg/kg) with CYP2B6 and CYPRED also enhanced tumour growth delay; this effect became significant when the schedule was repeated 14 days later (p = 0.0197). CONCLUSIONS: The results show the efficacy of a CYP2B6-mediated GDEPT strategy for bioreduction of AQ4N; this may offer an additional approach to target radiation- and chemo-resistant hypoxic tumours that should enhance overall tumour control.


Assuntos
Terapia Genética/métodos , Pró-Fármacos/metabolismo , Animais , Antraquinonas/metabolismo , Antraquinonas/farmacologia , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Hidrocarboneto de Aril Hidroxilases/farmacologia , Hipóxia Celular/efeitos dos fármacos , Terapia Combinada , Ciclofosfamida/administração & dosagem , Ciclofosfamida/uso terapêutico , Citocromo P-450 CYP2B6 , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , DNA de Neoplasias/efeitos dos fármacos , Fibrossarcoma/terapia , Humanos , Camundongos , Camundongos Endogâmicos C3H , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Oxirredutases N-Desmetilantes , Pró-Fármacos/farmacologia , Radioterapia , Proteínas Recombinantes/análise , Transfecção , Transgenes , Células Tumorais Cultivadas
19.
Curr Opin Mol Ther ; 6(4): 421-35, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15468601

RESUMO

Gene-directed enzyme prodrug therapy involves tumor-specific delivery of a gene encoding a drug-metabolizing enzyme allowing metabolism of a non-toxic prodrug to a toxic species directly within tumor cells. This review covers the wide range of enzyme-prodrug combinations currently under preclinical and clinical investigation. Issues of tumor specificity and enhanced cytotoxicity through bystander effects will be discussed.


Assuntos
Enzimas/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Neoplasias/terapia , Pró-Fármacos/metabolismo , Animais , Efeito Espectador , Ensaios Clínicos como Assunto , Humanos
20.
J Natl Cancer Inst ; 95(24): 1859-68, 2003 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-14679155

RESUMO

BACKGROUND: Cell cycle checkpoints function to maintain genetic stability by providing additional time for repair of DNA damage and completion of events that are necessary for accurate cell division. Some checkpoints, such as the DNA damage G1 checkpoint, are dependent on p53, whereas other checkpoints, such as the decatenation G(2) checkpoint, are not. Because bladder transitional cell carcinomas (TCCs) often contain numerous chromosomal aberrations and appear to have highly unstable genomes, we analyzed cell cycle checkpoint functions in a panel of TCC lines. METHODS: Cell cycle arrest was induced in normal human fibroblasts (NHF1-hTERT) and normal human uroepithelial cells (HUCs), and TCC lines and checkpoint functions were quantified using flow cytometry and fluorescence microscopy. The inducers and checkpoints were ionizing radiation (i.e., DNA damage) (G1 and G2 checkpoints), the mitotic inhibitor colcemid (polyploidy checkpoint), or the topoisomerase II catalytic inhibitor ICRF-193 (decatenation G2 checkpoint). Four of the five TCC lines expressed mutant p53. RESULTS: HUCs had an effective G1 checkpoint response to ionizing radiation, with 68% of cells inhibited from moving from G1 into S phase. By contrast, G1 checkpoint function was severely attenuated (<15% inhibition) in three of the five TCC lines and moderately attenuated (<50% inhibition) in the other two lines. NHF1-hTERT had an effective polyploidy checkpoint response, but three of five TCC lines were defective in this checkpoint. HUCs had effective ionizing radiation and decatenation G2 checkpoint responses. All TCC lines had a relatively effective G2 checkpoint response to DNA damage, although the responses of two of the TCC lines were moderately attenuated relative to HUCs. All TCC lines had a severe defect in the decatenation G2 checkpoint response. CONCLUSION: Bladder TCC lines have defective cell cycle checkpoint functions, suggesting that the p53-independent decatenation G2 checkpoint may cooperate with the p53-dependent G1 checkpoints to preserve chromosomal stability and suppress bladder carcinogenesis.


Assuntos
Carcinoma de Células de Transição/fisiopatologia , Ciclo Celular , Dano ao DNA , Reparo do DNA , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Bexiga Urinária/fisiopatologia , Carcinoma de Células de Transição/genética , Linhagem Celular Tumoral , Fase G1 , Fase G2 , Genes Supressores de Tumor , Humanos , Ploidias , Neoplasias da Bexiga Urinária/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...