Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Eur J Pharm Biopharm ; 117: 400-407, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28478160

ABSTRACT

We address, for the first time, the impact of skin insertion on multiple occasions of polymeric microneedle arrays in an animal model in vivo. Dissolving microneedle arrays prepared from aqueous blends of 20% w/w Gantrez® S-97 BF and 40% w/w poly(vinyl pyrrolidone) 58kDa and hydrogel-forming microneedle arrays prepared from aqueous blends of and poly(ethyleneglycol) 10kDa were repeatedly applied to the skin of hairless mice in vivo. Skin appearance and skin barrier function, as illustrated by measurement of transepidermal water loss, were not measurably altered during the entire study period. Biomarkers of infection, immunity and inflammation/irritation were also statistically unchanged, regardless of the microneedle formulation, needle density or number of applications. Mice remained healthy throughout and continued to gain weight during the study. For example, transepidermal water loss values were typically in the range 10-15gm-2h-1 immediately prior to microneedle insertion and 15-25gm-2h-1 immediately following microneedle removal, regardless of when they were measured during the study periods. Serum biomarker levels, measured immediately post-mortem were always in the range 10-20µgml-1 for C-reactive protein, 0.5-1.5mgml-1 for Immunoglobulin G and 1000-2500pgml-1 for interleukin 1-ß and were never statistically different from untreated controls. No measurable levels of tumour necrosis factor-α were found in any animals. These findings are encouraging for the formulations investigated, suggesting that their repeated use by patients will not cause undesirable side-effects. By beginning to address potential regulatory questions at an early stage, the microneedles field will be ideally-placed to take advantage of the potential market. This work illustrates a potential pre-clinical strategy for development of regulatory dossiers on microneedle technologies.


Subject(s)
Immunity, Cellular/physiology , Microinjections/methods , Needles , Skin/immunology , Skin/metabolism , Water Loss, Insensible/physiology , Administration, Cutaneous , Animals , Biomarkers/blood , Female , Inflammation/blood , Inflammation/immunology , Male , Mice , Mice, Hairless , Microinjections/adverse effects , Needles/adverse effects
2.
Oncotarget ; 6(14): 12209-23, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25906750

ABSTRACT

FK506-binding protein-like (FKBPL) has established roles as an anti-tumor protein, with a therapeutic peptide based on this protein, ALM201, shortly entering phase I/II clinical trials. Here, we evaluated FKBPL's prognostic ability in primary breast cancer tissue, represented on tissue microarrays (TMA) from 3277 women recruited into five independent retrospective studies, using immunohistochemistry (IHC). In a meta-analysis, FKBPL levels were a significant predictor of BCSS; low FKBPL levels indicated poorer breast cancer specific survival (BCSS) (hazard ratio (HR) = 1.30, 95% confidence interval (CI) 1.14-1.49, p < 0.001). The prognostic impact of FKBPL remained significant after adjusting for other known prognostic factors (HR = 1.25, 95% CI 1.07-1.45, p = 0.004). For the sub-groups of 2365 estrogen receptor (ER) positive patients and 1649 tamoxifen treated patients, FKBPL was significantly associated with BCSS (HR = 1.34, 95% CI 1.13-1.58, p < 0.001, and HR = 1.25, 95% CI 1.04-1.49, p = 0.02, respectively). A univariate analysis revealed that FKBPL was also a significant predictor of relapse free interval (RFI) within the ER positive patient group, but it was only borderline significant within the smaller tamoxifen treated patient group (HR = 1.32 95% CI 1.05-1.65, p = 0.02 and HR = 1.23 95% CI 0.99-1.54, p = 0.06, respectively). The data suggests a role for FKBPL as a prognostic factor for BCSS, with the potential to be routinely evaluated within the clinic.


Subject(s)
Breast Neoplasms/genetics , Immunophilins/genetics , Immunophilins/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cohort Studies , Female , Humans , Precision Medicine , Prognosis , Survival Analysis , Tacrolimus Binding Proteins
3.
Thorax ; 70(5): 426-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25770093

ABSTRACT

INTRODUCTION: Secretory leucocyte protease inhibitor and elafin are members of the whey acidic protein (WAP), or WAP four disulfide-core (WFDC), family of proteins and have multiple contributions to innate defence including inhibition of neutrophil serine proteases and inhibition of the inflammatory response to lipopolysaccharide (LPS). This study aimed to explore potential activities of WFDC12, a previously uncharacterised WFDC protein expressed in the lung. METHODS: Recombinant expression and purification of WFDC12 were optimised in Escherichia coli. Antiprotease, antibacterial and immunomodulatory activities of recombinant WFDC12 were evaluated and levels of endogenous WFDC12 protein were characterised by immunostaining and ELISA. RESULTS: Recombinant WFDC12 inhibited cathepsin G, but not elastase or proteinase-3 activity. Monocytic cells pretreated with recombinant WFDC12 before LPS stimulation produced significantly lower levels of the pro-inflammatory cytokines interleukin-8 and monocyte chemotactic protein-1 compared with cells stimulated with LPS alone. Recombinant WFDC12 became conjugated to fibronectin in a transglutaminase-mediated reaction and retained antiprotease activity. In vivo WFDC12 expression was confirmed by immunostaining of human lung tissue sections. WFDC12 levels in human bronchoalveolar lavage fluid from healthy and lung-injured patients were quantitatively compared, showing WFDC12 to be elevated in both patients with acute respiratory distress syndrome and healthy subjects treated with LPS, relative to healthy controls. CONCLUSIONS: Together, these results suggest a role for this lesser known WFDC protein in the regulation of lung inflammation.


Subject(s)
Lung/metabolism , Monocytes/drug effects , Proteins/pharmacology , Serine Endopeptidases/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Humans , Lipopolysaccharides , Lung/pathology , Microbial Sensitivity Tests , Monocytes/metabolism , Proteins/metabolism , Recombinant Proteins/pharmacology , Tissue Culture Techniques
4.
Int J Cancer ; 133(9): 2102-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23629809

ABSTRACT

Recent murine studies have demonstrated that tumor-associated macrophages in the tumor microenvironment are a key source of the pro-tumorigenic cysteine protease, cathepsin S. We now show in a syngeneic colorectal carcinoma murine model that both tumor and tumor-associated cells contribute cathepsin S to promote neovascularization and tumor growth. Cathepsin S depleted and control colorectal MC38 tumor cell lines were propagated in both wild type C57Bl/6 and cathepsin S null mice to provide stratified depletion of the protease from either the tumor, tumor-associated host cells, or both. Parallel analysis of these conditions showed that deletion of cathepsin S inhibited tumor growth and development, and revealed a clear contribution of both tumor and tumor-associated cell derived cathepsin S. The most significant impact on tumor development was obtained when the protease was depleted from both sources. Further characterization revealed that the loss of cathepsin S led to impaired tumor vascularization, which was complemented by a reduction in proliferation and increased apoptosis, consistent with reduced tumor growth. Analysis of cell types showed that in addition to the tumor cells, tumor-associated macrophages and endothelial cells can produce cathepsin S within the microenvironment. Taken together, these findings clearly highlight a manner by which tumor-associated cells can positively contribute to developing tumors and highlight cathepsin S as a therapeutic target in cancer.


Subject(s)
Carcinoma, Lewis Lung/pathology , Cathepsins/physiology , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Melanoma, Experimental/pathology , Neovascularization, Pathologic , Animals , Apoptosis , Blotting, Western , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Cell Adhesion , Cell Cycle , Cells, Cultured , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Macrophages/cytology , Macrophages/metabolism , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Microenvironment
5.
Nat Commun ; 2: 555, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22109529

ABSTRACT

Metastases are the major cause of death from melanoma, a skin cancer that has the fastest rising incidence of any malignancy in the Western world. Molecular pathways that drive melanoblast migration in development are believed to underpin the movement and ultimately the metastasis of melanoma. Here we show that mice lacking P-Rex1, a Rac-specific Rho GTPase guanine nucleotide exchange factor, have a melanoblast migration defect during development evidenced by a white belly. Moreover, these P-Rex1(-/-) mice are resistant to metastasis when crossed to a murine model of melanoma. Mechanistically, this is associated with P-Rex1 driving invasion in a Rac-dependent manner. P-Rex1 is elevated in the majority of human melanoma cell lines and tumour tissue. We conclude that P-Rex1 has an important role in melanoblast migration and cancer progression to metastasis in mice and humans.


Subject(s)
Cell Movement/physiology , Guanine Nucleotide Exchange Factors/metabolism , Melanoma/metabolism , Melanoma/pathology , Neoplasm Metastasis/physiopathology , Animals , Cell Movement/genetics , Cells, Cultured , Guanine Nucleotide Exchange Factors/genetics , Humans , Immunohistochemistry , In Vitro Techniques , Melanoma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/genetics , Tissue Array Analysis
6.
Biochem Soc Trans ; 39(2): 663-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21428958

ABSTRACT

FKBPs (FK506-binding proteins) have long been recognized as key regulators of the response to immunosuppressant drugs and as co-chaperones of steroid receptor complexes. More recently, evidence has emerged suggesting that this diverse protein family may also represent cancer biomarkers owing to their roles in cancer progression and response to treatment. FKBPL (FKBP-like) is a novel FKBP with roles in GR (glucocorticoid receptor), AR (androgen receptor) and ER (oestrogen receptor) signalling. FKBPL binds Hsp90 (heat-shock protein 90) and modulates translocation, transcriptional activation and phosphorylation of these steroid receptors. It has been proposed as a novel prognostic and predictive biomarker, where high levels predict for increased recurrence-free survival in breast cancer patients and enhanced sensitivity to endocrine therapy. Since this protein family has roles in a plethora of signalling pathways, its members represent novel prognostic markers and therapeutic targets for cancer diagnosis and treatment.


Subject(s)
Biomarkers, Tumor/physiology , Immunophilins/physiology , Neoplasms/diagnosis , Tacrolimus Binding Proteins/physiology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Immunophilins/genetics , Immunophilins/metabolism , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction/physiology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
7.
Melanoma Res ; 20(5): 392-400, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20848733

ABSTRACT

DNA methylation plays a major role in cancer by silencing tumour suppressor genes. In melanoma, only a discrete number of methylated genes have been identified so far. After the treatment of melanoma cells with a DNA methyltransferase inhibitor and subsequent transcriptomic profiling, we had identified earlier a cohort of melanoma progression-associated genes regulated by methylation. Here, we identified which of these genes are directly methylated in melanoma cell lines and tissues. First, we examined 16 genes by bisulphite sequencing in the WM793 isogenic cell line model series. Five of these genes (CYBA, FABP5, MT1E, TSPY1 and TAC1) displayed increased methylation in several invasive cell lines compared with the parental WM793 cells, indicating their involvement in progression. Next, we analyzed several matched primary/metastatic tumours using methylation-specific PCR, which revealed that MT1E (one of the five genes assessed) was methylated in the largest proportion of tumours. Examination of a larger cohort of samples showed that 1 of 17 (6%) of the benign naevi, 16 of 43 (37%) primary tumours and 6 of 13 (46%) of the metastases displayed MT1E methylation. In addition, ectopic over-expression of MT1E mediated sensitization to cisplatin-induced apoptosis. Overall, these studies suggest that MT1E is a potential tumour suppressor gene, whose loss may promote resistance to apoptosis-inducing therapies.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , DNA Methylation , Drug Resistance, Neoplasm/genetics , Melanoma/genetics , Metallothionein/genetics , Skin Neoplasms/genetics , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Cell Line, Tumor , Cells, Cultured , DNA Methylation/physiology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Humans , Melanoma/metabolism , Metallothionein/metabolism , Metallothionein/physiology , Microarray Analysis , Skin Neoplasms/metabolism , Transfection
8.
Pigment Cell Melanoma Res ; 23(4): 542-53, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20465595

ABSTRACT

In this study, we used array-comparative genomic hybridization (aCGH) and fluorescent in situ hybridization (FISH) to examine genetic aberrations in melanoma cell lines and tissues. Array-comparative genomic hybridization revealed that the most frequent genetic changes found in melanoma cell lines were amplifications on chromosomes 7p and 20q, along with disruptions on Chr 9, 10, 11, 12, 22 and Y. Validation of the results using FISH on tissue microarrays (TMAs) identified TOP1 as being amplified in melanoma tissues. TOP1 amplification was detected in a high percentage (33%) of tumours and was associated with thicker, aggressive tumours. These results show that TOP1 amplification is associated with advanced tumours and poor prognosis in melanoma. These observations open the possibility that TOP1-targeted therapeutics may be of benefit in a particular subgroup of advanced stage melanoma patients.


Subject(s)
DNA Topoisomerases, Type I/genetics , Gene Amplification , Melanoma/diagnosis , Melanoma/enzymology , Cell Line, Tumor , Comparative Genomic Hybridization , DNA Topoisomerases, Type I/metabolism , Humans , In Situ Hybridization, Fluorescence , Male , Melanoma/pathology , Prognosis
9.
Clin Cancer Res ; 15(19): 6042-51, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789302

ABSTRACT

PURPOSE: Cathepsin S is a cysteine protease that promotes the invasion of tumor and endothelial cells during cancer progression. Here we investigated the potential to target cathepsin S using an antagonistic antibody, Fsn0503, to block these tumorigenic effects. EXPERIMENTAL DESIGN: A panel of monoclonal antibodies was raised to human cathepsin S. The effects of a selected antibody were subsequently determined using invasion and proteolysis assays. Endothelial cell tube formation and aorta sprouting assays were done to examine antiangiogenic effects. In vivo effects were also evaluated using HCT116 xenograft studies. RESULTS: A selected cathepsin S antibody, Fsn0503, significantly blocked invasion of a range of tumor cell lines, most significantly HCT116 colorectal carcinoma cells, through inhibition of extracellular cathepsin S-mediated proteolysis. We subsequently found enhanced expression of cathepsin S in colorectal adenocarcinoma biopsies when compared with normal colon tissue. Moreover, Fsn0503 blocked endothelial cell capillary tube formation and aortic microvascular sprouting. We further showed that administration of Fsn0503 resulted in inhibition of tumor growth and neovascularization of HCT116 xenograft tumors. CONCLUSIONS: These results show that blocking the invasive and proangiogenic effects of cathepsin S with antibody inhibitors may have therapeutic utility upon further preclinical and clinical evaluation.


Subject(s)
Adenocarcinoma/pathology , Antibodies, Monoclonal/pharmacology , Cathepsins/antagonists & inhibitors , Colorectal Neoplasms/pathology , Neovascularization, Pathologic/prevention & control , Adenocarcinoma/blood supply , Adenocarcinoma/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cathepsins/immunology , Cell Movement/drug effects , Cells, Cultured , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/therapy , Female , HCT116 Cells , Humans , Immunotherapy/methods , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Rats , Rats, Wistar , Xenograft Model Antitumor Assays
10.
Int J Cancer ; 124(9): 2088-97, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19142967

ABSTRACT

The role of intercellular tight junctions in breast epithelial cells is traditionally thought to be in maintaining polarity and barrier function. However, claudin-4, a tight junction protein, is overexpressed in breast tumour cells compared to normal epithelial cells, which generally corresponds to a loss in polarity. The aim of this study was to investigate the distribution and potential clinical value of claudin-4 in breast cancer, and to evaluate its usefulness as a prognostic and predictive biomarker. Expression of claudin-4 was initially examined by Western blot analysis in a cohort of 88 breast tumours, and was found to correlate positively with tumour grade and negatively with ER. Claudin-4 expression was then evaluated by immunohistochemistry in a larger cohort of 299 tumours represented on a tissue microarray. Claudin-4 expression correlated positively with tumour grade and Her2, and negatively with ER. High claudin-4 expression was also associated with worse breast cancer-specific survival (p = 0.003), recurrence-free survival (p = 0.025) and overall survival (p = 0.034). Multivariate analysis revealed that claudin-4 independently predicted survival in the entire cohort (HR 1.95; 95%CI 1.01-3.79; p = 0.047) and in the ER positive subgroup treated with adjuvant tamoxifen (HR 4.34; 95%CI 1.14-16.53; p = 0.032). This relationship between increased claudin-4 expression and adverse outcome was validated at the mRNA level in a DNA microarray dataset of 295 breast tumours. We conclude that high levels of claudin-4 protein are associated with adverse outcome in breast cancer patients, including the subgroup of patients treated with adjuvant tamoxifen.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Carcinoma, Medullary/metabolism , Membrane Proteins/metabolism , Adenocarcinoma, Mucinous/drug therapy , Adenocarcinoma, Mucinous/secondary , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/secondary , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/secondary , Carcinoma, Medullary/drug therapy , Carcinoma, Medullary/secondary , Claudin-4 , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Membrane Proteins/genetics , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , Receptor, ErbB-2/metabolism , Survival Rate , Tamoxifen/therapeutic use , Tissue Array Analysis , Tumor Cells, Cultured
11.
Int J Cancer ; 120(7): 1434-43, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17205517

ABSTRACT

DNA microarrays have the potential to classify tumors according to their transcriptome. Tissue microarrays (TMAs) facilitate the validation of biomarkers by offering a high-throughput approach to sample analysis. We reanalyzed a high profile breast cancer DNA microarray dataset containing 96 tumor samples using a powerful statistical approach, between group analyses. Among the genes we identified was centromere protein-F (CENP-F), a gene associated with poor prognosis. In a published follow-up breast cancer DNA microarray study, comprising 295 tumour samples, we found that CENP-F upregulation was significantly associated with worse overall survival (p<0.001) and reduced metastasis-free survival (p<0.001). To validate and expand upon these findings, we used 2 independent breast cancer patient cohorts represented on TMAs. CENP-F protein expression was evaluated by immunohistochemistry in 91 primary breast cancer samples from cohort I and 289 samples from cohort II. CENP-F correlated with markers of aggressive tumor behavior including ER negativity and high tumor grade. In cohort I, CENP-F was significantly associated with markers of CIN including cyclin E, increased telomerase activity, c-Myc amplification and aneuploidy. In cohort II, CENP-F correlated with VEGFR2, phosphorylated Ets-2 and Ki67, and in multivariate analysis, was an independent predictor of worse breast cancer-specific survival (p=0.036) and overall survival (p=0.040). In conclusion, we identified CENP-F as a biomarker associated with poor outcome in breast cancer and showed several novel associations of biological significance.


Subject(s)
Breast Neoplasms/genetics , Chromosomal Instability , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic , Microfilament Proteins/genetics , Adult , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Centromere/physiology , Chromosomal Proteins, Non-Histone/metabolism , Female , Follow-Up Studies , Gene Amplification , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Ki-67 Antigen/metabolism , Lymphatic Metastasis , Microfilament Proteins/metabolism , Middle Aged , Oligonucleotide Array Sequence Analysis , Phosphorylation , Prognosis , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Estrogen/metabolism , Survival Rate , Tissue Array Analysis , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...