Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Mol Pharm ; 18(9): 3464-3474, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34448393

ABSTRACT

Optimal cytoreduction for ovarian cancer is often challenging because of aggressive tumor biology and advanced stage. It is a critical issue since the extent of residual disease after surgery is the key predictor of ovarian cancer patient survival. For a limited number of cancers, fluorescence-guided surgery has emerged as an effective aid for tumor delineation and effective cytoreduction. The intravenously administered fluorescent agent, most commonly indocyanine green (ICG), accumulates preferentially in tumors, which are visualized under a fluorescent light source to aid surgery. Insufficient tumor specificity has limited the broad application of these agents in surgical oncology including for ovarian cancer. In this study, we developed a novel tumor-selective fluorescent agent by chemically linking ICG to mouse monoclonal antibody 10D7 that specifically recognizes an ovarian cancer-enriched cell surface receptor, CUB-domain-containing protein 1 (CDCP1). 10D7ICG has high affinity for purified recombinant CDCP1 and CDCP1 that is located on the surface of ovarian cancer cells in vitro and in vivo. Our results show that intravenously administered 10D7ICG accumulates preferentially in ovarian cancer, permitting visualization of xenograft tumors in mice. The data suggest CDCP1 as a rational target for tumor-specific fluorescence-guided surgery for ovarian cancer.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Cell Adhesion Molecules/antagonists & inhibitors , Fluorescent Dyes/administration & dosage , Optical Imaging/methods , Ovarian Neoplasms/diagnosis , Animals , Antibodies, Monoclonal/chemistry , Antigens, Neoplasm , Cell Line, Tumor , Female , Fluorescent Dyes/chemistry , Humans , Indocyanine Green/administration & dosage , Indocyanine Green/chemistry , Injections, Intravenous , Mice , Ovarian Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Cancers (Basel) ; 12(4)2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32260077

ABSTRACT

High stage and recurrent ovarian clear cell carcinoma (OCC) are associated with poor prognosis and resistance to chemotherapy. A distinguishing histological feature of OCC is abundant cytoplasmic stores of glucose, in the form of glycogen, that can be mobilized for cellular metabolism. Here, we report the effect on preclinical models of OCC of disrupting glycogen utilization using the glucose analogue 2-deoxy-D-glucose (2DG). At concentrations significantly lower than previously reported for other cancers, 2DG markedly improves the efficacy in vitro of carboplatin chemotherapy against chemo-sensitive TOV21G and chemo-resistant OVTOKO OCC cell lines, and this is accompanied by the depletion of glycogen. Of note, 2DG doses-of more than 10-fold lower than previously reported for other cancers-significantly improve the efficacy of carboplatin against cell line and patient-derived xenograft models in mice that mimic the chemo-responsiveness of OCC. These findings are encouraging, in that 2DG doses, which are substantially lower than previously reported to cause adverse events in cancer patients, can safely and significantly improve the efficacy of carboplatin against OCC. Our results thus justify clinical trials to evaluate whether low dose 2DG improves the efficacy of carboplatin in OCC patients.

3.
Oncogene ; 39(1): 219-233, 2020 01.
Article in English | MEDLINE | ID: mdl-31471585

ABSTRACT

Elevated CUB-domain containing protein 1 (CDCP1) is predictive of colorectal cancer (CRC) recurrence and poor patient survival. While CDCP1 expression identifies stem cell populations that mediate lung metastasis, mechanisms underlying the role of this cell surface receptor in CRC have not been defined. We sought to identify CDCP1 regulated processes in CRC using stem cell populations, enriched from primary cells and cell lines, in extensive in vitro and in vivo assays. These experiments, demonstrating that CDCP1 is functionally important in CRC tumor initiation, growth and metastasis, identified CDCP1 as a positive regulator of Wnt signaling. Detailed cell fractionation, immunoprecipitation, microscopy, and immunohistochemical analyses demonstrated that CDCP1 promotes translocation of the key regulators of Wnt signaling, ß-catenin, and E-cadherin, to the nucleus. Of functional importance, disruption of CDCP1 reduces nuclear localized, chromatin-associated ß-catenin and nuclear localized E-cadherin, increases sequestration of these proteins in cell membranes, disrupts regulation of CRC promoting genes, and reduces CRC tumor burden. Thus, disruption of CDCP1 perturbs pro-cancerous Wnt signaling including nuclear localization of ß-catenin and E-cadherin.


Subject(s)
Antigens, Neoplasm/genetics , Cadherins/genetics , Cell Adhesion Molecules/genetics , Colorectal Neoplasms/genetics , beta Catenin/genetics , Active Transport, Cell Nucleus/genetics , Carcinogenesis/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Humans , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Wnt Signaling Pathway/genetics
4.
Cell Rep ; 23(13): 3730-3740, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29949758

ABSTRACT

LINE-1 (L1) retrotransposons are a source of insertional mutagenesis in tumor cells. However, the clinical significance of L1 mobilization during tumorigenesis remains unclear. Here, we applied retrotransposon capture sequencing (RC-seq) to multiple single-cell clones isolated from five ovarian cancer cell lines and HeLa cells and detected endogenous L1 retrotransposition in vitro. We then applied RC-seq to ovarian tumor and matched blood samples from 19 patients and identified 88 tumor-specific L1 insertions. In one tumor, an intronic de novo L1 insertion supplied a novel cis-enhancer to the putative chemoresistance gene STC1. Notably, the tumor subclone carrying the STC1 L1 mutation increased in prevalence after chemotherapy, further increasing STC1 expression. We also identified hypomethylated donor L1s responsible for new L1 insertions in tumors and cultivated cancer cells. These congruent in vitro and in vivo results highlight L1 insertional mutagenesis as a common component of ovarian tumorigenesis and cancer genome heterogeneity.


Subject(s)
Evolution, Molecular , Long Interspersed Nucleotide Elements/genetics , Ovarian Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , DNA Methylation , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Loss of Heterozygosity/genetics , Mutagenesis, Insertional , Mutation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics
5.
Am J Cancer Res ; 7(11): 2257-2274, 2017.
Article in English | MEDLINE | ID: mdl-29218249

ABSTRACT

We provide evidence of a pericellular network of proteases that are elevated and co-expressed in prostate cancer. The network involves the membrane bound serine proteases hepsin and TMPRSS2, the secreted kallikrein-related peptidases KLK4 and KLK14, and the secreted matrix metalloproteinases MMP-3 and MMP-9. Western blot analysis of cell lysates, conditioned cell culture media, immunoprecipitates and cell surface proteins, demonstrates a network of interactions centred largely at the plasma membrane, with the Arg/Lys specific proteases hepsin and TMPRSS2 key regulators of the network. Our data demonstrate that like TMPRSS2, hepsin is able to autoactivate. Active hepsin degrades KLK4, generating a cell associated degradation product with corresponding reduction in levels of cell-free KLK4. In contrast hepsin activates KLK14. TMPRSS2 appears to cleave amino terminal to the KLK4 activation site such that it is available for further processing to generate the active KLK4 protease. In contrast with hepsin, TMPRSS2 degrades KLK14. In addition to these direct mechanisms of regulation, hepsin and TMPRSS2 indirectly modulate KLK4 activity by cleaving the KLK4-activating protease MMP-3. Hepsin and TMPRSS2 also activate MMP-9, which similar to MMP-3, associates with the cell surface. Interestingly our data also show that proteolysis occurs between the membrane spanning and catalytic domains of hepsin and TMPRSS2. Hepsin cleavage occurs via an autoproteolytic mechanism, whereas TMPRSS2 cleavage is mediated by KLK14. Hepsin and TMPRSS2 are not shed from the cell surface but proteolysis likely disrupts domains that regulate the proteolytic activity of these proteases. Immunocytochemical analyses demonstrate that hepsin and TMPRSS2 colocalize on the cell surface with the secreted serine proteases KLK4 and KLK14, only in membrane protrusions, suggesting that reciprocal proteolytic interactions occur in defined cellular structures that are important during cancer dissemination for cell migration, invasion and survival. Also of note, immunohistochemical analysis of serial sections of prostate tumor demonstrated significant overlapping expression of the six proteases in vivo. Collectively these data suggest the possibility that the novel proteolytic network identified by us, will be most important during active dissemination of prostate cancers, and that its disruption could inhibit metastasis.

6.
Br J Cancer ; 114(4): 417-26, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26882065

ABSTRACT

BACKGROUND: Development of targeted therapies for high-grade serous ovarian cancer (HGSC) remains challenging, as contributing molecular pathways are poorly defined or expressed heterogeneously. CUB-domain containing protein 1 (CDCP1) is a cell-surface protein elevated in lung, colorectal, pancreas, renal and clear cell ovarian cancer. METHODS: CUB-domain containing protein 1 was examined by immunohistochemistry in HGSC and fallopian tube. The impact of targeting CDCP1 on cell growth and migration in vitro, and intraperitoneal xenograft growth in mice was examined. Three patient-derived xenograft (PDX) mouse models were developed and characterised for CDCP1 expression. The effect of a monoclonal anti-CDCP1 antibody on PDX growth was examined. Src activation was assessed by western blot analysis. RESULTS: Elevated CDCP1 was observed in 77% of HGSC cases. Silencing of CDCP1 reduced migration and non-adherent cell growth in vitro and tumour burden in vivo. Expression of CDCP1 in patient samples was maintained in PDX models. Antibody blockade of CDCP1 significantly reduced growth of an HGSC PDX. The CDCP1-mediated activation of Src was observed in cultured cells and mouse xenografts. CONCLUSIONS: CUB-domain containing protein 1 is over-expressed by the majority of HGSCs. In vitro and mouse model data indicate that CDCP1 has a role in HGSC and that it can be targeted to inhibit progression of this cancer.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Cystadenocarcinoma, Serous/pathology , Neoplasm Proteins/metabolism , Ovarian Neoplasms/pathology , Animals , Antigens, CD/genetics , Antigens, Neoplasm , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cystadenocarcinoma, Serous/metabolism , Disease Models, Animal , Female , Heterografts , Humans , Mice , Neoplasm Grading , Neoplasm Proteins/genetics , Ovarian Neoplasms/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Survival Analysis
7.
Pathology ; 45(1): 49-54, 2013 01.
Article in English | MEDLINE | ID: mdl-23222243

ABSTRACT

AIMS: To examine AGR2 expression in ovarian epithelial tumours and its potential role as a prognostic biomarker. METHODS: Tissue microarray technology and immunohistochemistry were used to investigate AGR2 expression in ovarian epithelial tumours and in non-neoplastic ovarian epithelium. For the carcinomas, the expression data were correlated with clinicopathological features and disease outcome. RESULTS: AGR2 was expressed in all benign, borderline and malignant mucinous tumours and in a high proportion of endometrioid carcinomas (89%). AGR2 was frequently expressed in benign and borderline serous tumours (76% and 95%, respectively), but less commonly expressed in serous carcinomas (19%, p < 0.001). AGR2 expression in ovarian carcinomas was inversely correlated with p53 and p16 expression (p = 0.002 and p < 0.001, respectively), and independent of CA125 expression. AGR2 expression was more common in carcinomas which presented with early-stage compared with late-stage disease (p = 0.009) and AGR2 was expressed in carcinomas with better outcome (22% relapse rate of AGR2 positive cancers compared with 74% relapse rate for AGR2 negative cancers, p = 0.001). CONCLUSION: Our findings indicate that AGR2 expression is associated with mucinous carcinomas and their precursor lesions and endometrioid cancers. Additionally, AGR2 may be an important prognostic biomarker of ovarian cancer.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Endometrioid/metabolism , Ovarian Neoplasms/metabolism , Proteins/metabolism , Adenocarcinoma, Mucinous/pathology , Carcinoma, Endometrioid/pathology , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16 , Diagnosis, Differential , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Mucoproteins , Neoplasm Proteins/metabolism , Oncogene Proteins , Ovarian Neoplasms/pathology , Prognosis , Tissue Array Analysis , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...