Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Br J Cancer ; 111(8): 1634-45, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25167227

ABSTRACT

BACKGROUND: The level of plasma-derived naturally circulating anti-glycan antibodies (AGA) to P1 trisaccharide has previously been shown to significantly discriminate between ovarian cancer patients and healthy women. Here we aim to identify the Ig class that causes this discrimination, to identify on cancer cells the corresponding P1 antigen recognised by circulating anti-P1 antibodies and to shed light into the possible function of this glycosphingolipid. METHODS: An independent Australian cohort was assessed for the presence of anti-P1 IgG and IgM class antibodies using suspension array. Monoclonal and human derived anti-glycan antibodies were verified using three independent glycan-based immunoassays and flow cytometry-based inhibition assay. The P1 antigen was detected by LC-MS/MS and flow cytometry. FACS-sorted cell lines were studied on the cellular migration by colorimetric assay and real-time measurement using xCELLigence system. RESULTS: Here we show in a second independent cohort (n=155) that the discrimination of cancer patients is mediated by the IgM class of anti-P1 antibodies (P=0.0002). The presence of corresponding antigen P1 and structurally related epitopes in fresh tissue specimens and cultured cancer cells is demonstrated. We further link the antibody and antigen (P1) by showing that human naturally circulating and affinity-purified anti-P1 IgM isolated from patients ascites can bind to naturally expressed P1 on the cell surface of ovarian cancer cells. Cell-sorted IGROV1 was used to obtain two study subpopulations (P1-high, 66.1%; and P1-low, 33.3%) and observed that cells expressing high P1-levels migrate significantly faster than those with low P1-levels. CONCLUSIONS: This is the first report showing that P1 antigen, known to be expressed on erythrocytes only, is also present on ovarian cancer cells. This suggests that P1 is a novel tumour-associated carbohydrate antigen recognised by the immune system in patients and may have a role in cell migration. The clinical value of our data may be both diagnostic and prognostic; patients with low anti-P1 IgM antibodies present with a more aggressive phenotype and earlier relapse.


Subject(s)
Antigens, Neoplasm/immunology , Glycosphingolipids/immunology , Neoplasm Metastasis/immunology , Ovarian Neoplasms/immunology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/isolation & purification , Cell Line, Tumor , Chromatography, Affinity , Female , Flow Cytometry , Humans , Ovarian Neoplasms/pathology
2.
Ann Oncol ; 25(2): 322-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285017

ABSTRACT

Cytoreductive surgery and chemotherapy continue to be the mainstay of ovarian cancer treatment. However, as mortality from advanced ovarian cancer remains very high, novel therapies are required to be integrated into existing treatment regimens. Immunotherapy represents an alternative and rational therapeutic approach for ovarian cancer based on a body of evidence supporting a protective role of the immune system against these cancers, and on the clinical success of immunotherapy in other malignancies. Whether or not immunotherapy will have a role in the future management of ovarian cancer is too early to tell, but research in this field is active. This review will discuss recent clinical developments of selected immunotherapies for ovarian cancer which fulfil the following criteria: (i) they are antibody-based, (ii) target a distinct immunological pathway, and (iii) have reached the clinical trial stage. Specifically, the focus is on Catumaxomab (anti-EpCAM×anti-CD3), Abagovomab, Oregovomab (anti-CA125), Daclizumab (anti-CD25), Ipilimumab (anti-CTLA-4), and MXD-1105 (anti-PD-L1). Catumaxomab has reached phase III clinical trials and exhibits promise with reports, showing that it can cause a significant and sustained reduction in ascites. Phase I-III clinical trials continue to be conducted on the other antibodies, some of which have had encouraging reports. We will also provide our perspective on the future of immunotherapy for ovarian cancer, and how it may be best employed in treatment regimens.


Subject(s)
Antineoplastic Agents/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Clinical Trials as Topic , Daclizumab , Diphtheria Toxin/therapeutic use , Female , Humans , Immunoglobulin G/therapeutic use , Immunotherapy , Interleukin-2/therapeutic use , Ipilimumab , Recombinant Fusion Proteins/therapeutic use
3.
Br J Cancer ; 98(6): 1085-93, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18349819

ABSTRACT

Despite a high initial response rate to first-line platinum/paclitaxel chemotherapy, most women with epithelial ovarian cancer relapse with recurrent disease that becomes refractory to further cytotoxic treatment. We have previously shown that the E3 ubiquitin ligase, EDD, a regulator of DNA damage responses, is amplified and overexpressed in serous ovarian carcinoma. Given that DNA damage pathways are linked to platinum resistance, the aim of this study was to determine if EDD expression was associated with disease recurrence and platinum sensitivity in serous ovarian cancer. High nuclear EDD expression, as determined by immunohistochemistry in a cohort of 151 women with serous ovarian carcinoma, was associated with an approximately two-fold increased risk of disease recurrence and death in patients who initially responded to first-line chemotherapy, independently of disease stage and suboptimal debulking. Although EDD expression was not directly correlated with relative cisplatin sensitivity of ovarian cancer cell lines, sensitivity to cisplatin was partially restored in platinum-resistant A2780-cp70 ovarian cancer cells following siRNA-mediated knockdown of EDD expression. These results identify EDD as a new independent prognostic marker for outcome in serous ovarian cancer, and suggest that pathways involving EDD, including DNA damage responses, may represent new therapeutic targets for chemoresistant ovarian cancer.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Cystadenocarcinoma, Serous , Female , Humans , Middle Aged , Neoplasm Recurrence, Local , Ovarian Neoplasms/drug therapy , Prognosis , Retrospective Studies
4.
Br J Cancer ; 94(6): 904-13, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16508639

ABSTRACT

Mucinous epithelial ovarian cancers (MOC) are clinically and morphologically distinct from the other histological subtypes of ovarian cancer. To determine the genetic basis of MOC and to identify potential tumour markers, gene expression profiling of 49 primary ovarian cancers of different histological subtypes was performed using a customised oligonucleotide microarray containing >59 000 probesets. The results show that MOC express a genetic profile that both differs and overlaps with other subtypes of epithelial ovarian cancer. Concordant with its histological phenotype, MOC express genes characteristic of mucinous carcinomas of varying epithelial origin, including intestinal carcinomas. Differences in gene expression between MOC and other histological subtypes of ovarian cancer were confirmed by RT-PCR and/or immunohistochemistry. In particular, galectin 4 (LGALS4) was highly and specifically expressed in MOC, but expressed at lower levels in benign mucinous cysts and borderline (atypical proliferative) tumours, supporting a malignant progression model of MOC. Hence LGALS4 may have application as an early and differential diagnostic marker of MOC.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Gene Expression Profiling , Genetic Markers , Ovarian Neoplasms/genetics , Adenocarcinoma, Mucinous/pathology , Cell Transformation, Neoplastic , Disease Progression , Female , Galectin 4/biosynthesis , Humans , Immunohistochemistry , Middle Aged , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/pathology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
5.
Ann Oncol ; 14(6): 938-45, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12796033

ABSTRACT

BACKGROUND: Ataxia-telangiectasia is a pleiotropic autosomal recessive disorder caused by mutations in the ATM gene. In addition to a profound cancer predisposition, another hallmark of ataxia-telangiectasia is radiosensitivity. Recently, p53-null mouse fibroblasts have been reported to be radiosensitised by the concurrent loss of ATM. MATERIALS AND METHODS: We compared the sensitivity of atm(+/+)/p53(-/-) and atm(-/-)/p53(-/-) mouse embryonic fibroblasts to different classes of chemotherapeutic agents using the MTT assay, Trypan Blue exclusion and fluorescence-activated cell sorting for cell cycle and apoptosis analyses. RESULTS: Loss of ATM function in p53-deficient cells resulted in a 2- to 4-fold increase in sensitivity to the topoisomerase I poisons camptothecin and topotecan, to the topoisomerase II poisons doxorubicin, epirubicin and etoposide, and to the antimetabolites 5-fluorouracil and gemcitabine, but not to the platinum compounds cisplatin, carboplatin and oxaliplatin, the taxanes docetaxel and paclitaxel, or to busulfan. Loss of ATM function did not result in increased apoptosis, but resulted in increased Trypan Blue staining in response to epirubicin, suggesting that processes other than apoptosis may mediate cytotoxicity. ATM deficiency did not alter the extent of G(1)/S or G(2)/M cell cycle phase accumulation produced by epirubicin, suggesting that enhanced sensitivity was not due to failure of checkpoint activation. CONCLUSIONS: We provide further evidence that ATM is involved in regulating cellular defences against some cytotoxic agents in the absence of p53. Tumour-targeted functional inhibition of ATM may be a valuable strategy for increasing the efficacy of anticancer agents in the treatment of p53-mutant cancers.


Subject(s)
Antineoplastic Agents/toxicity , Fibroblasts/drug effects , Gene Deletion , Protein Serine-Threonine Kinases/physiology , Topoisomerase II Inhibitors , Tumor Suppressor Protein p53/deficiency , Animals , Antimetabolites/toxicity , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/drug effects , Cell Cycle Proteins , Cell Line , DNA-Binding Proteins , Drug Resistance, Neoplasm , Enzyme Inhibitors/toxicity , Fibroblasts/metabolism , Homozygote , In Situ Nick-End Labeling , Mice , Mice, Knockout , Trypan Blue , Tumor Suppressor Proteins
6.
Br J Cancer ; 87(9): 1027-33, 2002 Oct 21.
Article in English | MEDLINE | ID: mdl-12434296

ABSTRACT

A large fraction of human tumours carries mutations in the p53 gene. p53 plays a central role in controlling cell cycle checkpoint regulation, DNA repair, transcription, and apoptosis upon genotoxic stress. Lack of p53 function impairs these cellular processes, and this may be the basis of resistance to chemotherapeutic regimens. By virtue of the involvement of DNA mismatch repair in modulating cytotoxic pathways in response to DNA damaging agents, we investigated the effects of loss of Pms2 on the sensitivity to a panel of widely used anticancer agents in E1A/Ha-Ras-transformed p53-null mouse fibroblasts either proficient or deficient in Pms2. We report that lack of the Pms2 gene is associated with an increased sensitivity, ranging from 2-6-fold, to some types of anticancer agents including the topoisomerase II poisons doxorubicin, etoposide and mitoxantrone, the platinum compounds cisplatin and oxaliplatin, the taxanes docetaxel and paclitaxel, and the antimetabolite gemcitabine. In contrast, no change in sensitivity was found after treatment with 5-fluorouracil. Cell cycle analysis revealed that both, Pms2-deficient and -proficient cells, retain the ability to arrest at the G2/M upon cisplatin treatment. The data indicate that the concomitant loss of Pms2 function chemosensitises p53-deficient cells to some types of anticancer agents, that Pms2 positively modulates cell survival by mechanisms independent of p53, and that increased cytotoxicity is paralleled by increased apoptosis. Tumour-targeted functional inhibition of Pms2 may be a valuable strategy for increasing the efficacy of anticancer agents in the treatment of p53-mutant cancers.


Subject(s)
Adenosine Triphosphatases/physiology , Antineoplastic Agents/pharmacology , Cell Line, Transformed/drug effects , DNA Repair Enzymes , DNA-Binding Proteins/physiology , Fibroblasts/drug effects , Taxoids , Tumor Suppressor Protein p53/deficiency , Animals , Apoptosis/drug effects , Base Pair Mismatch , Bridged-Ring Compounds/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Transformed/metabolism , DNA Repair , Drug Resistance, Neoplasm , Fibroblasts/metabolism , Fibroblasts/pathology , Fluorouracil/pharmacology , Homozygote , Mice , Mice, Knockout , Mismatch Repair Endonuclease PMS2 , Organoplatinum Compounds/pharmacology , Trypan Blue
7.
Br J Cancer ; 86(7): 1130-5, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11953861

ABSTRACT

Loss of DNA mismatch repair is a common finding in hereditary nonpolyposis colon cancer as well as in many types of sporadic human tumours. DNA mismatch repair-deficient cells have been reported to be resistant to many chemotherapeutic agents and to radiotherapy, and to have the potential of rapidly acquiring additional mutations leading to tumour progression. Photodynamic therapy is a new treatment modality using light to activate a photosensitiser that preferentially localises in tumour cells. An oxygen dependent photochemical reaction ensues, resulting in selective tumour necrosis. The effect of loss of DNA mismatch repair activity on the sensitivity to photodynamic therapy was tested using pairs of cell lines proficient or deficient in mismatch repair due to loss of either MLH1 or MSH2 protein function. Cells were incubated with the photosensitiser 5,10,15,20-meta-tetra(hydroxyphenyl)chlorin and exposed to laser light at 652 nm with various optical doses ranging from 0-1 J cm(-2). Cell survival was assessed using the clonogenic assay. Loss of MLH1 or MSH2 function was not associated with resistance to photodynamic therapy. MCF-7 cells repeatedly treated with photodynamic therapy expressed parental levels of MLH1, MSH2, MSH6, and PMS2. DNA mismatch repair-deficient and -proficient cells showed similar subcellular distributions of meta-tetra(hydroxyphenyl)chlorin as analysed by laser scanning and fluorescence microscopy. Therefore, repeated exposure of tumour cells to photodynamic therapy does not seem to result in loss of DNA mismatch repair, and loss of mismatch repair, in turn, does not seem to contribute to resistance to photodynamic therapy. Our results suggest recommending photodynamic therapy as a strategy for circumventing resistance due to loss of DNA mismatch repair.


Subject(s)
Base Pair Mismatch , DNA Repair , DNA-Binding Proteins , Drug Resistance, Neoplasm , Neoplasm Proteins/genetics , Photochemotherapy , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing , Adenocarcinoma/pathology , Breast Neoplasms/pathology , Carrier Proteins , Colorectal Neoplasms/pathology , Humans , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/biosynthesis , Nuclear Proteins , Proto-Oncogene Proteins/biosynthesis , Tumor Cells, Cultured
8.
Lasers Surg Med ; 29(3): 252-9, 2001.
Article in English | MEDLINE | ID: mdl-11573228

ABSTRACT

BACKGROUND AND OBJECTIVE: The goal of this study was to evaluate various creams for their capability to protect photosensitized skin from visible light. STUDY DESIGN/MATERIALS AND METHODS: Two cover creams and creams containing various combinations of Vaseline with TiO(2), ZnO, and Fe(2)O(3) were used to measure the reduced light transmission and the light absorption spectrum. In vitro and in vivo tests were performed to assess the protection from light by above mentioned compounds. RESULTS: The cover creams and the 50% TiO(2) cream showed similar efficacy in reducing light transmission, while the sunscreen was less efficient by a factor of 5. Cell protection by 25% TiO(2)+25% ZnO, TiO(2), or the cover creams was more efficient than protection by the sunscreen or other compounds. In vivo, the dark cover cream protected the skin by a factor of 3.4 better than the sunscreen. CONCLUSION: The dark cover cream has acceptable properties to protect photosensitized skin.


Subject(s)
Ferric Compounds/therapeutic use , Photochemotherapy/adverse effects , Photosensitivity Disorders/drug therapy , Skin/drug effects , Sunscreening Agents/therapeutic use , Titanium/therapeutic use , Zinc Oxide/therapeutic use , Absorption/drug effects , Absorption/radiation effects , Erythema/etiology , Erythema/prevention & control , Female , Ferric Compounds/radiation effects , Humans , In Vitro Techniques , Keratinocytes/drug effects , Keratinocytes/radiation effects , Male , Microscopy, Atomic Force , Photosensitizing Agents/radiation effects , Photosensitizing Agents/therapeutic use , Skin/radiation effects , Spectrum Analysis , Sunscreening Agents/radiation effects , Titanium/radiation effects , Zinc Oxide/radiation effects
9.
Int J Cancer ; 93(4): 571-6, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11477562

ABSTRACT

Sporadic breast carcinomas demonstrate microsatellite instability, reflecting the presence of DNA mismatch repair-deficient cells, in about one fourth of cases at the time of diagnosis. Loss of DNA mismatch repair has been reported to result in resistance not only to cisplatin and alkylating agents but also to the topoisomerase II poison doxorubicin, suggesting an association between DNA mismatch repair and topoisomerase II poison-induced cytotoxicity. Our study investigates the relationship between loss of MSH2 or MLH1 function and sensitivity to the topoisomerase I and II poisons, and to the taxanes, 2 classes of cytotoxic drugs commonly used in breast cancer. Two pairs of cell lines proficient and deficient in mismatch repair due to loss of either MSH2 or MLH1 function were used. Loss of either MSH2 or MLH1 function resulted in resistance to the topoisomerase II poisons doxorubicin, epirubicin and mitoxantrone, whereas only loss of MLH1 function was associated with low-level resistance to the topoisomerase I poisons camptothecin and topotecan. In contrast, there was no resistance to docetaxel and paclitaxel. Our data support the hypothesis that both MSH2 and MLH1 are involved in topoisomerase II poison-mediated cytotoxicity, whereas only MLH1 is involved in topoisomerase I poison-mediated cytotoxicity. Since our study shows that loss of DNA mismatch repair does not result in resistance to the taxanes, these drugs can be recommended for use in breast cancer deficient in mismatch repair.


Subject(s)
Base Pair Mismatch , DNA Repair , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Taxoids , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Adaptor Proteins, Signal Transducing , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Antibiotics, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Carrier Proteins , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Docetaxel , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/genetics , Epirubicin/pharmacology , Female , Humans , Intercalating Agents/pharmacology , Mitoxantrone/pharmacology , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/physiology , Nuclear Proteins , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Proto-Oncogene Proteins/physiology , Gemcitabine
10.
Gynecol Oncol ; 80(1): 62-6, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11136571

ABSTRACT

OBJECTIVES: The aim of this study was twofold: first, to determine the feasibility of photodynamic therapy (PDT) of vulvar intraepithelial neoplasia III (VIN III) using topically applied 5-aminolevulinic acid (ALA) for photosensitization, and second, to compare PDT results with those of laser evaporation and local excision. METHODS: Fifteen patients with VIN III had 10 g of 10% ALA gel applied to the entire vulva. Two to three hours after drug application the vulva was irradiated with 120 J/cm(2) laser light at a wavelength of 635 nm. The procedure was performed without anesthesia in most patients. Thirty patients with VIN III treated by laser evaporation and 27 patients treated by surgical excision served as controls. RESULTS: Eight weeks following PDT, 11 of 15 patients were free of VIN III as determined by biopsy. Excellent tissue preservation was achieved and no ulcers or scarring occurred. Three recurrences were seen during follow-up, at 5, 6, and 7 months after PDT. Twelve months after treatment, analysis of disease-free survival revealed no statistically significant difference between patients treated with PDT and patients treated with conventional treatment modalities (P = 0.67) but the power of this analysis is low. In multivariate analysis, multifocal disease was the sole variable associated with a reduced disease-free survival. CONCLUSION: While PDT of VIN III seems to show efficacy similar to that of conventional treatment modalities it offers unique advantages: healing time is short, preservation of normal vulvar appearance is excellent, and PDT may be performed without anesthesia. Hence, PDT of VIN III deserves further investigation.


Subject(s)
Aminolevulinic Acid/administration & dosage , Carcinoma in Situ/drug therapy , Photochemotherapy , Photosensitizing Agents/administration & dosage , Vulvar Neoplasms/drug therapy , Administration, Topical , Adult , Aminolevulinic Acid/adverse effects , Disease-Free Survival , Feasibility Studies , Female , Gels , Humans , Laser Therapy , Middle Aged , Multivariate Analysis , Photochemotherapy/adverse effects , Photosensitizing Agents/adverse effects , Vulvar Neoplasms/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...