Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Vet J ; 239: 21-29, 2018 09.
Article in English | MEDLINE | ID: mdl-30197105

ABSTRACT

Precision medicine can be defined as the prevention, investigation and treatment of diseases taking individual variability into account. There are multiple ways in which the field of precision medicine may be advanced; however, recent innovations in the fields of electronics and microfabrication techniques have led to an increased interest in the use of implantable biosensors in precision medicine. Implantable biosensors are an important class of biosensors because of their ability to provide continuous data on the levels of a target analyte; this enables trends and changes in analyte levels over time to be monitored without any need for intervention from either the patient or clinician. As such, implantable biosensors have great potential in the diagnosis, monitoring, management and treatment of a variety of disease conditions. In this review, we describe precision medicine and the role implantable biosensors may have in this field, along with challenges in their clinical implementation due to the host immune responses they elicit within the body.


Subject(s)
Biosensing Techniques/veterinary , Precision Medicine/veterinary , Prostheses and Implants/veterinary , Veterinary Medicine/methods , Animals , Biosensing Techniques/statistics & numerical data , Precision Medicine/instrumentation , Precision Medicine/methods , Prostheses and Implants/statistics & numerical data , Veterinary Medicine/instrumentation
2.
Br J Cancer ; 110(12): 2975-84, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24867692

ABSTRACT

BACKGROUND: The response of ovarian cancer patients to carboplatin and paclitaxel is variable, necessitating identification of biomarkers that can reliably predict drug sensitivity and resistance. In this study, we sought to identify dynamically controlled genes and pathways associated with drug response and its time dependence. METHODS: Gene expression was assessed for 14 days post-treatment with carboplatin or carboplatin-paclitaxel in xenografts from two ovarian cancer models: platinum-sensitive serous adenocarcinoma-derived OV1002 and a mixed clear cell/endometrioid carcinoma-derived HOX424 with reduced sensitivity to platinum. RESULTS: Tumour volume reduction was observed in both xenografts, but more dominantly in OV1002. Upregulated genes in OV1002 were involved in DNA repair, cell cycle and apoptosis, whereas downregulated genes were involved in oxygen-consuming metabolic processes and apoptosis control. Carboplatin-paclitaxel triggered a more comprehensive response than carboplatin only in both xenografts. In HOX424, apoptosis and cell cycle were upregulated, whereas Wnt signalling was inhibited. Genes downregulated after day 7 from both xenografts were predictive of overall survival. Overrepresented pathways were also predictive of outcome. CONCLUSIONS: Late expressed genes are prognostic in ovarian tumours in a dynamic manner. This longitudinal gene expression study further elucidates chemotherapy response in two models, stressing the importance of delayed biomarker detection and guiding optimal timing of biopsies.


Subject(s)
Carboplatin/pharmacology , Gene Expression Regulation, Neoplastic/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Paclitaxel/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/genetics , Cell Cycle/genetics , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , Energy Metabolism/genetics , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/mortality , Prognosis , Wnt Signaling Pathway/genetics
3.
Br J Cancer ; 106(11): 1779-89, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22549178

ABSTRACT

BACKGROUND: Trastuzumab and pertuzumab target the Human Epidermal growth factor Receptor 2 (HER2). Combination therapy has been shown to provide enhanced antitumour activity; however, the downstream signalling to explain how these drugs mediate their response is not clearly understood. METHODS: Transcriptome profiling was performed after 4 days of trastuzumab, pertuzumab and combination treatment in human ovarian cancer in vivo. Signalling pathways identified were validated and investigated in primary ovarian xenografts at the protein level and across a timeseries. RESULTS: A greater number and variety of genes were differentially expressed by the combination of antibody therapies compared with either treatment alone. Protein levels of cyclin-dependent kinase inhibitors p21 and p27 were increased in response to both agents and further by the combination; pERK signalling was inhibited by all treatments; but only pertuzumab inhibited pAkt signalling. The expression of proliferation, apoptosis, cell division and cell-cycle markers was distinct in a panel of primary ovarian cancer xenografts, suggesting the heterogeneity of response in ovarian cancer and a need to establish predictive biomarkers. CONCLUSION: This first comprehensive study of the molecular response to trastuzumab, pertuzumab and combined therapy in vivo highlights both common and distinct downstream effects to agents used alone or in combination, suggesting that complementary pathways may be involved.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Expression/drug effects , Ovarian Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Blotting, Western , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Trastuzumab , Xenograft Model Antitumor Assays
4.
Br J Cancer ; 103(3): 401-10, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20628393

ABSTRACT

BACKGROUND: C35 is a 12 kDa membrane-anchored protein endogenously over-expressed in many invasive breast cancers. C35 (C17orf37) is located on the HER2 amplicon, between HER2 and GRB7. The function of over-expressed C35 in invasive breast cancer is unknown. METHODS: Tissue microarrays containing 122 primary human breast cancer specimens were used to examine the association of C35 with HER2 expression. Cell lines over-expressing C35 were generated and tested for evidence of cell transformation in vitro. RESULTS: In primary breast cancers high levels of C35 mRNA expression were associated with HER2 gene amplification. High levels of C35 protein expression were associated with hallmarks of transformation, such as, colony growth in soft agar, invasion into collagen matrix and formation of large acinar structures in three-dimensional (3D) cell cultures. The transformed phenotype was also associated with characteristics of epithelial to mesenchymal transition, such as adoption of spindle cell morphology and down-regulation of epithelial markers, such as E-cadherin and keratin-8. Furthermore, C35-induced transformation in 3D cell cultures was dependent on Syk kinase, a downstream mediator of signalling from the immunoreceptor tyrosine-based activation motif, which is present in C35. CONCLUSION: C35 functions as an oncogene in breast cancer cell lines. Drug targeting of C35 or Syk kinase might be helpful in treating a subset of patients with HER2-amplified breast cancers.


Subject(s)
Breast Neoplasms/genetics , Genes, erbB-2 , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, CD , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cadherins/genetics , Cell Line, Tumor , Colony-Forming Units Assay , DNA Primers , Down-Regulation , Female , Gene Amplification , Humans , Immunohistochemistry , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Open Reading Frames , RNA, Messenger/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Syk Kinase , Transfection , Trastuzumab
6.
Ann Oncol ; 21(6): 1254-1261, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19858088

ABSTRACT

BACKGROUND: Assessment of receptors [estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2)] is routinely carried out on primary tumour in order to select appropriate adjuvant therapy; the same analysis is not carried out on nodal metastases. Since de novo resistance to therapy is common, we quantified differences in receptor expression between primary and nodal disease in order to assess whether this might contribute to therapeutic resistance. PATIENTS AND METHODS: A total of 385 patients with invasive primary breast carcinomas and paired lymph nodes (n = 211) were assessed for ER, PR and HER2 expression using quantitative immunofluorescence. Cut-points were defined by comparison with tumours scored by immunohistochemistry (IHC) and FISH. Differences in expression for each of the markers and molecular phenotype were analysed. RESULTS: Quantitative receptor expression shows a wide dynamic range compared with IHC. Overall, 46.9% cases had disparate breast/node receptor status of at least one receptor. Many of the differences in expression between primary tumour and node are large magnitude (greater than fivefold) changes. Triple-negative phenotype changes in 23.1% of cases. CONCLUSIONS: A significant number of patients show discordant quantitative expression of molecular markers between primary and nodal disease. Appropriately measured, lymph node receptor status could be a more accurate measurement for guiding adjuvant therapy, which requires testing in a clinical trial.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Lymph Nodes/metabolism , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma/pathology , Female , Fluorescent Antibody Technique/methods , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Matched-Pair Analysis , Neoplasm Staging/methods , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Research Design , Tumor Burden
7.
Eur J Cancer ; 41(11): 1637-44, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15953713

ABSTRACT

Kahalalide F (KF) is a small natural peptide that showed activity in vitro and in vivo. The dose-limiting toxicity in clinical trials was transaminitis. We investigated the cytotoxicity of KF in cell lines from breast, ovary, prostate and colon cancers, but focused on hepatoma cell lines, performing mechanistic studies in HepG2 (IC50 = 0.3 microM) and PLC/PRF/5C (IC50 = 5 microM). Following KF exposure, HepG2 cells demonstrated profound ATP depletion, associated with cell swelling and cell blebbing, and increased permeability to propidium iodide (PI), annexin V (AV) and release of lactate dehydrogenase (LDH). PLC/PRF/5C cells retained their cell structure, but were permeable to PI and, following exposure to high concentrations of KF, to AV. The pattern of cell permeability is similar to maitotoxin, another small cytotoxic peptide, but the differential effects on the cell membrane induced by KF in HepG2 and PLC/PRF/5C suggest specific interactions with membranes or proteins. This could lead to better drug design aimed at exploiting the potential for cell selectivity.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Depsipeptides/pharmacology , Liver Neoplasms/metabolism , Adenosine Triphosphate/deficiency , Cell Line, Tumor , Cell Membrane/drug effects , Depsipeptides/pharmacokinetics , Humans , Permeability/drug effects
8.
Br J Cancer ; 92(10): 1927-33, 2005 May 23.
Article in English | MEDLINE | ID: mdl-15870720

ABSTRACT

Ovulation is believed to contribute to the development of ovarian cancers that derive from the ovarian surface epithelium (OSE). The process of ovulation is synonymous with inflammation and inflammatory cytokines such as interleukin-1alpha (IL-1alpha) have recently been shown to induce both inflammatory and anti-inflammatory responses in human OSE (HOSE) cells. In this study we directly compared levels of IL-1alpha-induced gene expression by analysing the levels of 11beta-hydroxysteroid dehydrogenase (11betaHSD) types 1 (11betaHSD-1) and 2 (11betaHSD-2), cyclooxygenase-2 (COX-2), IL-1 receptor (IL-1R) and glucocorticoid receptor alpha (GRalpha) mRNA between normal HOSE cells and cell lines derived from poorly differentiated (SKOV-3, BG-1, PEO-4) and well-differentiated (PEO-14) ovarian adenocarcinoma. In HOSE cell cultures, and to a lesser extent PEO-14 cells, the basal mRNA levels of COX-2 and 11betaHSD-1 were relatively high and further shown to be induced in response to IL-1alpha (for HOSE cells; >20-fold, P<0.05 and PEO-14 cells; >3fold, P<0.05). However, whereas HOSE cells expressed a low level of 11betaHSD-2 mRNA that was only mildly responsive to IL-1alpha (1.3-fold, P<0.001), all cell lines exhibited a higher basal level of 11betaHSD-2 mRNA that was in some cases further stimulated in PEO-4 cells (five-fold; P<0.05) or suppressed in SKOV-3 cells (two-fold; P<0.01) in response to IL-1alpha. All cells tested expressed IL-1R and, with the exception of BG-1, GRalpha. These results indicate that cell lines derived from ovarian cancers have lost the ability to respond normally to inflammatory cytokines such as IL-1alpha. The finding that normal OSE cells, in contrast to cell lines derived from patients with ovarian adenocarcinoma, abundantly express 11betaHSD-1 mRNA but are essentially devoid of 11betaHSD-2 mRNA supports the concept that the pattern of 11betaHSD isoform gene expression is a defining feature of neoplastic cellular transformation, which might have particular relevance to the ovary.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/immunology , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation , Inflammation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , 11-beta-Hydroxysteroid Dehydrogenases/biosynthesis , Cell Differentiation , Cell Transformation, Neoplastic , Cyclooxygenase 2 , Epithelial Cells , Female , Humans , Interleukin-1/pharmacology , Membrane Proteins , Ovulation , Prostaglandin-Endoperoxide Synthases/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
9.
Br J Cancer ; 86(3): 456-62, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11875715

ABSTRACT

The modulating effects of the orally active epidermal growth factor receptor-specific tyrosine kinase inhibitor ZD 1839 ("Iressa") on cell growth and signalling were evaluated in four ovarian cancer cell lines (PE01, PE04, SKOV-3, OVCAR-5) that express the epidermal growth factor receptor, and in A2780, which is epidermal growth factor receptor-negative. Transforming growth factor-alpha stimulated growth was completely inhibited by concentrations of ZD 1839 > or =0.3 microM in the epidermal growth factor receptor-expressing cell lines, as were transforming growth factor-alpha stimulated phosphorylation of the epidermal growth factor receptor and downstream components of the MAP kinase and PI-3 kinase signalling cascades. Growth inhibition in the absence of added transforming growth factor-alpha was also observed which could be consistent with suppression of action of autocrine epidermal growth factor receptor-activating ligands by ZD 1839. In support of this, transforming growth factor-alpha, EGF and amphiregulin mRNAs were detected by RT-PCR in the epidermal growth factor receptor-expressing cell lines. ZD 1839 inhibited growth of the PE04 ovarian cancer xenograft at 200 mg kg(-1)day(-1). These data lend further support to the view that targeting the epidermal growth factor receptor in ovarian cancer could have therapeutic benefit.


Subject(s)
ErbB Receptors/physiology , Ovarian Neoplasms/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/therapeutic use , Animals , Cell Division/drug effects , ErbB Receptors/drug effects , Female , Gefitinib , Humans , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Br J Cancer ; 85(11): 1753-8, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11742498

ABSTRACT

c-Raf is an essential component of the extracellular related kinase (ERK) signal transduction pathway. Immunohistochemical staining indicated that c-Raf was present in 49/53 ovarian adenocarcinomas investigated and high c-Raf expression correlated significantly with poor survival (P = 0.002). c-Raf protein was detected in 15 ovarian cancer cell lines. Antisense oligodeoxynucleotides (ODNs) (ISIS 5132 and ISIS 13650) reduced c-Raf protein levels and inhibited cell proliferation in vitro. Selectivity was demonstrated by the lack of effect of ISIS 5132 on A-Raf or ERK, while a random ODN produced only minor effects on growth and did not influence c-Raf expression. ISIS 5132 produced enhanced apoptosis and cells accumulated in S and G(2)/M phases of the cell cycle. In vivo, ISIS 5132 inhibited growth of the s.c. SKOV-3 xenograft while a mismatch ODN had no effect. These data indicate that high levels of c-Raf expression may be important in ovarian cancer and use of antisense ODNs targeted to c-Raf could provide a strategy for the treatment of this disease.


Subject(s)
DNA, Antisense/pharmacology , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-raf/drug effects , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , DNA, Antisense/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-raf/biosynthesis , Proto-Oncogene Proteins c-raf/genetics , Survival Analysis , Time Factors , Xenograft Model Antitumor Assays
11.
Cancer Res ; 61(5): 2169-76, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280782

ABSTRACT

Immunohistochemical expression of erbB4 protein was identified in 93% (49 of 53) of ovarian cancers using the HFR-1 antibody (targeted to the cytoplasmic domain of the erbB4 receptor) and in 89% (47 of 53) of ovarian cancers using the H4.77.16 antibody (targeted to the extracellular domain). Tumors of serous histology were more likely to express a higher level of erbB4 than endometrioid tumors, and for stage III serous tumors, long-term survival was associated with moderate to high coexpression of erbB4 and erbB2. Within ovarian cancer cell lines, high erbB4 expression was associated with cisplatin resistance. Using reverse transcription-PCR, the presence of multiple isoforms of erbB4 mRNA was identified in both ovarian primary tumors and cell lines. Splice variants in the juxtamembrane (JM-a and JM-d) and cytoplasmic (CT-a and CT-b) regions were identified in mRNA of both cell lines and primary tumors. The use of an anti-erbB4 blocking antibody suggested that erbB4 was not the mediator of the growth stimulatory effects of neuregulin in ovarian cancer cells and indeed could potentially antagonize this effect.


Subject(s)
ErbB Receptors/biosynthesis , Ovarian Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Alternative Splicing , Antibodies/pharmacology , Base Sequence , Blotting, Western , Cell Division/drug effects , Cell Division/physiology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Humans , Immunohistochemistry , Molecular Sequence Data , Neoplasm Staging , Neuregulin-1/pharmacology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Isoforms , Receptor, ErbB-4 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Cells, Cultured
12.
Methods Mol Med ; 39: 145-53, 2001.
Article in English | MEDLINE | ID: mdl-21340765

ABSTRACT

Perhaps the most fundamental question that faces the laboratory scientist is, "Which model system should I use to investigate the problem?" Failure to adequately address this issue can compromise even the most meticulous and inspired research program. If this is such a thorny issue, why use model systems at all? As with most biological systems, ovarian cancer is a complex disorder comprising tumor cells, stromal tissues, neovascularization, inflammatory responses, and other host responses to the tumor. Experimental science best progresses by controling all but a single variable and observing what occurs when that variable is modulated. Almost by definition, this requires a homogenous group of samples to work with. Using human cancer patients for research, it becomes rapidly apparent that the diverse nature of the tumors and the hosts greatly complicates such an approach, hence the development of various model systems. The problem with model systems is simple, they are models-not the true disease states, by their very nature they are less than perfect reflections of the way in which the system under investigation performs in vivo in the normal host. Model systems are essential research tools, but have to be used appropriately.

13.
Methods Mol Med ; 39: 155-9, 2001.
Article in English | MEDLINE | ID: mdl-21340766

ABSTRACT

Human tumor cell lines have provided valuable model systems to study a wide variety of tumor characteristics including the cell biology, genetics, and chemosensitivity profiles of disease. A large number of ovarian cancer cell lines have now been established and are in widespread use Table 1) (1-15). Many of these have been selected to reflect specific situations, e.g., pre- and postchemotherapy models or different histo- logical subtypes. Table 1 Properties of Established Ovarian Carcinoma Cell Lines Prior Cell Line Histology Source Treatment Ref. PE01 P.D. Serous adenoca Ascites P/FU/CHL 1 PE04 P.D. Serous adenoca Ascites P/FU/CHL 1 PE06 P.D. Serous adenoca Ascites P/FU/CHL 2 PEA1 P.D. Adenoca Pleural None 2 PEA2 P.D. Adenoca Ascites P/Pred 2 PE016 P.D. Serous adenoca Ascites Radioth 2 PE014 W.D.Serous adenoca Ascites None 2 T014 W.D.Serous adenoca Sol. Met None 2 PE023 W.D.Serous adenoca Ascites P/CHL 2 SKOV-3 Adenoca Ascites T 3 SW626 Adenoca - - 3 OVCAR-2 Adenoca Ascites P/Cy 4 OVCAR-3 P.D. papillary adenoca Ascites P/Cy/Adr 5 OVCAR-4 Adenoca Ascites P/Cy/Adr 6 OVCAR-5 Adenoca Ascites None 7 OAW 28 Adenoca Ascites P / Mel 8 OAW 42 Serous adenoca Ascites P 8 41M Adenoca Ascites None 9 59M Endometr adenoca Ascites None 8 CH1 Papillary adenoca Ascites P/ JM8 8 138D Serous adenoca Ascites Carb 9 180D Adenoca Ascites P 9 200D Serous adenoca Solid None 9 253D Serous adenoca Ascites Cy/MPA 9 HOC-1 W.D. Serous adenoca Ascites None 10 HOC-7 W.D. Serous adenoca Ascites None 10 CAOV-3 Adenoca Tumour Cy/Adr/FU 10 COLO 110 Serous adenoca Sol. Met None 11 COLO 316 Serous adenoca Pleural None 11 COLO 319 Serous adenoca Ascites None 11 COLO 330 Serous adenoca Ascites Mel/Radiother 11 IGROV1 Adenoca Primary None 12 HTOA W.D. serous adenoca Primary None 13 OV-1063 Papillary adenoca Ascites Cy/Adr/P/HMM 14 DO-s W.D. mucinous adenoca Ascites - 15 P.D. = Poorly differentiated; W.D. = Well differentiated; adenoca = adenocarcinoma; pleural = pleural effusion; Sol.met. = solid metastasis; P = cisplatin; FU = 5-fluorouracil; CHL = chlorambucil; Pred = prednimustine; Radioth = radiotherapy; T = thiotepa; Cy = cyclophosphamide; Adr = adriamycin; Mel = melphalan; Carb = carboplatin; MPA = medroxyprogesterone actetate; HMM = examethylmelamine.

14.
Methods Mol Med ; 39: 193-8, 2001.
Article in English | MEDLINE | ID: mdl-21340772

ABSTRACT

The use of human tumor xenografts grown in immunodeficient animals as a model for human cancers is well established and their value depends on the extent to which their characteristics reflect the properties of a particular cancer in the clinical situation. For endocrine-sensitive tumors, the retention of hormone receptors in xenografts and their responsiveness to hormonal stimuli are essential criteria if they are to serve as appropriate models. Provided these criteria are met, such experimental systems allow detailed studies of the effects of hormones on hormone-sensitive tumor cells and the potential of endocrine therapies in an in vivo system (1).

15.
Methods Mol Med ; 39: 199-203, 2001.
Article in English | MEDLINE | ID: mdl-21340773

ABSTRACT

The fact that human tumor xenografts grown in immunodeficient mice have proven to be useful models of human cancer is well documented. However, the establishment of such xenografts from cell lines cultured in vitro has proven to be fraught with difficulties-these problems become even more apparent when one tries to establish xenografts from primary clinical material (1-3). Nevertheless, the inclusion of Matrigel has been shown to enhance the tumorigenicity of a number of cell lines in vivo (4-10), including those of ovarian origin (11). There are also reports of Matrigel enhancing the tumorigenic potential of breast primary material (12). Furthermore, it has been shown that xenografts established in the presence of Matrigel can be excised, transferred into recipient animals, and subsequently grown in the absence of Matrigel, which suggests that it is required only for the initial establishment of tumors (11). Matrigel is, therefore, extremely useful for establishing a variety of cell lines as routine xenografts, which could otherwise prove difficult to take, as illustrated in Table 1 and Fig. 1 (adapted from [11]).

16.
Ann Oncol ; 10(10): 1233-40, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10586342

ABSTRACT

BACKGROUND: Ecteinascidin-743 (ET743) is a novel antitumour agent originating from the Caribbean tunicate Ecteinascidia turbinata. It has potent cytotoxic and antitumour activity and a potential new mechanism of action. The aim of the present study was to further explore the antitumour activity of ET743 in human tumour xenografts from melanoma, non-small-cell lung and ovarian cancer. DESIGN: As the antitumour profile of ET743 was largely unknown a chemo-sensitive and a marginal chemo-resistant human tumour xenograft were selected for each tumour type. ET743 was administered intravenously using two administration schedules (days 0, 4, 8 and 0-2, 13-15). RESULTS: ET743 was very active at the maximum tolerated dose (MTD) in the chemo-sensitive xenograft melanoma MEXF 989, non-small-cell lung cancer LXFL 529, and ovarian cancers HOC22 and (marginally resistant to cisplatin) HOC18. Activity was also seen at 1/2 MTD. Apart from HOC18, ET743 caused complete remissions in the responding xenografts. The compound was inactive in the chemo-resistant xenograft melanoma MEXF 514 and non-small-cell lung cancer LXFA 629. In terms of antitumour activity the days 0, 4, 8 schedule had advantages over the days 0-2, 13-15 schedule. CONCLUSIONS: ET743 is a very effective agent in chemo-sensitive and marginal chemo-resistant xenografts, but inactive in chemo-resistant tumour xenografts. The activity of ET743 in the marginally cisplatin-resistant ovarian cancer HOC18 might indicate absence or incomplete cross-resistance against cisplatin. It is recommended to include melanoma, non-small-cell lung cancer, and ovarian cancer in phase II clinical trials and to use an intermittent schedule.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Dioxoles/therapeutic use , Isoquinolines/therapeutic use , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Ovarian Neoplasms/drug therapy , Adult , Animals , Antineoplastic Agents, Alkylating/adverse effects , Dioxoles/adverse effects , Female , Humans , Isoquinolines/adverse effects , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Tetrahydroisoquinolines , Trabectedin
17.
Br J Cancer ; 80(5-6): 685-92, 1999 May.
Article in English | MEDLINE | ID: mdl-10360644

ABSTRACT

The extracellular matrix glycoprotein tenascin-C (TN) is overexpressed in the stroma of malignant ovarian tumours particularly at the interface between epithelia and stroma leading to suggestions that it may be involved in the process of invasion (Wilson et al (1996) Br J Cancer 74: 999-1004). To define regulation of TN further and investigate its function in ovarian cancer, a range of cell line models were studied. Concentrations of secreted TN in media from cultures of ovarian fibroblast cell lines were at least 100-fold greater than from carcinoma cell lines. Evidence for paracrine regulation of TN secretion was obtained by co-culture of carcinoma cells with fibroblast cells wherein secretion into the media was greater than from fibroblasts alone. Transforming growth factor (TGF)-beta1, insulin-like growth factor (IGF)-II and progesterone all stimulated TN secretion while human choriogonadotropin (hCG), follicle-stimulating hormone (FSH) and gamma-interferon inhibited secretion. TGF-beta1 produced the greatest stimulation of TN in cultured fibroblasts and its co-expression with TN was examined in primary ovarian tumours. There was a significant association between the presence of moderate-strong expression of TN and TGF-beta1. Evidence for TN having a functional role in ovarian carcinoma was obtained from adhesion and migration assays. The PE01, PE04, SKOV-3 and 59M cell lines all demonstrated marked adhesion to plastic coated with TN relative to the control protein bovine serum albumin (BSA) and expressed alpha2beta1 and alpha3beta1 integrins. The SKOV-3 cell line migrated more rapidly through TN than through BSA indicating that TN can facilitate migration of ovarian carcinoma cells.


Subject(s)
Ovarian Neoplasms/metabolism , Tenascin/physiology , Animals , Cattle , Cell Adhesion/physiology , Cell Movement/physiology , Chorionic Gonadotropin/pharmacology , Coculture Techniques , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Follicle Stimulating Hormone/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Integrins/biosynthesis , Interferon-gamma/pharmacology , Mice , Neoplasm Invasiveness , Ovarian Neoplasms/pathology , Progesterone/pharmacology , Protein Isoforms , Stromal Cells/metabolism , Stromal Cells/pathology , Tenascin/biosynthesis , Tenascin/metabolism , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
18.
Br J Cancer ; 79(7-8): 1098-103, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10098742

ABSTRACT

The modulating effects of the epidermal growth factor (EGF) receptor-specific tyrosine kinase inhibitor ZM 252868 on cell growth and signalling have been evaluated in four ovarian carcinoma cell lines PE01, PE04, SKOV-3 and PE01CDDP. Transforming growth factor alpha (TGF-alpha)-stimulated growth was completely inhibited by concentrations > or =0.3 microM in the PE01 and PE04 cell lines and by > or =0.1 microM in SKOV-3 cells. TGF-alpha inhibition of PE01CDDP growth was reversed by concentrations > or =0.1 microM ZM 252868. TGF-alpha-stimulated tyrosine phosphorylation of both the EGF receptor and c-erbB2 receptor in all four cell lines. The inhibitor ZM 252868, at concentrations > or =0.3 microM, completely inhibited TGF-alpha-stimulated tyrosine phosphorylation of the EGF receptor and reduced phosphorylation of the c-erbB2 protein. EGF-activated EGF receptor tyrosine kinase activity was completely inhibited by 3 microM ZM 252868 in PE01, SKOV-3 and PE01CDDP cells. These data indicate that the EGF receptor-targeted TK inhibitor ZM 252868 can inhibit growth of ovarian carcinoma cells in vitro consistent with inhibition of tyrosine phosphorylation at the EGF receptor.


Subject(s)
Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/drug effects , Ovarian Neoplasms/drug therapy , Quinazolines/pharmacology , Transforming Growth Factor alpha/antagonists & inhibitors , Cell Division/drug effects , ErbB Receptors/metabolism , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/metabolism , Receptor, ErbB-3 , Signal Transduction , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured/drug effects
19.
Eur J Cancer ; 35(9): 1381-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10658531

ABSTRACT

Previous studies have demonstrated that endothelin (ET) isoforms (ET-1, ET-2 and ET-3) can act in an autocrine manner in ovarian cancer while in breast cancer their role has been proposed to be that of a paracrine mitogen. To explore the possibility that endothelin isoforms might function not only as autocrine regulators but also as paracrine mitogens in ovarian cancers, we investigated their effects on the growth of ovarian fibroblasts derived from ovarian carcinomas, the interaction between ovarian carcinoma and fibroblast cells and the location of the isoform expression in primary ovarian tumours. ET-1, ET-2 and ET-3 stimulated the growth of three ovarian fibroblast cell lines at concentrations ranging from 10(-12) M to 10(-7) M. Inhibition of 125I-ET binding by the ETA receptor antagonist BQ123 and the ETB receptor antagonist BQ788 suggested the presence of both types of ET receptors in fibroblast cells. In the absence of ET-1, neither BQ 123 nor BQ 788 inhibited growth. However, both antagonists inhibited ET-1 stimulated growth suggesting the involvement of both receptor types in ET-1 growth regulation. In contrast to carcinoma cells which secrete measurable levels of ET-1, fibroblast cell lines did not secrete detectable protein. Co-culture experiments (using porous membrane insert wells) of fibroblasts with carcinoma cells demonstrated that growth of both populations of cells was increased compared with either grown in isolation. In this system, growth of the fibroblast cell line was partially inhibited by both BQ123 and BQ788, whilst growth of the PE014 carcinoma cell line was inhibited by only BQ123. RT-PCR measurements detected the presence of the ETA receptor subtype in 10/10 primary ovarian cancers but the presence of ETB receptor in only 6/10 cancers. Using specific antibodies, ET-1 was found in 11/15, ET-2 in 5 of 7 and ET-3 in 5/7 primary ovarian cancers predominantly in the epithelial cells but with some stromal expression. These data indicate that the ET isoforms may stimulate growth of the fibroblast population within an ovarian cancer in addition to stimulating the epithelial cells and since the ETs are expressed in the majority of ovarian cancers, this paracrine effect may contribute to the overall growth of the tumour.


Subject(s)
Endothelins/physiology , Ovarian Neoplasms/pathology , Cell Division , Endothelin Receptor Antagonists , Endothelin-1/physiology , Endothelin-2/physiology , Endothelin-3/physiology , Female , Fibroblasts/pathology , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Cells, Cultured
20.
Cancer Chemother Pharmacol ; 42(5): 429-32, 1998.
Article in English | MEDLINE | ID: mdl-9771960

ABSTRACT

The activity of CD437¿6-[3-(1-adamantyl)-4 hydroxyphenyl]-2-naphthalene carboxylic acid¿, a relatively selective activator of RAR-gamma, was evaluated against four human ovarian-carcinoma cell lines : PE01, PE04 (a Pt-resistant in vivo-derived counterpart of PE01), PE01CDDP (a Pt-resistant in vitro-derived model of PE01) and PE014. Growth inhibition was observed after 3 and 6 days of exposure to sub-micromolar concentrations as assessed by a reduction in cell number. IC50 values against PE01, PE04, PE01CDDP and PE014 were 0.09, 0.21, 0.12 and 0.28 microM (day 3) and 0.1, 0.14, 0.07 and 0.17 microM (day 6), respectively. Cisplatin-resistant cell lines were as responsive as cisplatin-sensitive lines, indicating potential activity in resistant disease. CD437 was also evaluated against the PE04 xenograft grown in nude mice using daily doses of 20 (days 0-4) and 10 mg/kg (days 0-4 and 7-11) given either by i.p. delivery or oral administration. Significant growth inhibition (P < 0.05) was obtained for both doses and by both routes. These data provide further support for the view that retinoids have value for the treatment of ovarian cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Ovarian Neoplasms/drug therapy , Retinoids/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma/pathology , Cell Division/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Injections, Intraperitoneal , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Retinoids/administration & dosage , Retinoids/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...