Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
J Mech Behav Biomed Mater ; 60: 401-415, 2016 07.
Article in English | MEDLINE | ID: mdl-26974584

ABSTRACT

Mechanical characterisation of soft biological tissues using standard compression or tensile testing presents a significant challenge due to specimen geometrical irregularities, difficulties in cutting intact and appropriately sized test samples, and issues with slippage or damage at the grips. Indentation can overcome these problems but requires fitting a model to the resulting load-displacement data in order to calculate moduli. Despite the widespread use of this technique, few studies experimentally validate their chosen model or compensate for boundary effects. In this study, viscoelastic hydrogels of different concentrations and dimensions were used to calibrate an indentation technique performed at large specimen-strain deformation (20%) and analysed with a range of routinely used mathematical models. A rigid, flat-ended cylindrical indenter was applied to each specimen from which 'indentation moduli' and relaxation properties were calculated and compared against values obtained from unconfined compression. Only one indentation model showed good agreement (<10% difference) with all moduli values obtained from compression. A sample thickness to indenter diameter ratio ≥1:1 and sample diameter to indenter diameter ratio ≥4:1 was necessary to achieve the greatest accuracy. However, it is not always possible to use biological samples within these limits, therefore we developed a series of correction factors. The approach was validated using human diseased omentum and bovine articular cartilage resulting in mechanical properties closely matching compression values. We therefore present a widely useable indentation analysis method to allow more accurate calculation of material mechanics which is important in the study of soft tissue development, ageing, health and disease.


Subject(s)
Cartilage, Articular/pathology , Hydrogels , Animals , Calibration , Cattle , Elasticity , Humans , Models, Biological , Pressure , Stress, Mechanical
2.
Oncogene ; 31(48): 4987-95, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-22266861

ABSTRACT

Early genetic events in the development of high-grade serous ovarian cancer (HGSOC) may define the molecular basis of the profound structural and numerical instability of chromosomes in this disease. To discover candidate genetic changes we sequentially passaged cells from a karyotypically normal hTERT immortalised human ovarian surface epithelial line (IOSE25) resulting in the spontaneous formation of colonies in soft agar. Cell lines transformed ovarian surface epithelium 1 and 4 (TOSE 1 and 4) established from these colonies had an abnormal karyotype and altered morphology, but were not tumourigenic in immunodeficient mice. TOSE cells showed loss of heterozygosity (LOH) at TP53, increased nuclear p53 immunoreactivity and altered expression profile of p53 target genes. The parental IOSE25 cells contained a missense, heterozygous R175H mutation in TP53, whereas TOSE cells had LOH at the TP53 locus with a new R273H mutation at the previous wild-type TP53 allele. Cytogenetic and array CGH analysis of TOSE cells also revealed a focal genomic amplification of CXCR4, a chemokine receptor commonly expressed by HGSOC cells. TOSE cells had increased functional CXCR4 protein and its abrogation reduced epidermal growth factor receptor (EGFR) expression, as well as colony size and number. The CXCR4 ligand, CXCL12, was epigenetically silenced in TOSE cells and its forced expression increased TOSE colony size. TOSE cells had other cytogenetic changes typical of those seen in HGSOC ovarian cancer cell lines and biopsies. In addition, enrichment of CXCR4 pathway in expression profiles from HGSOC correlated with enrichment of a mutated TP53 gene expression signature and of EGFR pathway genes. Our data suggest that mutations in TP53 and amplification of the CXCR4 gene locus may be early events in the development of HGSOC, and associated with chromosomal instability.


Subject(s)
Cell Transformation, Neoplastic/genetics , Ovary/cytology , Receptors, CXCR4/genetics , Tumor Suppressor Protein p53/genetics , Animals , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Humans , Karyotyping , Loss of Heterozygosity , Mice , Ovary/metabolism , RNA, Messenger
3.
Br J Cancer ; 102(11): 1555-77, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20502460

ABSTRACT

Animal experiments remain essential to understand the fundamental mechanisms underpinning malignancy and to discover improved methods to prevent, diagnose and treat cancer. Excellent standards of animal care are fully consistent with the conduct of high quality cancer research. Here we provide updated guidelines on the welfare and use of animals in cancer research. All experiments should incorporate the 3Rs: replacement, reduction and refinement. Focusing on animal welfare, we present recommendations on all aspects of cancer research, including: study design, statistics and pilot studies; choice of tumour models (e.g., genetically engineered, orthotopic and metastatic); therapy (including drugs and radiation); imaging (covering techniques, anaesthesia and restraint); humane endpoints (including tumour burden and site); and publication of best practice.


Subject(s)
Animal Experimentation/standards , Animal Welfare/standards , Neoplasms/pathology , Neoplasms/therapy , Practice Guidelines as Topic , Algorithms , Animal Experimentation/ethics , Animal Welfare/ethics , Animal Welfare/organization & administration , Animals , Biomarkers, Pharmacological/analysis , Biomedical Research/ethics , Biomedical Research/legislation & jurisprudence , Biomedical Research/organization & administration , Biomedical Research/standards , Cell Line, Transformed , Diagnostic Imaging , Disease Models, Animal , Female , Humans , Male , Mice , Neoplasm Transplantation/methods , Neoplasm Transplantation/pathology , Neoplasm Transplantation/standards , Neoplasms/diagnosis , Neoplasms/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Clin Pharmacol Ther ; 87(4): 401-6, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200512

ABSTRACT

Smoldering, nonresolving inflammation is a component of the tumor microenvironment. The linkage between inflammation and cancer, first perceived in the nineteenth century, is now part of an accepted paradigm of carcinogenesis.


Subject(s)
Drug Delivery Systems , Inflammation/complications , Neoplasms/etiology , Animals , Anti-Inflammatory Agents/pharmacology , Clinical Trials as Topic , Cytokines/metabolism , Drug Evaluation, Preclinical , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Neoplasms/physiopathology , Neoplasms/prevention & control
5.
Oncogene ; 28(5): 773-80, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-18997822

ABSTRACT

The receptor for macrophage colony-stimulating factor 1 receptor (CSF1R) is a product of the proto-oncogene c-fms and a member of the class III transmembrane tyrosine kinase receptor family. Earlier, we described increased mRNA expression of CSF1R in human telomerase reverse transcriptase (hTERT) immortalized human ovarian surface epithelial (IOSE) cell lines derived from a single donor. Here, we further describe that CSF1R is upregulated at both the mRNA and protein level in hTERT immortalized human normal OSE cells from two different donors and in hTERT immortalized human pancreatic ductal epithelial cells. CSF1R was not upregulated in hTERT immortalized epithelial clones that subsequently underwent senescence or in immortalized fibroblasts. Upon stimulation by the CSF1R ligand CSF1, the immortalized epithelial cell lines showed rapid internalization of CSF1R with concomitant down-modulation and colocalization of phosphorylated NFkappaBp65 with hTERT protein, hTERT translocation into the nucleus and the binding of c-Myc to the hTERT promoter region. Reducing the expression of CSF1R using short hairpin interfering RNA abolished these effects and also decreased cell survival and the number of population doublings under suboptimal culture conditions. The telomerase inhibitor GRN163L confirmed a role for telomerase in the cleavage of the intracellular domain of CSF1R. On the basis of these findings, we suggest that CSF1R may be a critical factor facilitating hTERT immortalization of epithelial cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial Cells/pathology , Receptor, Macrophage Colony-Stimulating Factor/physiology , Telomerase/genetics , Cell Line, Transformed , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Proto-Oncogene Mas , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Telomerase/metabolism , Transfection , Up-Regulation/drug effects
6.
Ann Oncol ; 19(7): 1340-1346, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18325912

ABSTRACT

BACKGROUND: Tumour necrosis factor-alpha (TNF-alpha) is an important regulator of the chronic inflammation contributing to tumour progression. Infliximab, an anti-TNF-alpha monoclonal antibody was investigated in this trial of patients with advanced cancer. The primary objectives were to determine the safety profile and biological response of infliximab in a cancer population. Clinical response was a secondary objective. PATIENTS AND METHODS: Forty-one patients received infliximab at 5 mg/kg (n = 21) or 10 mg/kg (n = 20) i.v. at 0 and 2 weeks and then every 4 weeks. Post-treatment samples were measured for changes in plasma and serum TNF-alpha, CCL2, IL-6 and C-reactive protein (CRP). RESULTS: Infliximab was well tolerated with no dose-limiting toxic effects. At both doses of infliximab, neutralisation of serum TNF-alpha was observed after 1 h while plasma CCL2, IL-6 and serum CRP were decreased 24 and 48 h following infliximab administration. Seven patients experienced disease stablisation (range 10-50+ weeks). There was no evidence of disease acceleration in any patient. CONCLUSIONS: Infliximab treatment was safe and well tolerated in patients with advanced cancer. There was evidence of biological activity with baseline TNF-alpha and CCL2 being correlated with infliximab response.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Drug Hypersensitivity , Hypersensitivity, Delayed , Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , C-Reactive Protein/analysis , Chemokine CCL2/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Hypersensitivity, Delayed/chemically induced , Infliximab , Infusions, Intravenous , Interleukin-6/blood , Linear Models , Male , Middle Aged , Neoplasms/blood , Neoplasms/pathology , Sensitivity and Specificity , Stomatitis/chemically induced , Treatment Outcome , Tumor Necrosis Factor-alpha/blood
7.
Cell Prolif ; 40(5): 780-94, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17877616

ABSTRACT

OBJECTIVE: Cell immortalization is considered to be a prerequisite status for carcinogenesis. Normal human ovarian surface epithelial (OSE) cells, which are thought to be the origin of most of human ovarian carcinomas, have a very limited lifespan in culture. Establishment of immortalized OSE cell lines has, in the past, required inactivation of pRb and p53 functions. However, this often leads to increased chromosome instability during prolonged culture. MATERIALS AND METHODS: In this study, we have used a retroviral infection method to overexpress human telomerase reverse transcriptase (hTERT) gene, in primary normal OSE cells, under optimized culture conditions. RESULTS: In vitro and in vivo analysis of hTERT-immortalized cell lines confirmed their normal epithelial characteristics. Gene expression profiles and functional analysis of p16(INK4A), p15(INK4B), pRb and p53 confirmed the presence of their intact functions. Our study suggests that inactivation of pRb and p53 is not necessary for OSE immortalization. Furthermore, down-regulation of p15(INK4B) in the immortalized cells may indicate a functional role for this protein in them. CONCLUSION: These immortal OSE cell lines are likely to be an important tool for studying human OSE biology and carcinogenesis.


Subject(s)
Ovary/cytology , Ovary/metabolism , Retinoblastoma Protein/metabolism , Telomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Antigens, CD , Cadherins/genetics , Cell Cycle , Cell Line , Cyclin-Dependent Kinase Inhibitor p15/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Genes, Retinoblastoma , Genes, p53 , Humans , In Situ Hybridization, Fluorescence , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/genetics
8.
Br J Cancer ; 91(1): 30-6, 2004 Jul 05.
Article in English | MEDLINE | ID: mdl-15162145

ABSTRACT

Marimastat, low molecular weight heparins and captopril have antiangiogenic activity in vitro and in animal models. We studied the safety and efficacy of the combination of these drugs in patients with advanced cancer. In all, 50 patients were enrolled. Captopril was given orally at a dose of 50 mg bd daily. Fragmin was administered as a daily subcutaneous injection of 200 units kg(-1) for the first 28 days and 5000 units thereafter. Marimastat was given at 10 mg bd orally. Serum, plasma and urinary angiogenic factors were measured at baseline and after 1 month of treatment. Inhibition of release of tumour necrosis factor alpha (TNF-alpha) from peripheral lymphocytes was used as a surrogate pharmacodynamic end point. There was one case of haemorrhagic stroke and one upper gastrointestinal haemorrhage. The commonest toxicity was myalgia. One of 10 patients with renal cancer had a partial response, and three patients had a prolonged period of stable disease. The treatment significantly inhibited phytohaemagglutinin (PHA)-stimulated TNF-alpha release from patient's lymphocytes. The combination of marimastat, fragmin and captopril is well tolerated and has in vivo activity. Inhibition of PHA-stimulated TNF-alpha release from lymphocytes is a surrogate pharmacodynamic marker of metalloprotease inhibition.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Captopril/pharmacology , Dalteparin/pharmacology , Enzyme Inhibitors/pharmacology , Fibrinolytic Agents/pharmacology , Hydroxamic Acids/pharmacology , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Biomarkers , Captopril/administration & dosage , Dalteparin/administration & dosage , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Female , Fibrinolytic Agents/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Injections, Subcutaneous , Lymphocytes/physiology , Male , Middle Aged , Phytohemagglutinins/analysis , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
9.
Br J Cancer ; 89(8): 1418-22, 2003 Oct 20.
Article in English | MEDLINE | ID: mdl-14562010

ABSTRACT

Protein kinase C (PKC) has a critical role in several signal transduction pathways, and is involved in renal cancer pathogenesis. Bryostatin-1 modulates PKC activity and has antitumour effects in preclinical studies. We conducted a multicentre phase II clinical trial in patients with advanced renal cancer to determine the response rate, immunomodulatory activity and toxicity of bryostatin-1 given as a continuous 24 h infusion weekly for 3 out of 4 weeks at a dose of 25 mug m(-2). In all, 16 patients were recruited (11 males and five females). The median age was 59 years (range 44-68). Patients had been treated previously with nephrectomy (8) and/or interferon therapy (9) and/or hormone therapy (4) and/or radiotherapy (6). Eight, five and three patients had performance statuses of 0, 1 and 2, respectively. A total of 181 infusions were administered with a median of 12 infusions per patient (range 1-29). Disease response was evaluable in 13 patients. Three patients achieved stable disease lasting for 10.5, 8 and 5.5 months, respectively. No complete responses or partial responses were seen. Myalgia, fatigue, nausea, headache, vomiting, anorexia, anaemia and lymphopenia were the commonly reported side effects. Assessment of biological activity of bryostatin-1 was carried out using the whole-blood cytokine release assay in six patients, two of whom had a rise in IL-6 levels 24 h after initiating bryostatin-1 therapy compared to pretreatment values. However, the IL-6 level was found to be significantly lower at day 28 compared to the pretreatment level in all six patients analysed.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Lactones/pharmacology , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Bryostatins , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/surgery , Female , Humans , Infusions, Intravenous , Interleukin-6/blood , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Lactones/administration & dosage , Lactones/adverse effects , Macrolides , Male , Middle Aged , Treatment Outcome
10.
Eur J Cancer ; 39(13): 1818-27, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12932658

ABSTRACT

In March 2003, an international mulltidisciplinary group of scientists and clinicians with a specific interest in ovarian cancer met for 4 days to discuss research into and treatment of this challenging disease. Under the headings of molecular genetics, molecular biology, the biology of ovarian cancer, old therapies, new targets and the early detection of the disease, this Position Paper summarises the presentations and discussion from the 9th Biennial Helene Harris Memorial Trust Forum on Ovarian Cancer. In particular, we highlight the potential of international collaborations in translating laboratory science into useful clinical interventions.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Ovarian Neoplasms/therapy , Biomarkers, Tumor , Female , Forecasting , Gene Expression , Genes, BRCA1 , Genes, BRCA2 , Genes, Tumor Suppressor , Humans , Mutation/genetics , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Randomized Controlled Trials as Topic
12.
Br J Cancer ; 85(6): 891-7, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11556842

ABSTRACT

To understand the chemokine network in a tissue, both chemokine and chemokine receptor expression should be studied. Human epithelial ovarian tumours express a range of chemokines but little is known about the expression and localisation of chemokine receptors. With the aim of understanding chemokine action in this cancer, we investigated receptors for CC-chemokines and their ligands in 25 biopsies of human ovarian cancer. CC-chemokine receptor mRNA was generally absent from solid tumours, the exception being CCR1 which was detected in samples from 75% of patients. CCR1 mRNA localised to macrophages and lymphocytes and there was a correlation between numbers of CD8(+) and CCR1 expressing cells (P = 0.031). mRNA for 6 CC-chemokines was expressed in a majority of tumour samples. In a monocytic cell line in vitro, we found that CCR1 mRNA expression was increased 5-fold by hypoxia. We suggest that the CC-chemokine network in ovarian cancer is controlled at the level of CC-chemokine receptors and this may account for the phenotypes of infiltrating cells found in these tumours. The leukocyte infiltrate may contribute to tumour growth and spread by providing growth survival factors and matrix metalloproteases. Thus, CCR1 may be a novel therapeutic target in ovarian cancer.


Subject(s)
Chemokines, CC/genetics , Ovarian Neoplasms/metabolism , Receptors, Chemokine/genetics , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Base Sequence , Blotting, Northern , CD8-Positive T-Lymphocytes/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/metabolism , Carcinoma, Signet Ring Cell/pathology , Chemokines, CC/metabolism , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , In Situ Hybridization , Molecular Sequence Data , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Messenger/metabolism , Receptors, CCR1 , Receptors, Chemokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/metabolism
13.
Cancer Res ; 61(13): 4961-5, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431324

ABSTRACT

We investigated the possibility that chemokine gradients influence migration of human ovarian epithelial tumor cells. Of 14 chemokine receptors investigated, only CXCR4 was expressed on ovarian cancer cells. CXCR4 mRNA localized to a subpopulation of tumor cells in ovarian cancer biopsies. Ovarian cancer cell lines and cells freshly isolated from ascites expressed CXCR4 protein. The CXCR4 ligand, CXCL12, was found in ascites from 63 patients. CXCL12 elicited intracellular calcium flux and directed migration and changes in integrin expression in ovarian cancer cells. CXCR4 may influence cell migration in the peritoneum, a major route for ovarian cancer spread, and could be a therapeutic target.


Subject(s)
Cell Movement/physiology , Ovarian Neoplasms/pathology , Receptors, CXCR4/physiology , Ascitic Fluid/metabolism , Chemokine CXCL12 , Chemokines, CXC/metabolism , Chemokines, CXC/physiology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Stromal Cells/metabolism , Tumor Cells, Cultured
14.
Lancet ; 357(9255): 539-45, 2001 Feb 17.
Article in English | MEDLINE | ID: mdl-11229684

ABSTRACT

The response of the body to a cancer is not a unique mechanism but has many parallels with inflammation and wound healing. This article reviews the links between cancer and inflammation and discusses the implications of these links for cancer prevention and treatment. We suggest that the inflammatory cells and cytokines found in tumours are more likely to contribute to tumour growth, progression, and immunosuppression than they are to mount an effective host antitumour response. Moreover cancer susceptibility and severity may be associated with functional polymorphisms of inflammatory cytokine genes, and deletion or inhibition of inflammatory cytokines inhibits development of experimental cancer. If genetic damage is the "match that lights the fire" of cancer, some types of inflammation may provide the "fuel that feeds the flames". Over the past ten years information about the cytokine and chemokine network has led to development of a range of cytokine/chemokine antagonists targeted at inflammatory and allergic diseases. The first of these to enter the clinic, tumour necrosis factor antagonists, have shown encouraging efficacy. In this article we have provided a rationale for the use of cytokine and chemokine blockade, and further investigation of non-steroidal anti-inflammatory drugs, in the chemoprevention and treatment of malignant diseases.


Subject(s)
Inflammation/complications , Neoplasms/etiology , Animals , Antineoplastic Agents/therapeutic use , Cytokines/genetics , Cytokines/physiology , Humans , Immunity, Cellular , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/prevention & control , Wound Healing
15.
J Clin Oncol ; 19(5): 1485-92, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11230495

ABSTRACT

PURPOSE: N-Benzoyl staurosporine (PKC412) is a protein kinase C inhibitor with antitumor activity in laboratory models. We determined the toxicity of oral PKC412 administered daily for repeat cycles of 28 days. PATIENTS AND METHODS: Thirty-two patients with advanced solid cancers were treated at seven dose levels (12.5 to 300 mg daily) for a total of 68 cycles. RESULTS: The most frequent treatment-related toxicities were nausea, vomiting, fatigue, and diarrhea. At the two top dose levels (225 and 300 mg/d), 15 of 16 patients experienced nausea/vomiting (common toxicity criteria [CTC], version 1), grade 2 in nine of 16 and grade 3 in three of 16 patients; and six of 16 patients developed CTC grade 2 diarrhea. After 1 month of treatment, there were significant reductions in circulating lymphocyte (P <.02) and monocyte (P <.01) counts in patients receiving doses > or = 100 mg/d. Nevertheless, only two patients developed myelosuppression (both grade 2). Of two patients with progressive cholangiocarcinoma, one attained stable disease lasting 4.5 months and one a partial response lasting 4 months. There was a linear relationship between PKC412 dose and area under the curve (0-24 hours) and maximum plasma concentration with marked interpatient variability. The estimated median elimination half-life was 1.6 days (range, 0.9 to 4.0 days), and a metabolite with a median half-life of 36 days was detected. Steady-state PKC412 plasma levels at the top three dose cohorts (150 to 300 mg) were five to 10 times the cellular 50% inhibitory concentration for PKC412 of 0.2 to 0.7 micromol/L. CONCLUSION: PKC412 can be safely administered by chronic oral therapy, and 150 mg/d is suitable for phase II studies. The pharmacokinetics and lack of conventional toxicity indicate that pharmacodynamic measures may be additionally needed to optimize the drug dose and schedule.


Subject(s)
Enzyme Inhibitors/adverse effects , Neoplasms/drug therapy , Staurosporine/analogs & derivatives , Staurosporine/adverse effects , Administration, Oral , Adult , Aged , Area Under Curve , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Male , Middle Aged , Staurosporine/administration & dosage , Staurosporine/pharmacokinetics
16.
Eur J Immunol ; 31(2): 480-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11180113

ABSTRACT

Macrophages accumulate in areas of inflammation and necrosis that are likely to be hypoxic. Chemotaxis of monocytes and macrophages towards chemokines is rapidly (within 60-90 min) inhibited by hypoxia. Exposure to the inflammatory cytokine TNF-alpha has a similar effect on monocyte migration. We report here that neither changes in mitochondrial respiration nor intracellular pH are involved in migration arrest. However, hypoxic inhibition of migration was mimicked using chemical activators of hypoxia-inducible factor-1 and reversed by transcriptional inhibition. We used RNA arbitrarily primed PCR, a differential display technique, to investigate which genes were up-regulated within 90-min exposure to hypoxia. Of several thousand mRNA screened, only one was consistently up-regulated by hypoxia and this was identified as MAPK phosphatase 1 (MKP-1), which modulates MAPK activity. Levels of MKP-1 mRNA and protein were rapidly elevated in monocytic cells and primary macrophages after hypoxia or TNF-alpha treatment. The functional significance of MKP-1 was illustrated by hypoxia-induced decreases in phosphorylated MAPK in these cells and arrest of chemotaxis by MAPK inhibitors. We suggest that one of the important events in an 'emergency stop' response in monocytic cells and macrophages may be inhibition of the chemoattractant signaling cascade.


Subject(s)
Cell Cycle Proteins , Cell Hypoxia , Chemotaxis , Inflammation/immunology , Macrophages/physiology , Monocytes/physiology , Phosphoprotein Phosphatases , Catalase/pharmacology , Dactinomycin/pharmacology , Deferoxamine/pharmacology , Dual Specificity Phosphatase 1 , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Mitogen-Activated Protein Kinase Kinases/physiology , Mitogen-Activated Protein Kinases/metabolism , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/biosynthesis , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/physiology
18.
Br J Cancer ; 83(11): 1538-43, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11076665

ABSTRACT

The study of treatment-induced changes in the tumour microenvironment might lead to effective combinations of biological therapy. IL-12 induced tumour regression and cure of an experimental murine breast cancer, HTH-K, but only after long-term treatment that was associated with chronic toxicity. During IL-12 therapy, tumour levels of the matrix metalloprotease MMP-9 declined and its inhibitor TIMP-1 was strongly induced. We therefore administered alternate cycles of IL-12 and the MMP inhibitor Batimastat (BB94) to mice. Therapeutic efficacy was increased compared with short-term IL-12 therapy but without the chronic toxicity associated with long-term IL-12 treatment. Image analysis of treated tumours revealed that BB94 prevented regeneration of tumour and stromal compartments that normally occurred after short-term IL-12 therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Matrix Metalloproteinase Inhibitors , Phenylalanine/analogs & derivatives , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Disease Models, Animal , Drug Administration Schedule , Drug Synergism , Female , Interleukin-12/administration & dosage , Interleukin-12/pharmacology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/enzymology , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/prevention & control , Phenylalanine/administration & dosage , Phenylalanine/pharmacology , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacology , Thiophenes/administration & dosage , Thiophenes/pharmacology
19.
J Immunol ; 164(2): 733-8, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10623817

ABSTRACT

Monocyte chemotactic protein-1 (MCP-1, CCL2) is an important determinant of macrophage infiltration in tumors, ovarian carcinoma in particular. MCP-1 binds the chemokine receptor CCR2. Recent results indicate that proinflammatory and anti-inflammatory signals regulate chemokine receptor expression in monocytes. The present study was designed to investigate the expression of CCR2 in tumor-associated macrophages (TAM) from ovarian cancer patients. TAM isolated from ascitic or solid ovarian carcinoma displayed defective CCR2 mRNA (Northern blot and PCR) and surface expression and did not migrate in response to MCP-1. The defect was selective for CCR2 in that CCR1 and CCR5 were expressed normally in TAM. CCR2 gene expression and chemotactic response to MCP-1 were decreased to a lesser extent in blood monocytes from cancer patients. CCR2 mRNA levels and the chemotactic response to MCP-1 were drastically reduced in fresh monocytes cultured in the presence of tumor ascites from cancer patients. Ab against TNF-alpha restored the CCR2 mRNA level in monocytes cultured in the presence of ascitic fluid. The finding of defective CCR2 expression in TAM, largely dependent on local TNF production, is consistent with previous in vitro data on down-regulation of chemokine receptors by proinflammatory molecules. Receptor inhibition may serve as a mechanism to arrest and retain recruited macrophages and to prevent chemokine scavenging by mononuclear phagocytes at sites of inflammation and tumor growth. In the presence of advanced tumors or chronic inflammation, systemic down-regulation of receptor expression by proinflammatory molecules leaking in the systemic circulation may account for defective chemotaxis and a defective capacity to mount inflammatory responses associated with advanced neoplasia.


Subject(s)
Chemokine CCL2/metabolism , Macrophages/metabolism , Ovarian Neoplasms/immunology , Receptors, Chemokine/biosynthesis , Receptors, Cytokine/biosynthesis , Ascites/immunology , Ascites/pathology , Carcinoma/immunology , Carcinoma/metabolism , Carcinoma/pathology , Cell Movement/immunology , Chemokine CCL2/physiology , Female , Humans , Macrophages/immunology , Monocytes/immunology , Monocytes/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA, Messenger/biosynthesis , Receptors, CCR2 , Receptors, Chemokine/genetics , Receptors, Cytokine/genetics , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...