Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Cancer Educ ; 37(4): 1228-1235, 2022 08.
Article in English | MEDLINE | ID: mdl-33523406

ABSTRACT

For more than two decades, the International Summer School Oncology for Medical Students (ISOMS) has organized a biennial 2-week international summer school program in Groningen, the Netherlands. The summer school aims to increase knowledge about general cancer care, reduce fear of talking to cancer patients, and expose students to cancer-related problems. After 22 years, there was a need to improve the summer school format, the application procedure, and the intensity of the course. Here, we describe and evaluate these and additional changes that were made to the program. Several changes were made to the summer school format. The course was shortened from 10 days to a more intensive 7 days. The scientific program was integrated with the clinical program and students were taught scientific writing and presentation skills. The application process involved a personal video pitch. Importantly, the new summer school format was organized by a committee in which medical students had the lead. To evaluate the changes to the summer school, we conducted knowledge tests and regularly obtained feedback. There was a high overall student satisfaction, with a median score of a 9 out of 10. Students appreciated the interactive sessions and practicals and the scientific program, and were satisfied with the course level. All students had improved test scores. Improvement points highlighted the need for a less packed schedule and more lectures on basic oncology principles, or were related to specific lectures. The student-led innovation and adaptation of the ISOMS has been successful.


Subject(s)
Neoplasms , Students, Medical , Curriculum , Humans , Medical Oncology/education , Neoplasms/therapy , Netherlands , Schools
2.
Acta Neurochir (Wien) ; 160(8): 1539-1546, 2018 08.
Article in English | MEDLINE | ID: mdl-29858948

ABSTRACT

BACKGROUND: Meningiomas are the most frequently occurring primary intracranial tumours in adults. Surgical removal can only be curative by complete resection; however surgical access can be challenging due to anatomical localization and local invasion of bone and soft tissues. Several intraoperative techniques have been tried to improve surgical resection, including intraoperative fluorescence guided imaging; however, no meningioma-specific (fluorescent) targeting has been developed yet. Here, we aimed to identify the most promising biomarkers for targeted intra-operative fluorescence guided meningioma surgery. METHODS: One hundred forty-eight meningioma specimens representing all meningioma grades were analysed using immunohistochemistry (IHC) on tissue microarrays (TMAs) to determine expression patterns of meningioma biomarkers epithelial membrane antigen (EMA), platelet-derived growth factor ß (PDGF-ß), vascular endothelial growth factor α (VEGF-α), and somatostatin receptor type 2 (SSTR-2). Subsequently, the most promising biomarker was selected based on TArget Selection Criteria (TASC). Marker expression was examined by IHC in 3D cell culture models generated from freshly resected tumour material. RESULTS: TMA-IHC showed strongest staining for SSTR-2. All cases were positive, with 51.4% strong/diffuse, 30.4% moderate/diffuse and only 18.2% focal/weak staining patterns. All tested biomarkers showed at least weak positivity in all meningiomas, regardless of WHO grade. TASC analysis showed that SSTR-2 was the most promising target for fluorescence guided imaging, with a total score of 21 (out of 22). SSTR-2 expression was determined on original patient tumours and 3D cultures of three established cultures. CONCLUSIONS: SSTR-2 expression was highly sensitive and specific in all 148 meningiomas, regardless of WHO grade. According to TASC analysis, SSTR-2 is the most promising receptor for meningioma targeting. After establishing in vitro meningioma models, SSTR-2 cell membrane expression was confirmed in two of three meningioma cultures as well. This indicates that specific fluorescence in an experimental setting can be performed for the further development of targeted fluorescence guided meningioma surgery and near-infrared fluorescent tracers targeting SSTR-2.


Subject(s)
Biomarkers, Tumor/metabolism , Meningeal Neoplasms/surgery , Meningioma/surgery , Neurosurgical Procedures/methods , Receptors, Somatostatin/metabolism , Surgery, Computer-Assisted/methods , Adult , Aged , Biomarkers, Tumor/genetics , Female , Humans , Male , Middle Aged , Receptors, Somatostatin/genetics
3.
J Neurooncol ; 123(1): 53-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25894595

ABSTRACT

Formyl peptide receptor 1 (FPR1) activity in U87 glioblastoma (GBM) cells contributes to tumor cell motility. The present study aimed to evaluate the FPR1 expression in human GBM, the possibility to elicit agonist induced FPR1 activation of GBM cells and inhibit this activation with chemotaxis inhibitory protein of Staphylococcus aureus (CHIPS). Immunohistochemistry was used to assess FPR1 expression in GBM patient samples, which was present in all 178 samples. Also FPR1 mRNA levels measured with quantitative PCR, could be detected in all 25 GBM patient samples tested. Activation of FPR1 in U87 cells, as measured by human mitochondrial-derived agonists, increased calcium mobilization, AKT and ERK1/2 phosphorylation, and ligand-induced migration. Inhibition of all responses could be achieved with CHIPS. Eight early passage human Groningen Glioma (GG) cell lines, isolated from primary GBM tissue were screened for the presence of FPR1. FPR1 mRNA and protein expression as well as receptor activation could not be detected in any of these early passage GG cell lines. However FPR1 was present in ex vivo tumors formed by the same GG cell lines after being implanted in mouse brains. FPR1 is highly expressed in human GBM specimens, it can be activated by human mitochondrial-derived agonists in U87 and inhibited with CHIPS. FPR1 cannot be detected in early passage GG cell lines in vitro, however when engrafted in the mouse brain these cells show FPR1 expression. These results suggest a role of the brain microenvironment in FPR1 expression in GBM.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Interleukin-2/physiology , Receptors, Formyl Peptide/metabolism , Tumor Microenvironment , Animals , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Fluorescent Antibody Technique , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Formyl Peptide/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
4.
Cell Death Dis ; 5: e1443, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25275602

ABSTRACT

Different molecular subtypes of glioblastoma (GBM) have been recently identified, of which the mesenchymal subtype is associated with worst prognoses. Here, we report that transforming growth factor-ß (TGF-ß) is able to induce a mesenchymal phenotype in GBM that involves activation of SMAD2 and ZEB1, a known transcriptional inducer of mesenchymal transition in epithelial cancers. TGF-ß exposure of established and newly generated GBM cell lines was associated with morphological changes, enhanced mesenchymal marker expression, migration and invasion in vitro and in an orthotopic mouse model. TGF-ß-induced mesenchymal differentiation and invasive behavior was prevented by chemical inhibition of TGF-ß signaling as well as small interfering RNA (siRNA)-dependent silencing of ZEB1. Furthermore, TGF-ß-responding and -nonresponding GBM neurospheres were identified in vitro. Interestingly, nonresponding cells displayed already high levels of pSMAD2 and ZEB1 that could not be suppressed by inhibition of TGF-ß signaling, suggesting the involvement of yet unknown mechanisms. These different GBM neurospheres formed invasive tumors in mice as well as revealed mesenchymal marker expression in immunohistochemical analyses. Moreover, we also detected distinct zones with overlapping pSMAD2, elevated ZEB1 and mesenchymal marker expression in GBM patient material, suggestive of the induction of local, microenvironment-dependent mesenchymal differentiation. Overall, our findings indicate that GBM cells can acquire mesenchymal features associated with enhanced invasive potential following stimulation by secretory cytokines, such as TGF-ß. This property of GBM contributes to heterogeneity in this tumor type and may blur the boundaries between the proposed transcriptional subtypes. Targeting TGF-ß or downstream targets like ZEB1 might be of potential benefit in reducing the invasive phenotype of GBM in a subpopulation of patients.


Subject(s)
Epithelial-Mesenchymal Transition , Glioblastoma/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Line, Tumor , Cell Movement , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/physiopathology , Homeodomain Proteins/genetics , Humans , Mice , Mice, SCID , Neoplasm Invasiveness , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1
5.
Crit Rev Oncol Hematol ; 92(1): 25-37, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24935219

ABSTRACT

Barrett's metaplasia of the esophagus (BE) is the precursor lesion of esophageal adenocarcinoma (EAC), a deadly disease with a 5-year overall survival of less than 20%. The molecular mechanisms of BE development and its transformation to EAC are poorly understood and current surveillance and treatment strategies are of limited efficacy. Increasing evidence suggests that aberrant signaling through pathways active in the embryological development of the esophagus contributes to BE development and progression to EAC. We discuss the role that the Bone morphogenetic protein, Hedgehog, Wingless-Type MMTV Integration Site Family (WNT) and Retinoic acid signaling pathways play during embryological development of the esophagus and their contribution to BE development and malignant transformation. Modulation of these pathways provides new therapeutic opportunities. By integrating findings in developmental biology with those from translational research and clinical trials, this review provides a platform for future studies aimed at improving current management of BE and EAC.


Subject(s)
Barrett Esophagus/etiology , Barrett Esophagus/pathology , Cell Transformation, Neoplastic , Signal Transduction , Adenocarcinoma/etiology , Adenocarcinoma/therapy , Animals , Barrett Esophagus/therapy , Disease Progression , Esophageal Neoplasms/etiology , Esophageal Neoplasms/therapy , Humans , Molecular Targeted Therapy
6.
Br J Cancer ; 109(10): 2685-95, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24136147

ABSTRACT

BACKGROUND: Tumour cell-selective activation of apoptosis by recombinant human TNF-related apoptosis-inducing ligand (rhTRAIL) is enhanced through co-activation of p53 by chemotherapeutic drugs. The novel anticancer agent nutlin-3 provides a promising alternative for p53 activation by disrupting the interaction between p53 and its negative feedback regulator MDM2. METHODS: We examined whether nutlin-3 enhances apoptosis induction by rhTRAIL and the DR5-selective TRAIL variant D269H/E195R in wild-type p53-expressing ovarian, colon and lung cancer cell lines and in an ex vivo model of human ovarian cancer. RESULTS: Nutlin-3 enhanced p53, p21, MDM2 and DR5 surface expression. Although nutlin-3 did not induce apoptosis, it preferentially enhanced D269H/E195R-induced apoptosis over rhTRAIL. Combination treatment potentiated the cleavage of caspases 8, 9, 3 and PARP. P53 and MDM2 siRNA experiments showed that this enhanced apoptotic effect was mediated by wild-type p53. Indeed, nutlin-3 did not enhance rhTRAIL-induced apoptosis in OVCAR-3 cells harbouring mutant p53. Addition of the chemotherapeutic drug cisplatin to the combination further increased p53 and DR5 levels and rhTRAIL- and D269H/E195R-induced apoptosis. As a proof of concept, we show that the combination of D269H/E195R, nutlin-3 and cisplatin induced massive apoptosis in ex vivo tissue slices of primary human ovarian cancers. CONCLUSION: Nutlin-3 is a potent enhancer of D269H/E195R-induced apoptosis in wild-type p53-expressing cancer cells. Addition of DNA-damaging agents such as cisplatin further enhances DR5-mediated apoptosis.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Neoplasms/pathology , Piperazines/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Amino Acid Substitution , Apoptosis/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Genes, p53 , Humans , Neoplasms/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Recombinant Proteins/genetics , Substrate Specificity , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Cells, Cultured
7.
Cell Death Differ ; 20(7): 858-68, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23579241

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based therapy is currently evaluated in clinical studies as a tumor cell selective pro-apoptotic approach. However, besides activating canonical caspase-dependent apoptosis by binding to TRAIL-specific death receptors, the TRAIL ligand can activate non-canonical cell survival or proliferation pathways in resistant tumor cells through the same death receptors, which is counterproductive for therapy. Even more, recent studies indicate metastases-promoting activity of TRAIL. In this review, the remarkable dichotomy in TRAIL signaling is highlighted. An overview of the currently known mechanisms involved in non-canonical TRAIL signaling and the subsequent activation of various kinases is provided. These kinases include RIP1, IκB/ NF-κB, MAPK p38, JNK, ERK1/2, MAP3K TAK1, PKC, PI3K/Akt and Src. The functional consequences of their activation, often being stimulation of tumor cell survival and in some cases enhancement of their invasive behavior, are discussed. Interestingly, the non-canonical responses triggered by TRAIL in resistant tumor cells resemble that of TRAIL-induced signals in non-transformed cells. Better knowledge of the mechanism underlying the dichotomy in TRAIL receptor signaling may provide markers for selecting patients who will likely benefit from TRAIL-based therapy and could provide a rationalized basis for combination therapies with TRAIL death receptor-targeting drugs.


Subject(s)
Neoplasms/pathology , Neoplasms/physiopathology , Phosphotransferases/physiology , Receptors, Death Domain/physiology , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Disease Models, Animal , Humans , I-kappa B Kinase/physiology , Mice , Mitogen-Activated Protein Kinase Kinases/physiology , NF-kappa B/physiology , Neoplasms/drug therapy , Receptors, Death Domain/drug effects , TNF-Related Apoptosis-Inducing Ligand/drug effects
8.
Br J Cancer ; 108(3): 587-96, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23322202

ABSTRACT

BACKGROUND: High-grade astrocytomas are malignant brain tumours that infiltrate the surrounding brain tissue and have a poor prognosis. Activation of formyl peptide receptor (FPR1) on the human astrocytoma cell line U87 promotes cell motility, growth and angiogenesis. We therefore investigated the FPR1 inhibitor, Chemotaxis Inhibitory Protein of S. aureus (CHIPS), as a potential anti-astrocytoma drug. METHODS AND RESULTS: FPR1 expression was studied immunohistochemically in astrocytomas WHO grades I-IV. With intracellular calcium mobilisation and migration assays, human ligands were tested for their ability to activate FPR1 on U87 cells and on a cell line derived from primary astrocytoma grade IV patient material. Thereafter, we selectively inhibited these ligand-induced responses of FPR1 with an anti-inflammatory compound called Chemotaxis Inhibitory Protein of S. aureus (CHIPS). U87 xenografts in NOD-SCID mice served to investigate the effects of CHIPS in vivo. FPR1 was expressed in 29 out of 32 (90%) of all grades of astrocytomas. Two human mitochondrial-derived formylated peptides, formyl-methionil-leucine-lysine-isoleucine-valine (fMLKLIV) and formyl-methionil-methionil-tyrosine-alanine-leucine-phenylalanine (fMMYALF), were potent activators of FPR1 on tumour cells. Ligand-induced responses of FPR1-expressing tumour cells could be inhibited with FPR1 inhibitor CHIPS. Treatment of tumour-bearing mice with CHIPS slightly reduced tumour growth and improved survival as compared to non-treated animals (P=0.0019). CONCLUSION: Targeting FPR1 with CHIPS reduces cell motility and tumour cell activation, and prolongs the survival of tumour-bearing mice. This strategy could be explored in future research to improve treatment results for astrocytoma patients.


Subject(s)
Astrocytoma/pathology , Astrocytoma/prevention & control , Bacterial Proteins/pharmacology , Brain Neoplasms/prevention & control , Chemotaxis/drug effects , Peptide Fragments/pharmacology , Receptors, Formyl Peptide/antagonists & inhibitors , Animals , Astrocytoma/metabolism , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Calcium/metabolism , Cell Movement/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Grading , Receptors, Formyl Peptide/metabolism , Tumor Cells, Cultured
9.
Crit Rev Oncol Hematol ; 85(3): 363-72, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22944363

ABSTRACT

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine that selectively eradicates tumour cells via specific cell surface receptors and is intensively explored for use as a novel anticancer approach. To enhance the efficacy of TRAIL receptor agonists the proteasome inhibitor bortezomib is one of the most potent sensitizers. Here we review the main mechanisms underlying bortezomib-dependent TRAIL sensitization, including stimulation of apoptosis by increasing expression of TRAIL receptors, reduction of cFLIP and enhancement of caspase 8 activation, and modulation of Bcl-2 family proteins and inhibitor of apoptosis proteins (IAPs). Concomitantly, pro-survival signals are suppressed such as elicited by NF-κB and Akt. The different preclinical tumour models explored with this combination, including primary tumour (stem) cells, stroma co-culture and mice models, are discussed, as well as possible hurdles for clinical activity. Collectively, anticipating a solid rationale for bortezomib-TRAIL combination and very promising preclinical results, its clinical activity remains to be demonstrated.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Pyrazines/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Pyrazines/pharmacology , Signal Transduction/drug effects
10.
Ann Oncol ; 23(11): 2937-2942, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22689177

ABSTRACT

BACKGROUND: Initial response of small-cell lung cancer (SCLC) to chemotherapy is high, and recurrences occur frequently, leading to early death. This study investigated the prognostic value of circulating tumor cells (CTCs) in patients with SCLC and whether changes in CTCs can predict response to chemotherapy. Patients and methods In this multicenter prospective study, blood samples for CTC analysis were obtained from 59 patients with SCLC before, after one cycle, and at the end of chemotherapy. CTCs were measured using CellSearch systems. RESULTS: At baseline, lower numbers of CTCs were observed for 21 patients with limited SCLC (median = 6, range 0-220) compared with 38 patients with extensive stage (median = 63, range 0-14,040). Lack of measurable CTCs (27% of patients) was associated with prolonged survival (HR 3.4; P ≤ 0.001). CTCs decreased after one cycle of chemotherapy; this decrease was not associated with tumor response after four cycles of chemotherapy. CTC count after the first cycle of chemotherapy was the strongest predictor for overall survival (HR 5.7; 95% CI 1.7-18.9; P = 0.004). CONCLUSION: Absolute CTCs after one cycle of chemotherapy in patients with SCLC is the strongest predictor for response on chemotherapy and survival. Patients with low initial CTC numbers lived longer than those with higher CTCs.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Neoplastic Cells, Circulating , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/radiotherapy , Male , Middle Aged , Platinum Compounds/therapeutic use , Prognosis , Prospective Studies , Small Cell Lung Carcinoma/mortality , Small Cell Lung Carcinoma/radiotherapy , Treatment Outcome
11.
Br J Cancer ; 104(7): 1185-92, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21386840

ABSTRACT

BACKGROUND: Thymidine phosphorylase (TP) is often overexpressed in tumours and has a role in tumour aggressiveness and angiogenesis. Here, we determined whether TP increased tumour invasion and whether TP-expressing cancer cells stimulated angiogenesis. METHODS: Angiogenesis was studied by exposing endothelial cells (HUVECs) to conditioned medium (CM) derived from cancer cells with high (Colo320TP1=CT-CM, RT112/TP=RT-CM) and no TP expression after which migration (wound-healing-assay) and invasion (transwell-assay) were determined. The involvement of several angiogenic factors were examined by RT-PCR, ELISA and blocking antibodies. RESULTS: Tumour invasion was not dependent on intrinsic TP expression. The CT-CM and RT-CM stimulated HUVEC-migration and invasion by about 15 and 40%, respectively. Inhibition by 10 µM TPI and 100 µM L-dR, blocked migration and reduced the invasion by 50-70%. Thymidine phosphorylase activity in HUVECs was increased by CT-CM. Reverse transcription-polymerase chain reaction revealed a higher mRNA expression of bFGF (Colo320TP1), IL-8 (RT112/TP) and TNF-α, but not VEGF. Blocking antibodies targeting these factors decreased the migration and invasion that was induced by the CT-CM and RT-CM, except for IL-8 in CT-CM and bFGF in RT-CM. CONCLUSION: In our cell line panels, TP did not increase the tumour invasion, but stimulated the migration and invasion of HUVECs by two different mechanisms. Hence, TP targeting seems to provide a potential additional strategy in the field of anti-angiogenic therapy.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Cell Movement , Endothelial Cells/physiology , Neoplasms/enzymology , Thymidine Phosphorylase/physiology , Cell Line, Tumor , Cell Proliferation , Endothelial Cells/enzymology , Fibroblast Growth Factor 2/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Interleukin-8/genetics , Neoplasm Invasiveness , Neoplasms/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Tumor Necrosis Factor-alpha/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Biochem Pharmacol ; 80(6): 786-92, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20488166

ABSTRACT

Thymidine phosphorylase (TP) is often overexpressed in cancer and potentially plays a role in the stimulation of angiogenesis. The exact mechanism of angiogenesis induction is unclear, but is postulated to be related to thymidine-derived sugars. TP catalyzes the conversion of thymidine (TdR) to thymine and deoxyribose-1-phosphate (dR-1-P), which can be converted to dR-5-P, glyceraldehyde-3-phosphate (G3P) or deoxyribose (dR). However, it is unclear which sugar accumulates in this reaction. Therefore, in the TP overexpressing Colo320 TP1 and RT112/TP cells we determined by LC-MS/MS which sugars accumulated, their subcellular localization (using (3)H-TdR) and whether dR was secreted from the cells. In both TP-overexpressing cell lines, dR-1-P and dR-5-P accumulated intracellularly at high levels and dR was secreted extensively by the cells. A specific inhibitor of TP completely blocked TdR conversion, and thus no sugars were formed. To examine whether these sugars may be used for the production of angiogenic factors or other products, we determined with (3)H-TdR in which subcellular location these sugars accumulated. TdR-derived sugars accumulated in the cytoskeleton and to some extent in the cell membrane, while incorporation into the DNA was responsible for trapping in the nucleus. In conclusion, various metabolic routes were entered, of which the TdR-derived sugars accumulated in the cytoskeleton and membrane. Future studies should focus on which exact metabolic pathway is involved in the induction of angiogenesis.


Subject(s)
Sugar Phosphates/metabolism , Thymidine Phosphorylase/biosynthesis , Thymidine/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Endothelial Cells , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Ribosemonophosphates/metabolism , Substrate Specificity , Sugar Phosphates/chemistry , Thymidine/chemistry , Thymidine Phosphorylase/genetics , Thymidine Phosphorylase/metabolism
13.
Drug Resist Updat ; 13(1-2): 2-15, 2010.
Article in English | MEDLINE | ID: mdl-20036602

ABSTRACT

Non-small cell lung cancer (NSCLC) is a common and often fatal malignancy, diagnosed at an advanced stage in more than half of the cases. Chemo-resistance remains a major problem in the treatment of NSCLC patients with conventional chemotherapeutic agents. Therefore main research efforts are focused on the development of novel targeted agents. In this review we provide an overview on the use of TNF-related apoptosis-inducing ligand (TRAIL) receptor targeting agents in NSCLC models and in early clinical studies. Different TRAIL receptor targeting agents are available which have been tested in NSCLC models and some were tested in the clinic. The efficacy of these drugs as single agents in NSCLC models is discussed as well as different mechanisms of resistance that are found in NSCLC cell lines. In order to maximize sensitivity to TRAIL receptor targeting drugs, combined use with other drugs is of interest. The current status of tested combinations of TRAIL receptor targeting agents with other therapeutics, such as classical cytotoxics, Bcl-2 family targeting agents, proteasome inhibitors, EGFR inhibitors, histone deacetylase inhibitors and COX-2 inhibitors as well as their mechanisms in preclinical studies are discussed. Clinical studies on TRAIL targeted therapies in which NSCLC patients were included are discussed and future perspectives are considered.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Delivery Systems/methods , Lung Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Clinical Trials as Topic , Combined Modality Therapy/methods , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor/methods , Humans , Models, Biological , Signal Transduction/drug effects
15.
Br J Cancer ; 96(3): 450-6, 2007 Feb 12.
Article in English | MEDLINE | ID: mdl-17285121

ABSTRACT

Doxorubicin (DOX) is an antitumour agent for different types of cancer, but the dose-related cardiotoxicity limits its clinical use. To prevent this side effect we have developed the flavonoid monohydroxyethylrutoside (monoHER), a promising protective agent, which did not interfere with the antitumour activity of DOX. To obtain more insight in the mechanism underlying the selective protective effects of monoHER, we investigated whether monoHER (1 mM) affects DOX-induced apoptosis in neonatal rat cardiac myocytes (NeRCaMs), human endothelial cells (HUVECs) and the ovarian cancer cell lines A2780 and OVCAR-3. DOX-induced cell death was effectively reduced by monoHER in heart, endothelial and A2780 cells. OVCAR-3 cells were highly resistant to DOX-induced apoptosis. Experiments with the caspase-inhibitor zVAD-fmk showed that DOX-induced apoptosis was caspase-dependent in HUVECs and A2780 cells, whereas caspase-independent mechanisms seem to be important in NeRCaMs. MonoHER suppressed DOX-dependent activation of the mitochondrial apoptotic pathway in normal and A2780 cells as illustrated by p53 accumulation and activation of caspase-9 and -3 cleavage. Thus, monoHER acts by suppressing the activation of molecular mechanisms that mediate either caspase-dependent or -independent cell death. In light of the current work and our previous studies, the use of clinically achievable concentrations of monoHER has no influence on the antitumour activity of DOX whereas higher concentrations as used in the present study could influence the antitumour activity of DOX.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Caspases/physiology , Doxorubicin/pharmacology , Flavonoids/pharmacology , Cell Cycle/drug effects , Cell Line , Cytoprotection , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Humans , Myocytes, Cardiac/drug effects , Tumor Suppressor Protein p53/analysis , bcl-2-Associated X Protein/analysis
16.
Cancer Gene Ther ; 13(12): 1105-14, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16841078

ABSTRACT

Chemotherapy, including microtubule (MT)-interacting agents, can enhance the tumor-eradicating activity of replication-competent adenoviruses. The purpose of this study was to obtain more insight into the mechanism underlying this enhancement that may be exploited for the development of improved therapy. Two MT-interacting agents with opposite activity, paclitaxel (PTX) that stabilizes and vincristine (VCR) that destabilizes MTs, were found to synergistically enhance adenoviral oncolysis in non-small-cell lung cancer (NSCLC) cells. To explore the possibility that these drugs affect the viral life cycle by modulating adenoviral gene expression, we used a quantitative reverse transcription-polymerase chain reaction assay and found that PTX, but not VCR, increased the expression of E1A13S, ADP and Penton genes, which correlated with an increase in viral particle assembly and release. Next, the effect of combined treatment on cell-cycle progression was studied. Both drugs suppressed adenovirus-induced S-phase arrest and instead caused G2/M arrest, which was accompanied by an increase in apoptotic cells. Taken together, the enhancement of oncolysis by MT-interacting drugs appears not to require specific MT transport or scaffold functions. Our findings suggest that MT-interacting drug-induced cellular signals that modulate cell-cycle arrest and apoptosis are primarily on the basis of their oncolysis-enhancing activity.


Subject(s)
Adenoviridae/genetics , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Vincristine/pharmacology , Adenoviridae/drug effects , Adenovirus E3 Proteins/drug effects , Adenovirus E3 Proteins/genetics , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/genetics , Capsid Proteins/drug effects , Capsid Proteins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Gene Expression Regulation/drug effects , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Microtubules/drug effects , Tumor Cells, Cultured
17.
Oncogene ; 25(35): 4867-79, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16547492

ABSTRACT

As mitosis progresses, the chromosomal passenger proteins (CPPs) Survivin, Aurora B, INCENP and Borealin dynamically colocalize to mitotic structures. Chromosomal passenger proteins are already expressed during G2, whereas the nuclear envelope is only disassembled at the end of prophase. However, the mechanisms that modulate their nucleocytoplasmic localization before nuclear envelope breakdown (NEB) are poorly characterized. Using epitope-tagged proteins, we show that Aurora B, like Survivin, undergoes CRM1-mediated nucleocytoplasmic shuttling, although both proteins lack identifiable 'classical' nuclear transport signals. On the other hand, INCENP resides more stably in the nucleus and contains multiple nuclear localization signals. Finally, Borealin was detected in the nucleolus and the cytoplasm, but its cytoplasmic localization is not directly regulated by CRM1. Coexpression experiments indicate that the nuclear localization of Aurora B, but not of Survivin, is modulated by INCENP and that Survivin prevents the nucleolar accumulation of Borealin. Our data reveal that, in contrast to their closely related localization during mitosis, the nucleocytoplasmic localization of the CPPs before NEB is largely unrelated. Furthermore, the specific effect of INCENP and Survivin on the localization of Aurora B and Borealin, respectively, suggests that different complexes of CPPs may exist not only during mitosis, as recently reported, but also before NEB.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/physiology , Nuclear Envelope/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cytoplasm/enzymology , Cytoplasm/metabolism , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Subcellular Fractions/metabolism , Survivin
18.
Ann Oncol ; 15(11): 1654-60, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15520067

ABSTRACT

BACKGROUND: Expression of survivin, a member of the inhibitor of apoptosis protein family, is commonly detected in cancers but not in normal differentiated tissues. Survivin is usually localized in the cytoplasm of cancer cells, but nuclear localization has also been described, and we recently reported that survivin is a nuclear-cytoplasmic shuttling protein. PATIENTS AND METHODS: Fifty-three tumor specimens from patients with inoperable non-small-cell lung cancer (NSCLC) (55% stage IIIA, 17% stage IIIB and 28% stage IV) who underwent chemotherapy treatment were evaluated with immunohistochemistry for survivin expression and localization. These two sets of data were processed and tested for correlation with major patient characteristics, response to chemotherapy, and overall and relapse-free survival. RESULTS: Survivin was present only in malignant tissues, and 47/53 (89%) of the specimens were positive. The overall median expression of tumor cells was 40%, and this value was used as a cut-off point for statistical analysis. By dichotomizing the specimens as expressing low or high levels of survivin, a significant association was seen between the expression of survivin and the histology of the tumors (P=0.020), squamous cell carcinoma being the histotype with lower levels of survivin expression. Three patterns of localization were observed: 42% of cases (22/53) showed reactivity confined to the nucleus, 17% (nine of 53) only in the cytoplasm and 30% (16/53) in both the nucleus and the cytoplasm. Interestingly, nuclear survivin levels predicted longer overall and relapse-free survival, in univariate and multivariate analyses. Expression and localization of survivin did not correlate with response to chemotherapy. CONCLUSIONS: Our results indicate that differential localization of survivin may be a prognostic factor for NSCLC. Further studies are warranted.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Microtubule-Associated Proteins/analysis , Adult , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/therapy , Cell Nucleus/chemistry , Female , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Lung Neoplasms/metabolism , Male , Middle Aged , Neoplasm Proteins , Neoplasm Staging , Prognosis , Survival Analysis , Survivin
19.
Br J Cancer ; 91(1): 171-7, 2004 Jul 05.
Article in English | MEDLINE | ID: mdl-15173860

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL, also known as Apo-2L) is a promising novel anticancer agent that selectively induces apoptosis in tumour cells and the activity of which can be enhanced by combined treatment with chemo- or radiotherapy. For therapeutic purposes, the use of full-length TRAIL may be favourable to recombinant TRAIL based on its increased tumour cell killing potential, and the delivery of TRAIL at the tumour site by adenovirus vectors may provide an approach to overcome the short half-life of recombinant TRAIL and hepatocyte toxicity in vivo. Here, we constructed an adenoviral vector expressing full-length TRAIL (AdTRAIL) and studied the potential of chemo- and radiotherapy in enhancing AdTRAIL-induced apoptosis in non-small cell lung cancer (NSCLC) H460 cells and normal cells and, in addition, investigated the mechanism of AdTRAIL-induced apoptosis. AdTRAIL effectively killed H460 cells, which we previously showed to have a deficiency in mitochondria-dependent apoptosis by downstream activation of caspase-8 rather than caspase-9. Further analyses revealed that AdTRAIL induces death receptor- and mitochondria-dependent apoptosis that could be partially suppressed by Bcl2 overexpression. Combined treatment with doxorubicin (DOX), cisplatin (CDDP), paclitaxel (PTX) and radiation strongly enhanced AdTRAIL-induced cytotoxicity in a synergistic way. Synergy was accompanied by the cleavage of Bid and an increase in caspase-8 processing that was abolished by Bcl2 overexpression, indicating that the Bid-mitochondrial amplification loop is functional in H460 cells. Moreover, combination treatment did not alter the tumour selectivity of AdTRAIL since normal human fibroblasts (NHFs) remained resistant under these conditions. These findings further indicate that the combined use of chemo/radiotherapy and adenovirus-produced full-length TRAIL may provide a valuable treatment option for NSCLC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Membrane Glycoproteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Radiation Tolerance , Tumor Necrosis Factor-alpha/biosynthesis , Adenocarcinoma/pathology , Adenoviridae , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins , Fibroblasts/physiology , Humans , Ligands , Membrane Glycoproteins/pharmacology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , fas Receptor
20.
Br J Cancer ; 88(3): 447-54, 2003 Feb 10.
Article in English | MEDLINE | ID: mdl-12569390

ABSTRACT

Cultured H35 hepatoma cells release a cytotoxic factor in response to irradiation with X-rays. When the conditioned medium from irradiated cells is given to nonirradiated cells, growth is inhibited and followed by cell death, possibly apoptosis, Analysis of the conditioned medium reveals a dramatic change in the ornithine (urea) cycle components after the irradiation. A strong decrease in medium arginine is accompanied with parallel increases in ornithine, citrulline and ammonia. The high level of ammonia appears to be largely responsible for the observed cytotoxicity. The development of hyperammonia by irradiated cells and the related toxicity depend on the radiation dose and the number of cells seeded thereafter for the medium conditioning. Development of cytotoxicity by irradiated cells is completely prevented with the arginase inhibitor L-norvaline, in arginine-deficient medium or when citrulline replaces arginine. These preventive measures result in subtoxic ammonia levels.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Culture Media, Conditioned/pharmacology , Liver Neoplasms/metabolism , Ornithine/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Count , Cell Division/drug effects , Cell Division/radiation effects , Dose-Response Relationship, Radiation , Liver Neoplasms/pathology , Radiation Dosage , Rats , Statistics as Topic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...