Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 28
Filtrer
1.
J Cancer Res Clin Oncol ; 149(10): 7197-7206, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-36884118

RÉSUMÉ

PURPOSE: The WHO 2016 re-classification of myeloproliferative neoplasms resulted in a separation of essential thrombocythemia (ET) from the pre-fibrotic and fibrotic (overt) phases of primary myelofibrosis (MF). This study reports on a chart review conducted to evaluate the real life approach regarding clinical characteristics, diagnostic assessment, risk stratification and treatment decisions for MPN patients classified as ET or MF after implementation of the WHO 2016 classification. METHODS: In this retrospective chart review, 31 office-based hematologists/oncologists and primary care centers in Germany participated between April 2021 and May 2022. Physicians reported available data obtained from patient charts via paper-pencil based survey (secondary use of data). Patient features were evaluated using descriptive analysis, also including diagnostic assessment, therapeutic strategies and risk stratification. RESULTS: Data of 960 MPN patients diagnosed with essential thrombocythemia (ET) (n = 495) or myelofibrosis (MF) (n = 465) after implementation of the revised 2016 WHO classification of myeloid neoplasms was collected from the patient charts. While they met at least one minor WHO-criteria for primary myelofibrosis, 39.8% of those diagnosed with ET did not have histological BM testing at diagnosis. 63.4% of patients who were classified as having MF, however, did not obtain an early prognostic risk assessment. More than 50% of MF patients showed characteristics consistent with the pre-fibrotic phase, which was emphasized by the frequent use of cytoreductive therapy. Hydroxyurea was the most frequently used cytoreductive medication in 84.7% of ET and 53.1% of MF patients. While both ET and MF cohorts showed cardiovascular risk factors in more than 2/3 of the cases, the use of platelet inhibitors or anticoagulants varied between 56.8% in ET and 38.1% in MF patients. CONCLUSIONS: Improved histopathologic diagnostics, dynamic risk stratification including genetic risk factors for cases of suspected ET and MF are recommended for precise risk assessment and therapeutic stratification according to WHO criteria.


Sujet(s)
Syndromes myéloprolifératifs , Tumeurs , Myélofibrose primitive , Thrombocytémie essentielle , Humains , Myélofibrose primitive/diagnostic , Myélofibrose primitive/thérapie , Études rétrospectives , Syndromes myéloprolifératifs/diagnostic , Syndromes myéloprolifératifs/épidémiologie , Syndromes myéloprolifératifs/thérapie
2.
Stud Health Technol Inform ; 296: 25-32, 2022 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-36073485

RÉSUMÉ

Machine learning based disease classification have already achieved amazing results in medicine: for example, models can find a tumor in computer tomography images at least as accurately as experts in the field. Since the development and widespread use of actigraphy watches, activity data has been used as a basis for diagnosing various diseases such as depression or Alzheimer's disease. In this study, we use a dataset with activity measurements of mentally ill and healthy people, calculate various features and achieve a classification accuracy of over 78%. The paper describes and motivates the used features, discusses differences between healthy, bipolar 2 and unipolar participants and compares several well-known machine learning classifiers on different classification tasks and with different feature sets.


Sujet(s)
Maladie d'Alzheimer , Dépression , Maladie d'Alzheimer/imagerie diagnostique , Dépression/diagnostic , Humains , Apprentissage machine , Activité motrice , Tomodensitométrie
3.
Int J Mol Sci ; 21(4)2020 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-32102484

RÉSUMÉ

Persistent signalling via the PI3K/AKT/mTOR pathway is a major driver of malignancy in NF1-associated malignant peripheral nerve sheath tumours (MPNST). Nevertheless, single targeting of this pathway is not sufficient to inhibit MPNST growth. In this report, we demonstrate that combined treatment with the allosteric pan-AKT inhibitor MK-2206 and the mTORC1/mTORC2 inhibitor AZD8055 has synergistic effects on the viability of MPNST cell lines in comparison to the treatment with each compound alone. However, when treating animals bearing experimental MPNST with the combined AKT/mTOR regime, no influence on tumour growth was observed. Further analysis of the MPNST xenograft tumours resistant to AKT/mTOR treatment revealed a reactivation of both AKT and mTOR in several tumour samples. Additional targeting of the RAS/RAF/MEK/MAPK pathway with the allosteric MEK1/2 inhibitor AZD6244 showed synergistic effects on the viability of MPNST cell lines in vitro in comparison to the dual AKT/mTOR inhibition. In summary, these data indicate that combined treatment with AKT and mTOR inhibitors is effective on MPNST cells in vitro but tumour resistance can occur rapidly in vivo by restoration of AKT/mTOR signalling. Our data further suggest that a triple treatment with inhibitors against AKT, mTORC1/2 and MEK1/2 may be a promising treatment option that should be further analysed in an experimental MPNST mouse model in vivo.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Neurofibrosarcome/traitement médicamenteux , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Animaux , Benzimidazoles/administration et posologie , Lignée cellulaire tumorale , Synergie des médicaments , Composés hétérocycliques 3 noyaux/administration et posologie , Humains , Souris SCID , Morpholines/administration et posologie , Neurofibromatose de type 1/complications , Neurofibromatose de type 1/métabolisme , Neurofibrosarcome/complications , Neurofibrosarcome/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme
4.
Neuro Oncol ; 20(12): 1594-1605, 2018 11 12.
Article de Anglais | MEDLINE | ID: mdl-30053149

RÉSUMÉ

Background: Aerobic glycolysis confers several advantages to tumor cells, including shunting of metabolites into anabolic pathways. In glioblastoma cells, hypoxia induces a flux shift from the pentose phosphate pathway toward glycolysis and a switch from proliferation to migration. The mechanistic link between glycolysis and migration is poorly understood. Since glucose-6-phosphate isomerase (GPI) is identical to the secreted cytokine autocrine motility factor (AMF), we investigated whether GPI/AMF regulates glioblastoma cell invasion. Methods: The expression and hypoxic regulation of GPI/AMF and its receptor AMFR were analyzed in glioblastoma tissue and cell lines. Functional effects were studied in vitro and in xenograft models. Results: High GPI/AMF expression in glioblastomas was found to be associated with a worse patient prognosis, and levels were highest in hypoxic pseudopalisades. Hypoxia upregulated both GPI/AMF and AMFR expression as well as GPI/AMF secretion in vitro. GPI/AMF stimulated cell migration in an autocrine fashion, and GPI/AMF expression was upregulated in migratory cells but reduced in rapidly proliferating cells. Knockdown or inhibition of GPI/AMF reduced glioblastoma cell migration but in part stimulated proliferation. In a highly invasive orthotopic glioblastoma model, GPI/AMF knockdown reduced tumor cell invasion but did not prolong survival. In a highly proliferative model, knockdown tumors were even larger and more proliferative than controls; however, perivascular invasion, provoked by simultaneous bevacizumab treatment, was reduced. Conclusions: GPI/AMF is a potent motogen for glioblastoma cells, explaining in part the association between glycolysis and migration. Targeting GPI/AMF is, however, problematic, since beneficial anti-invasive effects may be outweighed by unintended mitogenic effects. Key Points: 1.Increased glycolysis is linked with increased cell migration and invasion in glioblastoma cells. 2.The glycolysis enzyme GPI/AMF may serve as a target for antimetabolic and anti-invasive therapy. 3.Despite reducing tumor invasion, GPI/AMF targeting may have unwanted growth stimulatory effects.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Mouvement cellulaire , Prolifération cellulaire , Glioblastome/anatomopathologie , Glucose 6-phosphate isomerase/métabolisme , Récepteur du facteur autocrine de motilité/métabolisme , Animaux , Apoptose , Communication autocrine , Marqueurs biologiques tumoraux/génétique , Cytokines/génétique , Cytokines/métabolisme , Régulation de l'expression des gènes tumoraux , Glioblastome/génétique , Glioblastome/métabolisme , Glucose 6-phosphate isomerase/génétique , Humains , Souris , Invasion tumorale , Pronostic , Récepteur du facteur autocrine de motilité/génétique , Taux de survie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Oncotarget ; 8(4): 6155-6168, 2017 Jan 24.
Article de Anglais | MEDLINE | ID: mdl-28008153

RÉSUMÉ

Despite improvement of therapeutic treatments for breast cancer, the development of brain metastases has become a major limitation to life expectancy for many patients. Brain metastases show very commonly alterations in EGFR and HER2 driven pathways, of which PTEN is an important regulator. Here, we analyzed PTEN expression in 111 tissue samples of breast cancer brain metastases (BCBM). Loss of PTEN was found in a substantial proportion of BCBM samples (48.6%) and was significantly associated with triple-negative breast cancer (67.5%, p = 0.001) and a shorter survival time after surgical resection of brain metastases (p = 0.048). Overexpression of PTEN in brain-seeking MDA-MB-231 BR cells in vitro reduced activation of the AKT pathway, notably by suppression of Akt1 kinase activity. Furthermore, the migration of MDA-MB-231 BR cells in vitro was promoted by co-culturing with both astrocytes and microglial cells. Interestingly, when PTEN was overexpressed the migration was significantly inhibited. Moreover, in an ex vivo organotypic brain slice model, PTEN overexpression reduced invasion of tumor cells. This was accompanied by reduced astrocyte activation that was mediated by autocrine and paracrine activation of GM-CSF/ CSF2RA and AKT/ PTEN pathways. In conclusion, loss of PTEN is frequently detected in triple-negative BCBM patients and associated with poor prognosis. The findings of our functional studies suggest that PTEN loss promotes a feedback loop between tumor cells and glial cells, which might contribute to disease progression.


Sujet(s)
Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/secondaire , Névroglie/métabolisme , Phosphohydrolase PTEN/métabolisme , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du cerveau/chirurgie , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Techniques de coculture , Évolution de la maladie , Rétrocontrôle physiologique , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Invasion tumorale , Protéines proto-oncogènes c-akt/métabolisme , Analyse de survie , Tumeurs du sein triple-négatives/chirurgie
6.
Neuro Oncol ; 18(9): 1219-29, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-26917237

RÉSUMÉ

BACKGROUND: The dichotomy between glioblastoma cell migration and proliferation is regulated by various parameters including oxygen tension. In glioblastoma stem-like cells, hypoxia induces downregulation of pentose phosphate pathway (PPP) enzymes and a flux shift towards glycolysis. We investigated whether the 2 parallel glucose metabolic pathways are intrinsically linked with cell function and whether these pathways are mechanistically involved in regulating functional programs. METHODS: Enzyme expression, migration, and proliferation under hypoxia were studied in multiple cell types. Rapidly and slowly dividing or migrating glioblastoma cells were separated, and enzyme profiles were compared. Glucose-6-phosphate dehydrogenase (G6PD) and Aldolase C (ALDOC), the most strongly inversely regulated PPP and glycolysis enzymes, were knocked down by short hairpin RNA. RESULTS: Hypoxia caused downregulation of PPP enzymes and upregulation of glycolysis enzymes in a broad spectrum of cancer and nonneoplastic cells and consistently stimulated migration while reducing proliferation. PPP enzyme expression was increased in rapidly dividing glioblastoma cells, whereas glycolysis enzymes were decreased. Conversely, glycolysis enzymes were elevated in migrating cells, whereas PPP enzymes were diminished. Knockdown of G6PD reduced glioblastoma cell proliferation, whereas ALDOC knockdown decreased migration. Enzyme inhibitors had similar effects. G6PD knockdown in a highly proliferative but noninvasive glioblastoma cell line resulted in prolonged survival of mice with intracerebral xenografts, whereas ALDOC knockdown shortened survival. In a highly invasive glioblastoma xenograft model, tumor burden was unchanged by either knockdown. CONCLUSIONS: Cell function and metabolic state are coupled independently of hypoxia, and glucose metabolic pathways are causatively involved in regulating "go or grow" cellular programs.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Glioblastome/anatomopathologie , Glucose/métabolisme , Glycolyse/physiologie , Hypoxie/physiopathologie , Voie des pentoses phosphates , Animaux , Apoptose , Glioblastome/métabolisme , Humains , Souris , Souris hairless , Souris de lignée NOD , Souris SCID , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Target Oncol ; 11(1): 29-40, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26032687

RÉSUMÉ

Although the epidermal growth factor receptor (EGFR) is overexpressed and/or amplified in more than 50 % of all glioblastomas (GBM), therapeutic targeting of the EGFR has not yet been successful. Since histone deacetylases (HDAC) have been described as controlling EGFR expression, we combined the EGFR tyrosine kinase inhibitor erlotinib with different HDAC inhibitors (HDACi) and investigated the benefit of combinatorial therapy for glioblastoma cells. Using representative models of EGFR-amplified, erlotinib-sensitive and -resistant GBM with or without EGFRvIII expression, we determined proliferation, migration, and EGFR-dependent signaling in response to erlotinib and HDACi alone or in combination. HDACi significantly inhibited proliferation of erlotinib-resistant GBM cells, partially restored their sensitivity to erlotinib, and also significantly reduced proliferation of all treatment-naïve cell lines tested. In combination with erlotinib, the development of resistance was prevented. The multitargeted EGFR/HDAC-inhibitor CUDC-101 exhibited similar effects. However, inhibition of cell migration was only achieved by targeting EGFR, and HDACi exhibited no additive effect. Mechanistically, we identified an HDACi-dependent decrease of EGFR/EGFRvIII protein expression underlying the anti-proliferative effects of HDACi. In conclusion, HDACi in combination with erlotinib might serve as a treatment option for newly diagnosed, treatment-naïve tumors irrespective of their EGFR status, as well as for treatment-refractory, EGFR-overexpressing GBM.


Sujet(s)
Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/antagonistes et inhibiteurs , Chlorhydrate d'erlotinib/pharmacologie , Glioblastome/traitement médicamenteux , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Humains , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR , Cellules cancéreuses en culture
8.
Oncotarget ; 6(32): 33867-77, 2015 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-26418954

RÉSUMÉ

BACKGROUND: Glioblastomas (GBM) are often characterized by an elevated expression of the epidermal growth factor receptor variant III (EGFRvIII). We used GBM cell lines with native EGFRvIII expression to determine whether this EGFR variant affects radiosensitivity with or without EGFR targeting. METHODS: Experiments were performed with GBM cell lines lacking (LN229, U87MG, U251, CAS-1) or endogenously expressing EGFRvIII (BS153, DKMG). The two latter cell lines were also used to establish sublines with a low (-) or a high proportion (+) of cells expressing EGFRvIII. EGFR signaling and the cell cycle were analyzed using Western blot and flow cytometry; cell survival was assessed by colony forming assay and double-strand break repair capacity by immunofluorescence. RESULTS: DKMG and BS153 parental cells with heterogeneous EGFRvIII expression were clearly more radiosensitive compared to other GBM cell lines without EGFRvIII expression. However, no significant difference was observed in cell proliferation, clonogenicity or radiosensitivity between the EGFRvIII- and + sublines derived from DKMG and BS153 parental cells. Expression of EGFRvIII was associated with decreased DSB repair capacity for BS153 but not for DKMG cells. The effects of EGFR targeting by gefitinib alone or in combination with irradiation were also found not to depend on EGFRvIII expression. Gefitinib was only observed to influence the proliferation of EGFRvIII- BS153 cells. CONCLUSION: The data indicate that EGFRvIII does not alter radiosensitivity with or without anti-EGFR treatment.


Sujet(s)
Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/radiothérapie , Récepteurs ErbB/métabolisme , Régulation de l'expression des gènes tumoraux , Glioblastome/traitement médicamenteux , Glioblastome/radiothérapie , Quinazolines/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/métabolisme , Cycle cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Séparation cellulaire , Survie cellulaire , Cassures double-brin de l'ADN , Réparation de l'ADN , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/génétique , Cytométrie en flux , Géfitinib , Glioblastome/métabolisme , Humains , Microscopie de fluorescence , Radiotolérance , Transduction du signal , Rayons X
9.
Stem Cells Int ; 2015: 427518, 2015.
Article de Anglais | MEDLINE | ID: mdl-26136784

RÉSUMÉ

Glioblastoma (GBM), the most common malignant brain tumor in adults, contains a subpopulation of cells with a stem-like phenotype (GS-cells). GS-cells can be maintained in vitro using serum-free medium supplemented with epidermal growth factor, basic fibroblast growth factor-2, and heparin. However, this method does not conserve amplification of the Epidermal Growth Factor Receptor (EGFR) gene, which is present in over 50% of all newly diagnosed GBM cases. GS-cells with retained EGFR amplification could overcome the limitations of current in vitro model systems and contribute significantly to preclinical research on EGFR-targeted therapy. This review recapitulates recent methodological approaches to expand stem-like cells from GBM with different EGFR status in order to maintain EGFR-dependent intratumoral heterogeneity in vitro. Further, it will summarize the current knowledge about the impact of EGFR amplification and overexpression on the stem-like phenotype of GBM-derived GS-cells and different approaches to target the EGFR-dependent GS-cell compartment of GBM.

10.
Oncotarget ; 6(14): 12035-47, 2015 May 20.
Article de Anglais | MEDLINE | ID: mdl-26059438

RÉSUMÉ

Acquired resistance to epidermal growth factor receptor (EGFR) targeted antibodies represents a clinical challenge in the treatment of gastrointestinal tumors such as metastatic colorectal cancer, but its molecular mechanisms are incompletely understood. We scanned KRAS exon 2/3/4, NRAS exon 2/3/4 and the overlapping epitopes of the EGFR antibodies cetuximab and panitumumab for mutations in pre- and post-treatment tumor tissue of 21 patients with gastrointestinal cancer treated with chemotherapy +/- EGFR antibodies by next-generation sequencing ("tumor tissue" cohort). We describe a novel EGFR exon 12 mutation acquired in tumors of 1 out of 3 patients treated with panitumumab. The EGFR G465R mutation introduces a positive charge within the overlap of the panitumumab and cetuximab epitopes. It abrogates antibody binding and mediates cross-resistance to both antibodies in EGFR G465R-transfected Ba/F3 cells. In circulating tumor DNA from an independent "liquid biopsy" cohort of 27 patients, we found this novel mutation in 1 out of 6 panitumumab-treated cases while about one third of patients show acquired RAS mutations. We show that acquired resistance by epitope-changing mutations also emerges during panitumumab treatment, which can be easily detected by a liquid biopsy approach even before clinical resistance occurs and this may help in tailoring EGFR-targeted therapies.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Cétuximab/usage thérapeutique , Récepteurs ErbB/génétique , Tumeurs gastro-intestinales/traitement médicamenteux , Adulte , Sujet âgé , Anticorps monoclonaux/administration et posologie , Antinéoplasiques/administration et posologie , Cétuximab/administration et posologie , Récepteurs ErbB/métabolisme , Femelle , Humains , Mâle , Adulte d'âge moyen , Mutation , Panitumumab
11.
J Immunother ; 38(5): 197-210, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25962108

RÉSUMÉ

Natural killer (NK) cells are promising effector cells for adjuvant immunotherapy of cancer. So far, several preclinical studies have shown the feasibility of gene-engineered NK cells, which upon expression of chimeric antigen receptors (CARs) are redirected to otherwise NK cell-resistant tumors. Yet, we reasoned that the efficiency of an immunotherapy using CAR-modified NK cells critically relies on efficient migration to the tumor site and might be improved by the engraftment of a receptor specific for a chemokine released by the tumor. On the basis of the DNAX-activation protein 12 (DAP12), a signaling adapter molecule involved in signal transduction of activating NK cell receptors, we constructed an epidermal growth factor variant III (EGFRvIII)-CAR, designated MR1.1-DAP12 which confers specific cytotoxicity of NK cell towards EGFRvIII glioblastoma cells in vitro and to established subcutaneous U87-MG tumor xenografts. So far, infusion of NK cells with expression of MR1.1-DAP12 caused a moderate but significantly delayed tumor growth and increased median survival time when compared with NK cells transduced with an ITAM-defective CAR. Notably, the further genetic engineering of these EGFRvIII-specific NK cells with the chemokine receptor CXCR4 conferred a specific chemotaxis to CXCL12/SDF-1α secreting U87-MG glioblastoma cells. Moreover, the administration of such NK cells resulted in complete tumor remission in a number of mice and a significantly increased survival when compared with the treatment of xenografts with NK cells expressing only the EGFRvIII-specific CAR or mock control. We conclude that chemokine receptor-engineered NK cells with concomitant expression of a tumor-specific CAR are a promising tool to improve adoptive tumor immunotherapy.


Sujet(s)
Récepteurs ErbB/immunologie , Expression des gènes , Glioblastome/étiologie , Immunothérapie , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Récepteurs CXCR4/génétique , Animaux , Lignée cellulaire , Thérapie cellulaire et tissulaire , Chimiokine CXCL12/biosynthèse , Chimiotaxie/génétique , Chimiotaxie/immunologie , Cytotoxicité immunologique , Modèles animaux de maladie humaine , Expression génique ectopique , Femelle , Ordre des gènes , Thérapie génétique , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique , Glioblastome/métabolisme , Glioblastome/mortalité , Glioblastome/anatomopathologie , Glioblastome/thérapie , Humains , Lentivirus/génétique , Souris , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs CXCR4/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Transduction génétique , Charge tumorale , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Oncotarget ; 6(15): 13579-90, 2015 May 30.
Article de Anglais | MEDLINE | ID: mdl-25944688

RÉSUMÉ

Liquid biopsies come of age offering unexploited potential to monitor and react to tumor evolution. We developed a cost-effective assay to non-invasively determine the immune status of glioblastoma (GBM) patients. Employing newly developed printed peptide microarrays we assessed the B-cell response against tumor-associated antigens (TAAs) in 214 patients. Firstly, sera of long-term (36+ months, LTS, n=10) and short-term (6-10 months, STS, n=14) surviving patients were screened for prognostic antibodies against 1745 13-mer peptides covering known TAAs (TNC, EGFR, GLEA2, PHF3, FABP5, MAGEA3). Next, survival associations were investigated in two retrospective independent multicenter validation sets (n=61, n=129, all IDH1-wildtype). Reliability of measurements was tested using a second array technology (spotted arrays). LTS/STS screening analyses identified 106 differential antibody responses. Evaluating the Top30 peptides in validation set 1 revealed three prognostic peptides. Prediction of TNC peptide VCEDGFTGPDCAE was confirmed in a second set (p=0.043, HR=0.66 [0.44-0.99]) and was unrelated to TNC protein expression. Median signals of printed arrays correlated with pre-synthesized spotted microarrays (p<0.0002, R=0.33). Multiple survival analysis revealed independence of age, gender, KPI and MGMT status. We present a novel peptide microarray immune assay that identified increased anti-TNC VCEDGFTGPDCAE serum antibody titer as a promising non-invasive biomarker for prolonged survival.


Sujet(s)
Autoanticorps/sang , Tumeurs du cerveau/immunologie , Glioblastome/immunologie , Oligopeptides/composition chimique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Séquence d'acides aminés , Tumeurs du cerveau/sang , Tumeurs du cerveau/anatomopathologie , Études de cohortes , Femelle , Glioblastome/sang , Glioblastome/anatomopathologie , Humains , Mâle , Adulte d'âge moyen , Pronostic , Analyse par réseau de protéines/méthodes , Analyse de survie
13.
Neuro Oncol ; 17(8): 1076-85, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-25543125

RÉSUMÉ

BACKGROUND: Signaling by insulin-like growth factor 1 receptor (IGF-1R) can contribute to the formation and progression of many diverse tumor types, including glioblastoma. We investigated the effect of the IGF-1R blocking antibody IMC-A12 on glioblastoma growth in different in vivo models. METHODS: U87 cells were chosen to establish rapidly growing, angiogenesis-dependent tumors in the brains of nude mice, and the GS-12 cell line was used to generate highly invasive tumors. IMC-A12 was administered using convection-enhanced local delivery. Tumor parameters were quantified histologically, and the functional relevance of IGF-1R activation was analyzed in vitro. RESULTS: IMC-A12 treatment inhibited the growth of U87 and GS-12 tumors by 75% and 50%, respectively. In GS-12 tumors, the invasive tumor extension and proliferation rate were significantly reduced by IMC-A12 treatment, while apoptosis was increased. In IMC-A12-treated U87 tumors, intratumoral vascularization was markedly decreased, and tumor cell proliferation was moderately reduced. Flow cytometry showed that <2% of U87 cells but >85% of GS-12 cells expressed IGF-1R. Activation of IGF-1R by IGF-1 and IGF-2 in GS-12 cells was blocked by IMC-A12. Both ligands stimulated GS-12 cell proliferation, and IGF-2 also stimulated migration. IMC-A12 inhibited these stimulatory effects and increased apoptosis. In U87 cells, stimulation with either ligand had no functional effect. CONCLUSIONS: IGF-1R blockade can inhibit glioblastoma growth by different mechanisms, including direct effects on the tumor cells as well as indirect anti-angiogenic effects. Hence, blocking IGF-1R may be useful to target both the highly proliferative, angiogenesis-dependent glioblastoma core component as well as the infiltrative periphery.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Glioblastome/anatomopathologie , Protéine-1 de liaison aux IGF/antagonistes et inhibiteurs , Animaux , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux humanisés , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du cerveau/physiopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Glioblastome/physiopathologie , Humains , Protéine-1 de liaison aux IGF/immunologie , Protéine-1 de liaison aux IGF/physiologie , Souris , Néovascularisation pathologique , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Sci Transl Med ; 6(247): 247ra101, 2014 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-25080476

RÉSUMÉ

Glioblastoma multiforme (GBM) is the most frequent and aggressive brain tumor in adults. The dogma that GBM spread is restricted to the brain was challenged by reports on extracranial metastases after organ transplantation from GBM donors. We identified circulating tumor cells (CTCs) in peripheral blood (PB) from 29 of 141 (20.6%) GBM patients by immunostaining of enriched mononuclear cells with antibodies directed against glial fibrillary acidic protein (GFAP). Tumor cell spread was not significantly enhanced by surgical intervention. The tumor nature of GFAP-positive cells was supported by the absence of those cells in healthy volunteers and the presence of tumor-specific aberrations such as EGFR gene amplification and gains and losses in genomic regions of chromosomes 7 and 10. Release of CTCs was associated with EGFR gene amplification, suggesting a growth potential of these cells. We demonstrate that hematogenous GBM spread is an intrinsic feature of GBM biology.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Glioblastome/secondaire , Cellules tumorales circulantes/anatomopathologie , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/génétique , Tumeurs du cerveau/composition chimique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/chirurgie , Études cas-témoins , Lignée cellulaire tumorale , Chromosomes humains de la paire 10 , Chromosomes humains de la paire 7 , Hybridation génomique comparative , Récepteurs ErbB/génétique , Femelle , Amplification de gène , Protéine gliofibrillaire acide/analyse , Glioblastome/composition chimique , Glioblastome/génétique , Glioblastome/chirurgie , Humains , Immunohistochimie , Hybridation fluorescente in situ , Mâle , Adulte d'âge moyen , Cellules tumorales circulantes/composition chimique
15.
PLoS One ; 9(3): e91663, 2014.
Article de Anglais | MEDLINE | ID: mdl-24621597

RÉSUMÉ

High-frequency stimulation of the subthalamic nucleus (STN-HFS) is widely used as therapeutic intervention in patients suffering from advanced Parkinson's disease. STN-HFS exerts a powerful modulatory effect on cortical motor control by orthodromic modulation of basal ganglia outflow and via antidromic activation of corticofugal fibers. However, STN-HFS-induced changes of the sensorimotor cortex are hitherto unexplored. To address this question at a genomic level, we performed mRNA expression analyses using Affymetrix microarray gene chips and real-time RT-PCR in sensorimotor cortex of parkinsonian and control rats following STN-HFS. Experimental parkinsonism was induced in Brown Norway rats by bilateral nigral injections of 6-hydroxydopamine and was assessed histologically, behaviorally, and electrophysiologically. We applied prolonged (23h) unilateral STN-HFS in awake and freely moving animals, with the non-stimulated hemisphere serving as an internal control for gene expression analyses. Gene enrichment analysis revealed strongest regulation in major histocompatibility complex (MHC) related genes. STN-HFS led to a cortical downregulation of several MHC class II (RT1-Da, Db1, Ba, and Cd74) and MHC class I (RT1CE) encoding genes. The same set of genes showed increased expression levels in a comparison addressing the effect of 6-hydroxydopamine lesioning. Hence, our data suggest the possible association of altered microglial activity and synaptic transmission by STN-HFS within the sensorimotor cortex of 6-hydroxydopamine treated rats.


Sujet(s)
Électrothérapie , Régulation de l'expression des gènes , Antigènes d'histocompatibilité/génétique , Maladie de Parkinson/génétique , Maladie de Parkinson/thérapie , Cortex sensorimoteur/métabolisme , Noyau subthalamique , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Électrodes , Phénomènes électrophysiologiques/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Hypocinésie/complications , Locomotion/effets des médicaments et des substances chimiques , Mâle , Oxidopamine/pharmacologie , Maladie de Parkinson/anatomopathologie , Maladie de Parkinson/physiopathologie , Rats , Cortex sensorimoteur/anatomopathologie , Tyrosine 3-monooxygenase/métabolisme
16.
Acta Neuropathol ; 126(5): 763-80, 2013 Nov.
Article de Anglais | MEDLINE | ID: mdl-24005892

RÉSUMÉ

Fluctuations in oxygen tension during tissue remodeling impose a major metabolic challenge in human tumors. Stem-like tumor cells in glioblastoma, the most common malignant brain tumor, possess extraordinary metabolic flexibility, enabling them to initiate growth even under non-permissive conditions. We identified a reciprocal metabolic switch between the pentose phosphate pathway (PPP) and glycolysis in glioblastoma stem-like (GS) cells. Expression of PPP enzymes is upregulated by acute oxygenation but downregulated by hypoxia, whereas glycolysis enzymes, particularly those of the preparatory phase, are regulated inversely. Glucose flux through the PPP is reduced under hypoxia in favor of flux through glycolysis. PPP enzyme expression is elevated in human glioblastomas compared to normal brain, especially in highly proliferative tumor regions, whereas expression of parallel preparatory phase glycolysis enzymes is reduced in glioblastomas, except for strong upregulation in severely hypoxic regions. Hypoxia stimulates GS cell migration but reduces proliferation, whereas oxygenation has opposite effects, linking the metabolic switch to the "go or grow" potential of the cells. Our findings extend Warburg's observation that tumor cells predominantly utilize glycolysis for energy production, by suggesting that PPP activity is elevated in rapidly proliferating tumor cells but suppressed by acute severe hypoxic stress, favoring glycolysis and migration to protect cells against hypoxic cell damage.


Sujet(s)
Gliome/métabolisme , Glycolyse/physiologie , Cellules souches tumorales/métabolisme , Oxygène/métabolisme , Voie des pentoses phosphates/physiologie , Animaux , Apoptose/physiologie , Hypoxie cellulaire/physiologie , Prolifération cellulaire , Cellules cultivées , Cytométrie en flux , Hétérogreffes , Humains , Immunohistochimie , Souris , Souris nude , Réaction de polymérisation en chaine en temps réel , Transcriptome
17.
Neuro Oncol ; 15(10): 1289-301, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23877316

RÉSUMÉ

BACKGROUND: The treatment efficacy of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors like erlotinib has not met expectations for glioblastoma therapy, even for EGFR-overexpressing tumors. We determined possible mechanisms of therapy resistance using the unique BS153 glioblastoma cell line, which has retained amplification of the egfr gene and expression of EGFR variant (v)III. METHODS: Functional effects of erlotinib, gefitinib, and cetuximab on BS153 proliferation, migration, and EGFR-dependent signal transduction were systematically compared in vitro. The tumor-initiating capacity of parental and treatment-resistant BS153 was studied in Naval Medical Research Institute/Foxn1(nu) mice. Potential mediators of resistance were knocked down using small interfering (si)RNA. RESULTS: Erlotinib and gefitinib inhibited proliferation and migration of BS153 in a dose-dependent manner, whereas cetuximab had no effect. BS153 developed resistance to erlotinib (BS153(resE)) but not to gefitinib. Resistance was associated with strong upregulation of EGFRvIII and subsequent activation of the phosphatidylinositol-3-OH kinase (PI3K) pathway in BS153(resE) and an increased expression of the regulatory 110-kDa delta subunit of PI3K (p110δ). Knockdown of EGFRvIII in BS153(resE) largely restored sensitivity to erlotinib. Targeting PI3K pharmacologically caused a significant decrease in cell viability, and specifically targeting p110δ by siRNA partially restored erlotinib sensitivity in BS153(resE). In vivo, BS153 formed highly invasive tumors with an unusual growth pattern, displaying numerous satellites distant from the initial injection site. Erlotinib resistance led to delayed onset of tumor growth as well as prolonged overall survival of mice without changing tumor morphology. CONCLUSIONS: EGFRvIII can mediate resistance to erlotinib in EGFR-amplified glioblastoma via an increase in PI3Kp110δ. Interfering with PI3Kp110δ can restore sensitivity toward the tyrosine kinase inhibitor.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Récepteurs ErbB/métabolisme , Amplification de gène , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Quinazolines/pharmacologie , Animaux , Apoptose , Technique de Western , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Survie cellulaire , Récepteurs ErbB/génétique , Chlorhydrate d'erlotinib , Cytométrie en flux , Technique d'immunofluorescence , Facteurs de transcription Forkhead/physiologie , Glioblastome/anatomopathologie , Humains , Techniques immunoenzymatiques , Hybridation fluorescente in situ , Souris , Souris nude , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal , Cellules cancéreuses en culture
18.
Neoplasia ; 14(11): 1023-31, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-23226096

RÉSUMÉ

Cetuximab and panitumumab, two antibodies targeting the extracellular domain of the epidermal growth factor receptor (EGFR), are of major clinical importance particularly in the treatment of metastatic colorectal cancer. As patients may acquire resistance-mediating mutations within the extracellular EGFR domain, functional dissection of the exact binding sites of EGFR targeting antibodies may help predict treatment responses. We therefore assessed the epitope recognition of panitumumab by screening phage-displayed random cyclic 7mer and linear 12mer peptide libraries on this antibody. Phage screenings revealed two strong, potentially epitope-mimicking consensus motifs targeted by panitumumab. A computational approach was used to map the sequences back to the potential epitope region on domain III of EGFR. The presumed epitope regions (386)WPEXRT(391) and a biochemically similar though discontinuous region P349-F352-D355 on a neighboring loop of domain III could be confirmed as part of the functionally relevant binding site of panitumumab by site-directed mutational analysis. To more accurately differentiate the panitumumab epitope from the previously characterized cetuximab epitope, binding studies were performed on a broad range of additional mutants. Taken together, this analysis revealed two large, partially overlapping functional epitopes consisting of 17 critical amino acid positions. Four of these positions were selectively targeted by cetuximab (I467, S468, Q408, and H409), whereas another four were selectively recognized by panitumumab (W386, E388, R390, and T391). In view of the clinical significance of extracellular domain mutations, our data may help guide treatment decisions in selected patients receiving EGFR-targeted therapies.


Sujet(s)
Anticorps monoclonaux/immunologie , Cartographie épitopique , Épitopes/immunologie , Récepteurs ErbB/immunologie , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/métabolisme , Anticorps monoclonaux humanisés , Techniques d'exposition à la surface cellulaire , Cétuximab , Épitopes/composition chimique , Épitopes/génétique , Épitopes/métabolisme , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Humains , Modèles moléculaires , Mimétisme moléculaire/immunologie , Mutation , Panitumumab , Banque de peptides , Peptides/immunologie , Peptides/métabolisme , Liaison aux protéines , Conformation des protéines
19.
PLoS One ; 7(8): e43468, 2012.
Article de Anglais | MEDLINE | ID: mdl-22927971

RÉSUMÉ

Glioblastomas are highly aggressive brain tumors of adults with poor clinical outcome. Despite a broad range of new and more specific treatment strategies, therapy of glioblastomas remains challenging and tumors relapse in all cases. Recent work demonstrated that the posttranslational hypusine modification of the eukaryotic initiation factor 5A (eIF-5A) is a crucial regulator of cell proliferation, differentiation and an important factor in tumor formation, progression and maintenance. Here we report that eIF-5A as well as the hypusine-forming enzymes deoxyhypusine synthase (DHS) and deoxyhypusine hydroxylase (DOHH) are highly overexpressed in glioblastoma patient samples. Importantly, targeting eIF-5A and its hypusine modification with GC7, a specific DHS-inhibitor, showed a strong antiproliferative effect in glioblastoma cell lines in vitro, while normal human astrocytes were not affected. Furthermore, we identified p53 dependent premature senescence, a permanent cell cycle arrest, as the primary outcome in U87-MG cells after treatment with GC7. Strikingly, combined treatment with clinically relevant alkylating agents and GC7 had an additive antiproliferative effect in glioblastoma cell lines. In addition, stable knockdown of eIF-5A and DHS by short hairpin RNA (shRNA) could mimic the antiproliferative effects of GC7. These findings suggest that pharmacological inhibition of eIF-5A may represent a novel concept to treat glioblastomas and may help to substantially improve the clinical course of this tumor entity.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Glioblastome/génétique , Lysine/analogues et dérivés , Mixed function oxygenases/génétique , Thérapie moléculaire ciblée , Oxidoreductases acting on CH-NH group donors/génétique , Facteurs initiation chaîne peptidique/génétique , Protéines de liaison à l'ARN/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antinéoplasiques alcoylants/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Carmustine/pharmacologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Dacarbazine/analogues et dérivés , Dacarbazine/pharmacologie , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Techniques de knock-down de gènes , Glioblastome/traitement médicamenteux , Glioblastome/enzymologie , Glioblastome/anatomopathologie , Guanine/analogues et dérivés , Guanine/pharmacologie , Humains , Lysine/biosynthèse , Mâle , Mixed function oxygenases/métabolisme , Grading des tumeurs , Oxidoreductases acting on CH-NH group donors/antagonistes et inhibiteurs , Oxidoreductases acting on CH-NH group donors/déficit , Oxidoreductases acting on CH-NH group donors/métabolisme , Facteurs initiation chaîne peptidique/déficit , Témozolomide ,
20.
Clin Cancer Res ; 18(7): 1901-13, 2012 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-22316604

RÉSUMÉ

PURPOSE: Despite the high incidence of epidermal growth factor receptor (EGFR) gene amplification and rearrangement in glioblastomas, no suitable cell line exists that preserves these alterations in vitro and is tumorigenic in immunocompromised mice. On the basis of previous observations that glioblastoma cells cultured with serum lose the EGFR amplification rapidly and that EGF can inhibit the growth of EGFR-amplified tumor cells, we hypothesized that serum-free and EGF-free culture conditions could promote maintenance of the EGFR amplification. EXPERIMENTAL DESIGN: Cells from EGFR-amplified glioblastomas were taken into culture using neural stem cell conditions with modifications, including varying oxygen concentrations and omission of routine EGF supplementation. RESULTS: High-level EGFR amplification was rapidly lost in 5 glioblastoma cultures supplemented with EGF, whereas it was preserved in cultures from the same tumors established without EGF. Cultures from 2 glioblastomas developed into pairs of cell lines, with either stable maintenance or irreversible loss of high-level EGFR amplification in the majority of cells. One EGFR-amplified cell line preserved expression of the receptor variant EGFRvIII. Cell lines with high-level EGFR amplification/EGFRvIII expression formed highly aggressive tumors in nude mice, whereas nonamplified cell lines were either nontumorigenic or grew significantly more slowly. In contrast, nonamplified cell lines proliferated faster in vitro. All cell lines responded to erlotinib, with inhibition of receptor activation and proliferation but partly different effects on downstream signaling and migration. CONCLUSIONS: Isogenic glioblastoma cell lines maintaining stable differences in EGFR/EGFRvIII status can be derived by varying exposure to EGF ligand and reflect the intratumoral genetic heterogeneity.


Sujet(s)
Facteur de croissance épidermique/pharmacologie , Récepteurs ErbB/génétique , Amplification de gène/effets des médicaments et des substances chimiques , Glioblastome/métabolisme , Animaux , Technique de Western , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Récepteurs ErbB/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Glioblastome/génétique , Glioblastome/anatomopathologie , Humains , Immunohistochimie , Estimation de Kaplan-Meier , Ligands , Souris , Lignées consanguines de souris , Souris nude , Tumeurs expérimentales/génétique , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , RT-PCR , Transplantation hétérologue , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...