Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Adrian Staab is active.

Publication


Featured researches published by Adrian Staab.


Investigative Radiology | 2008

Computed tomography, positron emission tomography, positron emission tomography/computed tomography, and magnetic resonance imaging for staging of limited pleural mesothelioma: initial results.

Christian Plathow; Adrian Staab; Astrid Schmaehl; Philip Aschoff; Ivan Zuna; Christina Pfannenberg; Schlemmer Heinz Peter; Susanne Eschmann; Michael Klopp

Objective:To evaluate and compare the role of computed tomography (CT), positron emission tomography (PET), PET/CT, and magnetic resonance imaging (MRI) in the correct staging of patients with limited malignant pleural mesothelioma (MPM). Materials and Methods:Fifty-four patients with an epithelial MPM (34 men and 20 women) were included in this study. Patients were referred to our department for staging in a predicted resectable state (stage II/III). Within 3 days, PET/CT and MRI was performed in all patients. Images were evaluated by 3 specialists in the field of PET/CT and MRI. The subexaminations of PET/CT, PET, and CT were independently evaluated with respect to tumor stage. Subexaminations were compared with each other, with MRI and PET/CT. N-stage was verified by mediastinoscopy. Afterward, consensus reading was performed. In 52 patients, surgery served as gold standard. In 2 patients, follow-up control served as gold standard as an inoperable situation with distant metastases was found. Additionally, interobserver variability (&kgr; value) was calculated. Results:In stage II, accuracy was 0.77 (CT), 0.86 (PET), 0.8 (MRI), 1.0 (PET/CT), and in stage III 0.75, 0.83, 0.9, 1.0. PET/CT was significantly more accurate (P < 0.05) in stages II and III compared with all other techniques. CT and MRI were not able to detect distant metastases in 2 patients, which changed therapy (operable vs. inoperable). Interobserver variability was 0.7, 0.9, 0.8, 1.0 in stage II and 0.9, 0.9, 0.9, 1.0 in stage III. Conclusion:PET/CT makes it possible to stage patients with limited MPM with high accuracy and low interobserver variability.


Current Pharmaceutical Design | 2010

Modulation of Carbonic Anhydrase 9 (CA9) in Human Brain Cancer

Harun M. Said; Claudiu T. Supuran; Carsten Hageman; Adrian Staab; Buelent Polat; Astrid Katzer; Andrea Scozzafava; Jelena Anacker; Michael Flentje; Dirk Vordermark

Hypoxia is a crucial factor in tumour aggressiveness and its treatment resistance, particularly in human brain cancer. Tumour resistance against radiation- and chemo- therapy is facilitated by oxygenation reduction at tumour areas. HIF-1α regulated genes are mostly responsible for this type of resistance. Among these genes, carbonic anhydrase isoform 9 (CA9) is highly overexpressed in many types of cancer especially in high grade brain cancer like GBM. CA IX contributes to tumour environment acidification by catalyzing the carbon dioxide hydration to bicarbonate and protons, leading to the acquisition of metastasic phenotypes and chemoresistance to weakly basic anticancer drugs and therefore to inadequate application of radio-therapeutic or chemotherapeutic anti-cancer treatment strategies. Inhibition of this enzymatic activity by application of specific chemical CA9 inhibitors (sulphonamide derivative compounds) or indirect inhibitors like HIF-1α inhibitors (chetomin) or molecular inhibitors like CA9-siRNA leads to reversion of these processes, leading to the CA9 functional role inhibition during tumourigenesis. Hypoxia significantly influences the tumour microenvironment behaviour via activation of genes involved in the adaptation to the hypoxic stress. It also represents an important cancer prognosis indicator and is associated with aggressive growth, malignant progression, metastasis and poor treatment response. The main objective in malignant GBM therapy is either to eradicate the tumour or to convert it into a controlled, quiescent chronic disease. Sulfonamide derivative compounds with CA9 inhibitory characteristics represent one of the optimal treatment options beside other CA9 inhibitory agents or chemical inhibitory compounds against its main regulating transcription factor which is the hypoxia induced HIF-1α when applied against human cancers with hypoxic regions like GBM, bearing potential for an effective role in human brain tumour therapeutic strategies. Glycolytic inhibitors, when added in controlled doses under hypoxia, lead to a reduced accumulation of HIF-1α and can function as indirect hypoxia regulated genes inhibitors like CA9. These may be used as alternative or in conjunction with other direct inhibitors like the sulphonamide derivate compounds, chetomin or specific siRNAs, or other different chemical compounds possessing similar functionality making them as optimal tools for optimized therapy development in cancer treatment, especially against human brain cancer. Further experimental analysis towards the tumour stage specific inhibitory CA9 characteristics determination are necessary to find the optimal therapeutic solutions among the different available modalities; whether they are direct or indirect chemical, molecular or natural inhibitors to be able to set up successful treatment approaches against the different human tumour diseases.


Bioorganic & Medicinal Chemistry | 2013

Hypoxia induced CA9 inhibitory targeting by two different sulfonamide derivatives including Acetazolamide in human Glioblastoma

Harun M. Said; Carsten Hagemann; Fabrizio Carta; Astrid Katzer; Bülent Polat; Adrian Staab; Andrea Scozzafava; Jelena Anacker; Giles Hamilton Vince; Michael Flentje; Claudiu T. Supuran

HIF-1α regulated genes are mainly responsible for tumour resistance to radiation- and chemo-therapy. Among these genes, carbonic anhydrase isoform IX (CA9) is highly over expressed in many types of cancer especially in high grade brain cancer like Glioblastoma (GBM). Inhibition of the enzymatic activity by application of specific chemical CA9 inhibitor sulphonamides (CAI) like Acetazolamide (Aza.), the new sulfonamide derivative carbonic anhydrase inhibitor (SU.D2) or indirect inhibitors like the HIF-1α inhibitor Chetomin or molecular inhibitors like CA9-siRNA are leading to an inhibition of the functional role of CA9 during tumorigenesis. Human GBM cells were treated with in vitro hypoxia (1, 6, or 24 h at 0.1%, O2). Aza. application was at a range between 250 and 8000 nM and the HIF-1α inhibitor Chetomin at a concentration range of 150-500 nM. Cell culture plates were incubated for 24 h under hypoxia (0.1% O2). Further, CA9-siRNA constructs were transiently transfected into GBM cells exposed to extreme hypoxic aeration conditions. CA9 protein expression level was detectable in a cell-type specific manner under normoxic conditions. Whereas U87-MG exhibited a strong aerobic expression, U251 and U373 displayed moderate and GaMG very weak normoxic CA9 protein bands. Aza. as well as SU.D2 displayed inhibitory characteristics to hypoxia induced CA9 expression in the four GBM cell lines for 24 h of hypoxia (0.1% O2) at concentrations between 3500 and 8000 nM, on both the protein and mRNA level. Parallel experiments using CA9-siRNA confirmed these results. Application of 150-500 nM of the glycolysis inhibitor Chetomin under similar oxygenation conditions led to a sharply reduced expression of both CA IX protein and CA9 mRNA levels, indicating a clear glucose availability involvement for the hypoxic HIF-1α and CA9 expression in GBM cells. Hypoxia significantly influences the behaviour of human tumour cells by activation of genes involved in the adaptation to hypoxic stress. The main objective in malignant GBM therapy is either to eradicate the tumour or to convert it into a controlled, quiescent chronic disease. Aza., SU.D2, Chetomin or CA9-siRNA possesses functional CA9 inhibitory characteristics when applied against human cancers with hypoxic regions like GBM. They may be used as alternative or in conjunction with other direct inhibitors possessing similar functionality, thereby rendering them as potential optimal tools for the development of an optimized therapy in human brain cancer treatment.


Strahlentherapie Und Onkologie | 2011

Small interfering RNA targeting HIF-1α reduces hypoxia-dependent transcription and radiosensitizes hypoxic HT 1080 human fibrosarcoma cells in vitro

Adrian Staab; Markus Fleischer; Juergen Loeffler; Harun M. Said; Astrid Katzer; Christian Plathow; H. Einsele; Michael Flentje; Dirk Vordermark

Background:Hypoxia inducible factor-1 has been identified as a potential target to overcome hypoxia-induced radioresistance The aim of the present study was to investigate whether selective HIF-1 inhibition via small interfering RNA (siRNA) targeting hypoxia-inducible factor 1α (HIF-1α) affects hypoxia-induced radioresistance in HT 1080 human fibrosarcoma cells.Material and Methods:HIF-1α expression in HT 1080 human fibrosarcoma cells in vitro was silenced using HIF-1α siRNA sequence primers. Quantitative real-time polymerase chain reaction assay was performed to quantify the mRNA expression of HIF-1α. HIF-1α protein levels were studied by Western blotting at 20% (air) or after 12 hours at 0.1% O2 (hypoxia). Cells were assayed for clonogenic survival after irradiation with 2, 5, or 10 Gy, under normoxic or hypoxic conditions in the presence of HIF-1α-targeted or control siRNA sequences. A modified oxygen enhancement ratio (OER´) was calculated as the ratio of the doses to achieve the same survival at 0.1% O2 as at ambient oxygen tensions. OER´ was obtained at cell survival levels of 50%, 37%, and 10%.Results:HIF-1α-targeted siRNA enhanced radiation treatment efficacy under severely hypoxic conditions compared to tumor cells treated with scrambled control siRNA. OER was reduced on all survival levels after treatment with HIF-1α-targeted siRNA, suggesting that inhibition of HIF-1 activation by using HIF-1α-targeted siRNA increases radiosensitivity of hypoxic tumor cells in vitro.Conclusion:Inhibition of HIF-1 activation by using HIF-1α-targeted siRNA clearly acts synergistically with radiotherapy and increase radiosensitivity of hypoxic cells in vitro.Hintergrund und Ziel:Hypoxia-inducible Factor-1 (HIF-1) wurde als potentielles therapeutisches Target identifiziert. Ziel der Arbeit war es, zu untersuchen, ob die selektive HIF-1-Inhibition mittels Small Interfering RNA (siRNA) gegen HIF-1α die Strahlensensibilität von hypoxischen HT-1080-Zellen beeinflussen kann.Material und Methodik: Die HIF-1α-Expression in humanen HT-1080-Fibrosarkomzellen wurde mittels RNA-Interferenz nach Transfektion der Zellen mit siRNA unter hypoxischen Bedingungen (0,1%, O2, 12 h), bzw. Normoxie (20% O2) in vitro inhibiert. Die HIF-1α-Genexpression wurde mit quantitativer Realtime-Polymerasekettenreaktion (qRT-PCR), das HIF-1α-Protein mittels Western Blot quantifiziert. Das klonogene Überleben wurde nach Bestrahlung unter Hypoxie und Normoxie bestimmt und daraus eine Oxygen Enhancement Ratio (OER´) bei den Überlebensniveaus 50%, 37% and 10% berechnet.Resultate:HIF-1α-siRNA erhöht die Strahlensensibilität unter hypoxischen Bedingen, verglichen mit HT-1080-Zellen, die mit Kontroll-siRNA behandelt wurden. Die OER` war bei allen Überlebensniveaus reduziert. Schlussfolgerung:Eine selektive Inhibition der HIF-1-Aktivierung durch HIF-1α-siRNA wirkt synergistisch mit einer Bestrahlung und erhöht die Strahlensensitivität hypoxischer Tumorzellen in vitro.


Strahlentherapie Und Onkologie | 2007

Modulation of Glucose Metabolism Inhibits Hypoxic Accumulation of Hypoxia-Inducible Factor-1α (HIF-1α)

Adrian Staab; Jürgen Löffler; Harun M. Said; Astrid Katzer; Melanie Beyer; Bülent Polat; Hermann Einsele; Michael Flentje; Dirk Vordermark

Background and Purpose:The hypoxic accumulation of the transcription factor subunit hypoxia-inducible factor-1α (HIF-1α), a potential endogenous hypoxia marker and therapeutic target, has recently been shown to strongly depend on glucose availability. The aim of this study was to investigate the underlying mechanism of this effect.Material and Methods:HIF-1α protein levels were studied by Western blotting in HT 1080 human fibrosarcoma cells and in a hypoxia-responsive element green fluorescent protein (HRE-GFP) reporter assay in stably transfected HT 1080 cells treated with hypoxia (0.1% O2, 12 h) and glycolysis inhibitors 2-deoxyglucose (2-DG) or iodoacetate (IAA). HIF-1α mRNA expression was quantified via real-time polymerase chain reaction (RT-PCR).Results:Both inhibitors drastically reduced hypoxic HIF-1α accumulation (2-DG + hypoxia 2% mean HIF-1α protein level vs. 59% hypoxia alone; IAA + hypoxia 13% mean HIF-1α protein level vs. 96% hypoxia alone), an effect not rescued by the addition of pyruvate and confirmed in an HRE-GFP reporter assay in stably transfected HT 1080 cells. RT-PCR under identical conditions showed no effect of glycolysis inhibition on HIF-1α mRNA levels, suggesting a translational or posttranslational mechanism.Conclusion:The effect of glycolysis modulation on the HIF-1α levels in tumor cells may provide a novel approach to therapeutically target HIF-1α.Hintergrund und Ziel:Die hypoxische Akkumulation des Hypoxia-inducible factor-1α (HIF-1α), eines potentiellen Hypoxiemarkers und therapeutischen Targets, ist in hohem Maße von der Glucoseverfügbarkeit abhängig. Ziel dieser Arbeit war es, den zugrundeliegenden Mechanismus zu untersuchen.Material und Methodik:Das Protein HIF-1α wurde in humanen HT-1080-Fibrosarkomzellen nach Behandlung der Zellen mit den Glykolyseinhibitoren 2-Desoxy-D-Glucose (2-DG) und Iodacetat (IAA) unter hypoxischen Bedingungen (0,1% O2, 12 h) mittels Western-Blot und mit einem HRE-GFP-(„hypoxia-responsive element green fluorescent protein“-)Reporter-Assay in stabil transfizierten HT-1080-Zellen detektiert. Die Expressionsrate der HIF-1α-mRNA wurde mit Real-Time-Polymerase-Kettenreaktion (RTPCR) gemessen.Ergebnisse:Nach Zugabe von Glykolyseinhibitoren zeigte sich eine deutlich verringerte hypoxische HIF-1α-Akkumulation (Abbildungen 1, 2 und 5: 2-DG + Hypoxie: 2% mittleres HIF-1α-Protein-Level vs. 59% Hypoxie ohne 2-DG; IAA + Hypoxie: 13% mittleres HIF-1α-Protein-Level vs. 96% Hypoxie ohne IAA), die durch die Zugabe von Pyruvat nicht rückgängig gemacht werden konnte (Abbildung 3). Diese Ergebnisse wurden mit einem HRE-GFP-Reporter-Assay in stabil transfizierten HT-1080-Zellen bestätigt (Abbildung 4). Die HIF-1α-RT-PCR zeigte keine veränderten Expressionsraten von HIF-1α-mRNA nach Zugabe von IAA bzw. 2-DG (Abbildung 6). Diese Daten deuten auf einen translationalen bzw. posttranslationalen Mechanismus der Inhibition der hypoxischen HIF-1a-Akkumulation durch Glykolyseinhibitoren hin.Schlussfolgerung:Der Einfluss einer Modulation der Glykolyse auf HIF-1α-Level in Tumorzellen könnte einen neuen Ansatz einer HIF-1α-gerichteten Therapie darstellen.


Current Medicinal Chemistry | 2008

Detection and Specific Targeting of Hypoxic Regions within Solid Tumors: Current Preclinical and Clinical Strategies

Matthias Bache; Matthias Kappler; Harun M. Said; Adrian Staab; Dirk Vordermark


Radiotherapy and Oncology | 2007

Expression patterns of the hypoxia-related genes osteopontin, CA9, erythropoietin, VEGF and HIF-1α in human glioma in vitro and in vivo

Harun M. Said; Carsten Hagemann; Adrian Staab; Jelena Stojic; Siglinde Kühnel; Giles Hamilton Vince; Michael Flentje; Klaus Roosen; Dirk Vordermark


BMC Cancer | 2007

Effects of HIF-1 inhibition by chetomin on hypoxia-related transcription and radiosensitivity in HT 1080 human fibrosarcoma cells

Adrian Staab; Jürgen Loeffler; Harun M. Said; Désirée Diehlmann; Astrid Katzer; Melanie Beyer; Markus Fleischer; Franz Schwab; Kurt Baier; Hermann Einsele; Michael Flentje; Dirk Vordermark


Journal of Neuro-oncology | 2006

Distinct patterns of hypoxic expression of carbonic anhydrase IX (CA IX) in human malignant glioma cell lines.

Harun M. Said; Adrian Staab; Carsten Hagemann; Giles Hamilton Vince; Astrid Katzer; Michael Flentje; Dirk Vordermark


Oncology Reports | 1994

Oxygen-dependent regulation of NDRG1 in human glioblastoma cells in vitro and in vivo

Harun M. Said; Susanne Stein; Carsten Hagemann; Buelent Polat; Adrian Staab; Jelena Anacker; Beate Schoemig; Matthias Theobald; Michael Flentje; Dirk Vordermark

Collaboration


Dive into the Adrian Staab's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge