Susanne Gahr
University of Marburg
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Susanne Gahr.
British Journal of Cancer | 2013
Susanne Gahr; R Stoehr; E Geissinger; Joachim H. Ficker; Wolfgang M. Brueckl; A Gschwendtner; S Gattenloehner; Florian S. Fuchs; C Schulz; Ralf J. Rieker; Arndt Hartmann; P Ruemmele; W Dietmaier
Background:The prognosis of metastatic non-small cell lung cancer (NSCLC) is still poor. Activating epithelial growth factor receptor (EGFR) mutations are important genetic alterations with dramatic therapeutical implications. Up to now, in contrast to Asian populations only limited data on the prevalence of those mutations are available from patients with Caucasian and especially European ethnicity.Methods:In this multicentre study, 1201 unselected NSCLC patients from Southern Germany were tested in the daily clinical routine for EGFR mutation status.Results:Activating EGFR mutations were found in 9.8% of all tumours. Mutations in exons 18, 19 and 21 accounted for 4.2%, 61.9% and 33.1% of all mutations, respectively. Non-smokers had a significantly higher rate of EGFR mutations than smokers or ex-smokers (24.4% vs 4.2%; P<0.001). Non-lepidic-non-mucinous adenocarcinomas (G2) accounted for 45.5% of all activating EGFR mutations and 3.5% of all squamous cell carcinomas were tested positive. Thyroid transcription factor 1 protein expression was significantly associated with EGFR mutational status.Conclusion:These comprehensive data from clinical routine in Germany add to the knowledge of clinical and histopathological factors associated with EGFR mutational status in NSCLC.
Analytical Cellular Pathology | 2010
Pietro Di Fazio; Regine Schneider-Stock; Daniel Neureiter; Kinya Okamoto; Tt Wissniowski; Susanne Gahr; Karl Quint; Matthias Meissnitzer; Beate Alinger; Roberta Montalbano; Gabriele Sass; Bernd Hohenstein; Eckhart G. Hahn; Matthias Ocker
Inhibition of deacetylases represents a new treatment option for human cancer diseases. We applied the novel and potent pan-deacetylase inhibitor panobinostat (LBH589) to human hepatocellular carcinoma models and investigated by which pathways tumor cell survival is influenced. HepG2 (p53wt) and Hep3B (p53null) responded to panobinostat treatment with a reduction of cell proliferation and a significant increase in apoptotic cell death at low micromolar concentrations. Apoptosis was neither mediated by the extrinsic nor the intrinsic pathway but quantitative RT-PCR showed an upregulation of CHOP, a marker of the unfolded protein response and endoplasmic reticulum stress with subsequent activation of caspase 12. Dependent on the p53 status, a transcriptional upregulation of p21cip1/waf1, an increased phosphorylation of H2AX, and an activation of the MAPK pathway were observed. In a subcutaneous xenograft model, daily i.p. injections of 10 mg/kg panobinostat lead to a significant growth delay with prolonged overall survival, mediated by reduced tumor cell proliferation, increased apoptosis and reduced angiogenesis in tumor xenografts. Panobinostat increased the acetylation of histones H3 and H4. Panobinostat is a well tolerated new treatment option for HCC that activates alternative pathways of apoptosis, also in p53-deficient tumors.
Pancreatology | 2009
Karl Quint; Sebastian Stintzing; Beate Alinger; Cornelia Hauser-Kronberger; Otto Dietze; Susanne Gahr; Eckhart G. Hahn; Matthias Ocker; Daniel Neureiter
Background and Aims: Pancreatic cancer cells have been shown to possess stem-cell-like properties, especially by reactivating embryonic transcription factors involved in tissue differentiation. We therefore investigated if and to what extent developmental genes of the human pancreas are expressed in pancreatic ductal adenocarcinomas and precursor lesions, pancreatic intraepithelial neoplasia (PanIN), and if this correlates or predicts response to treatment and overall survival. Material and Methods: Invasive ductal adenocarcinomas of the pancreas [UICC pT3pN0 (n = 13) vs. pT3pN1 (n = 25)] and tumors after neoadjuvant chemotherapy [5-fluorouracil (FU)/folic-acid and gemcitabine; UICC ypN0 (n = 7) vs. ypN1 (n = 6)] resected between 1997 and 2003 were characterized histochemically and immunohistochemically [pancreas duodenum homeobox 1 (PDX-1), Sonic hedgehog protein (SHH), Patched (Ptc) and Gli-1]. Gene distribution was compared with morphological patterns of the pancreatic carcinoma and PanIN as well as with peritumorous reactions of normal pancreas. Results: The overall expression of PDX-1, SHH, Ptc and Gli-1 was low, but showed a distinctive and topographic linkage inside pancreatic carcinomas as well as inside PanINs. Additionally, a topographic and significant association of these markers with nodal status (PDX-1, Ptc, Gli-1), tumor size (PDX-1, Gli-1) and R status (PDX-1) was found. After stratification with the strongest outcome predictor, grading, survival analysis revealed that Ptc expression in grade 2 and PDX-1 expression in grade 3 carcinomas are independent survival factors. Conclusions: Markers of pancreas development are reexpressed in invasive ductal adenocarcinomas and their expression is essentially associated with general clinical and pathological features such as survival or nodal status.
BMC Cancer | 2012
Steffen Zopf; Matthias Ocker; Daniel Neureiter; Beate Alinger; Susanne Gahr; Markus F. Neurath; Pietro Di Fazio
BackgroundHepatocellular carcinoma (HCC) still represents an unmet medical need. Epigenetic inactivation of tumor suppressor genes like RASSF1A or APC by overexpression of DNA methyltransferases (DNMTs) has been shown to be common in HCC and to be linked to the overall prognosis of patients. Inhibitors of protein and histone deacetylases (DACi) have been demonstrated to possess strong anti-tumor effects in HCC models.MethodsWe therefore investigated whether DACi also has any influence on the expression and activity of DNMTs and methylated target genes in HepG2 and Hep3B cell culture systems and in a xenograft model by immunohistochemistry, westernblotting, RT-qPCR and methylation-specific PCR.ResultsOur findings demonstrate a rapid inhibition of DNMT activity 6 h after treatment with 0.1 μM of the pan-DACi panobinostat. A downregulation of DNMT mRNAs and protein were also observed at later points in time. This loss of DNMT activity and expression was paralleled by a diminished methylation of the target genes RASSF1A and APC and a concomitant re-expression of APC mRNA and protein. Analysis of HepG2 xenograft specimens confirmed these results in vivo.ConclusionWe suggest a dual mode of action of DACi on DNA methylation status: a rapid inhibition of enzyme activity due to interference with posttranslational acetylation and a delayed effect on transcriptional control of DNMT genes by HDAC or miRNA mechanisms.
Journal of Cancer | 2012
Susanne Gahr; Tt Wissniowski; Steffen Zopf; D Strobel; Anette Pustowka; Matthias Ocker
Advanced hepatocellular carcinoma still represents an unmet medical need that has only a limited overall survival despite the introduction of the multi-kinase inhibitor sorafenib. Recently, inhibitors of histone and other protein deacetylases have been established as novel therapeutic approaches to cancer diseases. We here review the molecular rationale for combining these two novel targeted therapies and report a patient with metastasized hepatocellular carcinoma who showed a partial remission of primary and metastatic lesions for five months after a combination therapy with sorafenib and the orally available pan-deacetylase inhibitor panobinostat.
Nuclear Medicine and Biology | 2016
Simone Maschauer; Susanne Gahr; Muktheshwar Gandesiri; Philipp Tripal; Regine Schneider-Stock; Torsten Kuwert; Matthias Ocker; Olaf Prante
INTRODUCTION Deacetylase inhibitors have recently been established as a novel therapeutic approach to solid and hematologic cancers and have also been demonstrated to possess anti-angiogenic properties. Although these compounds show a good efficacy in vitro and in vivo, no data on monitoring and predicting treatment response are currently available. We therefore investigated the effect of the pan-deacetylase inhibitor panobinostat (LBH589) on gastrointestinal cancer models and the suitability of 2-[(18)F]FGlc-RGD as a specific agent for imaging integrin αvβ3 expression during tumor angiogenesis using small animal positron emission tomography (PET). METHODS The effect of panobinostat on cell viability in vitro was assessed with a label-free impedance based real-time analysis. Nude mice bearing HT29 and HepG2 tumors were treated with daily i.p. injections of 10mg/kg panobinostat for 4 weeks. During this time, tumor size was determined with a calliper and mice were repeatedly subjected to PET imaging. Tumor tissues were analyzed immunohistochemically with a focus on proliferation and endothelial cell markers (Ki-67, Meca-32) and by Western blot applying specific markers of apoptosis. RESULTS In vitro, panobinostat inhibited the proliferation of HepG2 and HT29 cells. Contrary to the situation in HepG2 tumors in vivo, where panobinostat treatment is known to reduce proliferation and vascularization resulting in a decreased tumor growth, HT29 tumors did not show any effect on these parameters. We demonstrated by Western blotting, that panobinostat induced apoptosis in HT29 tumors in vivo. Longitudinal PET imaging studies in HepG2 tumor-bearing mice using 2-[(18)F]FGlc-RGD demonstrated that the standard uptake value (SUVmax) in HepG2 tumors was significantly decreased by 39% at day 7 after treatment. The comparative PET study using HT29 tumor-bearing animals did not reveal any response of the tumors to panobinostat treatment. CONCLUSIONS Small-animal PET imaging using 2-[(18)F]FGlc-RGD was successfully applied to the non-invasive monitoring of the HepG2-tumor response to panobinostat in nude mice early after begin of treatment. Thus, PET imaging of angiogenesis using 2-[(18)F]FGlc-RGD could be a valuable tool to monitor panobinostat therapy in further preclinical studies. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE When successfully translated to the clinical surrounding, PET imaging of angiogenesis could therefore facilitate therapy planning and monitoring of therapy success with panobinostat in hepatocellular carcinoma.
Zeitschrift Fur Gastroenterologie | 2009
Susanne Gahr; G. Peter; Tth Wissniowski; E. G. Hahn; Christoph Herold; M Ocker
Hintergrund: Effektive Therapien fur fortgeschrittene Stadien des hepatozellularen Karzinoms (HCC) konnten bislang nicht entwickelt werden. Wir haben untersucht, inwieweit eine Kombination des Histondeazetylaseinhibitors MS-275 und des CDK-Inhibitors CYC-202 zusammenwirkt, um das Proliferationsverhalten von Hepatomzellen in vitro zu inhibieren und deren Apoptose zu fordern . Methoden: Die humanen Hepatomzelllinien HEP3B und HEPG2 sowie die Kontrollzelllinie humaner Vorhautfibroblasten wurden unter Standardbedingungen kultiviert und mit ansteigenden Konzentrationen von CYC-202 und MS-275 als Einzelsubstanzen oder in Kombination inkubiert. Nach 24 bis 72h erfolgte eine Bestimmung der Apoptoserate mittels Durchflusszytometrie (Propidiumjodid, JC-1) und Immunhistochemie fur Zytokeratin 18-Fragmentierung. Die DNA-Synthese wurde mittels BrdU-Inkorporations-ELISA bestimmt. Gesamtprotein wurde fur Westernblots gegen p21, Bax und Bcl-2 sowie fur fluorimetrische Aktivitatsassays gegen Caspase 3 und Caspase 8 isoliert. Ergebnisse: Die Kombination von CYC-202 und MS-275 fuhrt zu besseren pro-apoptotischen Effekten als die Verwendung von Einzelsubstanzen. Apoptose wurde uber eine Aktivierung des Mitochondrien-vermittelten Signaltransduktionsweges induziert, offensichtlich uber eine Verschiebung in der bax/bcl-2 ratio und einen Zusammenbruch des Mitochondrienmembranpotentials. Caspase Assays erzielten eine starke Aktivierung der Caspase 3, aber keine Aktivierung des extrinsischen Initiators Caspase 8. Schlussfolgerungen: Die Kombinationstherapie mit den Biomodulatoren MS-275 und CYC-202 ist eine vielversprechende Behandlungsoption fur das HCC.
International Journal of Oncology | 2009
Samir Jabari; Matthias Meissnitzer; Karl Quint; Susanne Gahr; Tt Wissniowski; Eckhart G. Hahn; Daniel Neureiter; Matthias Ocker
Oncology Reports | 1994
Susanne Gahr; Gisela Peter; Thadäus Till Wissniowski; E. G. Hahn; Christoph Herold; Matthias Ocker
International Journal of Oncology | 2007
Susanne Gahr; Matthias Ocker; Marion Ganslmayer; Steffen Zopf; Kinya Okamoto; Andrea Hartl; Sandra Leitner; Eckhart G. Hahn; Christoph Herold