20 results on '"Roland T Ullrich"'
Search Results
2. In-vivo visualization of tumor microvessel density and response to anti-angiogenic treatment by high resolution MRI in mice.
- Author
-
Roland T Ullrich, Jan F Jikeli, Michael Diedenhofen, Philipp Böhm-Sturm, Maike Unruh, Stefan Vollmar, and Mathias Hoehn
- Subjects
Medicine ,Science - Abstract
PURPOSE: Inhibition of angiogenesis has shown clinical success in patients with cancer. Thus, imaging approaches that allow for the identification of angiogenic tumors and the detection of response to anti-angiogenic treatment are of high clinical relevance. EXPERIMENTAL DESIGN: We established an in vivo magnetic resonance imaging (MRI) approach that allows us to simultaneously image tumor microvessel density and tumor vessel size in a NSCLC model in mice. RESULTS: Using microvessel density imaging we demonstrated an increase in microvessel density within 8 days after tumor implantation, while tumor vessel size decreased indicating a switch from macro- to microvessels during tumor growth. Moreover, we could monitor in vivo inhibition of angiogenesis induced by the angiogenesis inhibitor PTK787, resulting in a decrease of microvessel density and a slight increase in tumor vessel size. CONCLUSIONS: We present an in vivo imaging approach that allows us to monitor both tumor microvessel density and tumor vessel size in the tumor. Moreover, this approach enables us to assess, early-on, treatment effects on tumor microvessel density as well as on tumor vessel size. Thus, this imaging-based strategy of validating anti-angiogenic treatment effects has high potential in applications to preclinical and clinical trials.
- Published
- 2011
- Full Text
- View/download PDF
3. Early detection of erlotinib treatment response in NSCLC by 3'-deoxy-3'-[F]-fluoro-L-thymidine ([F]FLT) positron emission tomography (PET).
- Author
-
Roland T Ullrich, Thomas Zander, Bernd Neumaier, Mirjam Koker, Takeshi Shimamura, Yannic Waerzeggers, Christa L Borgman, Samir Tawadros, Hongfeng Li, Martin L Sos, Heiko Backes, Geoffrey I Shapiro, Jürgen Wolf, Andreas H Jacobs, Roman K Thomas, and Alexandra Winkeler
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Inhibition of the epidermal growth factor receptor (EGFR) has shown clinical success in patients with advanced non-small cell lung cancer (NSCLC). Somatic mutations of EGFR were found in lung adenocarcinoma that lead to exquisite dependency on EGFR signaling; thus patients with EGFR-mutant tumors are at high chance of response to EGFR inhibitors. However, imaging approaches affording early identification of tumor response in EGFR-dependent carcinomas have so far been lacking. METHODOLOGY/PRINCIPAL FINDINGS: We performed a systematic comparison of 3'-Deoxy-3'-[(18)F]-fluoro-L-thymidine ([(18)F]FLT) and 2-[(18)F]-fluoro-2-deoxy-D-glucose ([(18)F]FDG) positron emission tomography (PET) for their potential to identify response to EGFR inhibitors in a model of EGFR-dependent lung cancer early after treatment initiation. While erlotinib-sensitive tumors exhibited a striking and reproducible decrease in [(18)F]FLT uptake after only two days of treatment, [(18)F]FDG PET based imaging revealed no consistent reduction in tumor glucose uptake. In sensitive tumors, a decrease in [(18)F]FLT PET but not [(18)F]FDG PET uptake correlated with cell cycle arrest and induction of apoptosis. The reduction in [(18)F]FLT PET signal at day 2 translated into dramatic tumor shrinkage four days later. Furthermore, the specificity of our results is confirmed by the complete lack of [(18)F]FLT PET response of tumors expressing the T790M erlotinib resistance mutation of EGFR. CONCLUSIONS: [(18)F]FLT PET enables robust identification of erlotinib response in EGFR-dependent tumors at a very early stage. [(18)F]FLT PET imaging may represent an appropriate method for early prediction of response to EGFR TKI treatment in patients with NSCLC.
- Published
- 2008
- Full Text
- View/download PDF
4. Hepatic FTO is dispensable for the regulation of metabolism but counteracts HCC development in vivo
- Author
-
Melanie J. Mittenbühler, Katarzyna Saedler, Hendrik Nolte, Lara Kern, Jun Zhou, Shu-Bing Qian, Lydia Meder, Roland T. Ullrich, Jens C. Brüning, and F. Thomas Wunderlich
- Subjects
Hepatocellular carcinoma ,FTO ,Cul4a ,m6A ,Internal medicine ,RC31-1245 - Abstract
Objective: Single-nucleotide polymorphisms in the FTO gene encoding an m6Am and an m6A demethylase are associated with obesity. Moreover, recent studies have linked a dysregulation of m6A modifications and its machinery, including FTO, to the development of several forms of cancers. However, the functional role of hepatic FTO in metabolism and the development and progression of hepatocellular carcinoma (HCC), a proteotypic obesity-associated cancer, remains unclear. Thus, we aimed to reveal the role of hepatic FTO in metabolism and in the initiation and progression of HCC in vivo. Methods: We generated mice with hepatic FTO deficiency (FTOL−KO). The effect of hepatic FTO on metabolism was investigated by extensive metabolic phenotyping. To determine the impact of hepatic FTO on HCC development, FTOL−KO and Ctrl mice were subjected to long-term diethylnitrosamine (DEN)-induced HCC-development and the tumor initiation phase was examined via a short-term DEN protocol. Results: In long-term DEN experiments, FTOL−KO mice exhibit increased HCC burden compared to Ctrl mice. In the tumor initiation phase, Ctrl mice display a dynamic regulation of FTO upon induction of liver damage, while this response is abrogated in FTO-deficient mice. Proteomic analyses revealed that liver damage-induced increases in FTO expression reduce CUL4A protein abundance. Functionally, simultaneous knockdown of Cul4a reverses the increased hepatocyte proliferation observed upon loss of FTO. Conclusion: Collectively, our study demonstrates that hepatic FTO is dispensable for the control of energy homeostasis and glucose metabolism. However, we show a protective function of FTO in liver carcinogenesis and suggest the FTO-dependent dynamic mRNA demethylation of Cul4a in the initiation of HCC development contributes to this effect.
- Published
- 2020
- Full Text
- View/download PDF
5. Inhibition of Tumor VEGFR2 Induces Serine 897 EphA2-Dependent Tumor Cell Invasion and Metastasis in NSCLC
- Author
-
Caroline Volz, Sara Breid, Carolin Selenz, Alina Zaplatina, Kristina Golfmann, Lydia Meder, Felix Dietlein, Sven Borchmann, Sampurna Chatterjee, Maike Siobal, Jakob Schöttle, Alexandra Florin, Mirjam Koker, Marieke Nill, Luka Ozretić, Niklas Uhlenbrock, Steven Smith, Reinhard Büttner, Hui Miao, Bingcheng Wang, H. Christian Reinhardt, Daniel Rauh, Michael Hallek, Amparo Acker-Palmer, Lukas C. Heukamp, and Roland T. Ullrich
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Anti-angiogenic treatment targeting vascular endothelial growth factor (VEGF)-VEGFR2 signaling has shown limited efficacy in lung cancer patients. Here, we demonstrate that inhibition of VEGFR2 in tumor cells, expressed in ∼20% of non-squamous non-small cell lung cancer (NSCLC) patients, leads to a pro-invasive phenotype. Drug-induced inhibition of tumor VEGFR2 interferes with the formation of the EphA2/VEGFR2 heterocomplex, thereby allowing RSK to interact with Serine 897 of EphA2. Inhibition of RSK decreases phosphorylation of Serine 897 EphA2. Selective genetic modeling of Serine 897 of EphA2 or inhibition of EphA2 abrogates the formation of metastases in vivo upon VEGFR2 inhibition. In summary, these findings demonstrate that VEGFR2-targeted therapy conditions VEGFR2-positive NSCLC to Serine 897 EphA2-dependent aggressive tumor growth and metastasis. These data shed light on the molecular mechanisms explaining the limited efficacy of VEGFR2-targeted anti-angiogenic treatment in lung cancer patients. : Anti-angiogenic treatment targeting VEGFR2 signaling has shown limited efficacy in lung cancer patients. Volz et al. show that VEGFR2 inhibition leads to a pro-invasive phenotype in VEGFR2-positive non-small cell lung cancer cells, which is mediated by phosphorylation of EphA2-S897. Keywords: VEGFR2 inhibition, tumor cell invasion, S897 EphA2, RSK, NSCLC
- Published
- 2020
- Full Text
- View/download PDF
6. Tripartite antigen-agnostic combination immunotherapy cures established poorly immunogenic tumors
- Author
-
Sven Borchmann, Carolin Selenz, Mia Lohmann, Hanna Ludwig, Asmae Gassa, Johannes Brägelmann, Philipp Lohneis, Lydia Meder, Julia Mattlener, Sara Breid, Marieke Nill, Jana Fassunke, Amy J. Wisdom, Anik Compes, Birgit Gathof, Hakan Alakus, David Kirsch, Khosro Hekmat, Reinhard Büttner, H. Christian Reinhardt, Michael Hallek, and Roland T. Ullrich
- Subjects
Pharmacology ,Cancer Research ,Immunology ,Medizin ,Combined Modality Therapy ,Toll-Like Receptor 3 ,Epitopes ,Mice ,Oncology ,Neoplasms ,Animals ,Molecular Medicine ,Immunology and Allergy ,Immunotherapy - Abstract
BackgroundSingle-agent immunotherapy has shown remarkable efficacy in selected cancer entities and individual patients. However, most patients fail to respond. This is likely due to diverse immunosuppressive mechanisms acting in a concerted way to suppress the host anti-tumor immune response. Combination immunotherapy approaches that are effective in such poorly immunogenic tumors mostly rely on precise knowledge of antigenic determinants on tumor cells. Creating an antigen-agnostic combination immunotherapy that is effective in poorly immunogenic tumors for which an antigenic determinant is not known is a major challenge.MethodsWe use multiple cell line and poorly immunogenic syngeneic, autochthonous, and autologous mouse models to evaluate the efficacy of a novel combination immunotherapy named tripartite immunotherapy (TRI-IT). To elucidate TRI-ITs mechanism of action we use immune cell depletions and comprehensive tumor and immune infiltrate characterization by flow cytometry, RNA sequencing and diverse functional assays.ResultsWe show that combined adoptive cellular therapy (ACT) with lymphokine-activated killer cells, cytokine-induced killer cells, Vγ9Vδ2-T-cells (γδ-T-cells) and T-cells enriched for tumor recognition (CTLs) display synergistic antitumor effects, which are further enhanced by cotreatment with anti-PD1 antibodies. Most strikingly, the full TRI-IT protocol, a combination of this ACT with anti-PD1 antibodies, local immunotherapy of agonists against toll-like receptor 3, 7 and 9 and pre-ACT lymphodepletion, eradicates and induces durable anti-tumor immunity in a variety of poorly immunogenic syngeneic, autochthonous, as well as autologous humanized patient-derived models. Mechanistically, we show that TRI-IT coactivates adaptive cellular and humoral, as well as innate antitumor immune responses to mediate its antitumor effect without inducing off-target toxicity.ConclusionsOverall, TRI-IT is a novel, highly effective, antigen-agnostic, non-toxic combination immunotherapy. In this study, comprehensive insights into its preclinical efficacy, even in poorly immunogenic tumors, and mode of action are given, so that translation into clinical trials is the next step.
- Published
- 2022
7. In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin's lymphoma and facilitates ultrasensitive residual disease detection
- Author
-
Peter Borchmann, Stefanie Kreissl, Esther E.E. Drees, Roland T. Ullrich, Sarah Reinke, Elena Gerhard-Hartmann, Anton Hagenbeek, Sophia Sobesky, Josée M. Zijlstra, Andreas Rosenwald, Bastian von Tresckow, Wolfram Klapper, D.M. Pegtel, Melita Cirillo, Jonathan Weiss, Janine Altmüller, Paul J Bröckelmann, Margaretha G.M. Roemer, Laman Mammadova, Stephanie Sasse, Helge Dörr, Julia Mattlener, Andreas Engert, Sven Borchmann, Zhiyuan Shi, Peter Nürnberg, Clinical Haematology, Pathology, CCA - Cancer biology and immunology, CCA - Imaging and biomarkers, and Hematology
- Subjects
Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,DNA Copy Number Variations ,Medizin ,Cancer ,Genomics ,General Medicine ,Sequence Analysis, DNA ,Biology ,Hodgkin's lymphoma ,medicine.disease ,Minimal residual disease ,Hodgkin Disease ,DNA sequencing ,Lymphoma ,Cell-free fetal DNA ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Copy-number variation ,Neoplasm Recurrence, Local ,Cell-Free Nucleic Acids - Abstract
Summary Background Individualization of treatment in Hodgkin’s lymphoma is necessary to improve cure rates and reduce treatment side effects. Currently, it is hindered by a lack of genomic characterization and sensitive molecular response assessment. Sequencing of cell-free DNA is a powerful strategy to understand the cancer genome and can be used for extremely sensitive disease monitoring. In Hodgkin’s lymphoma, a high proportion of cell-free DNA is tumor-derived, whereas traditional tumor biopsies only contain a little tumor-derived DNA. Methods We comprehensively genotype and assess minimal residual disease in 121 patients with baseline plasma as well as 77 follow-up samples from a subset of patients with our targeted cell-free DNA sequencing platform. Findings We present an integrated landscape of mutations and copy number variations in Hodgkin’s lymphoma. In addition, we perform a deep analysis of mutational processes driving Hodgkin’s lymphoma, investigate the clonal structure of Hodgkin’s lymphoma, and link several genotypes to Hodgkin’s lymphoma phenotypes and outcome. Finally, we show that minimal residual disease assessment by repeat cell-free DNA sequencing, as early as a week after treatment initiation, predicts treatment response and progression-free survival, allowing highly improved treatment guidance and relapse prediction. Conclusions Our targeted cell-free DNA sequencing platform reveals the genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive detection of minimal residual disease. Funding Mildred Scheel School of Oncology Aachen-Bonn-Cologne-Dusseldorf MD Research Stipend, Next Generation Sequencing Competence Network grant 423957469, Deutsche Krebshilfe grant 70112502, Deutsche Forschungsgemeinschaft (DFG) grant EN 179/13-1, the HL MRD consortium, and the Frau-Weiskam und Christel Ruranski-Stiftung.
- Published
- 2021
- Full Text
- View/download PDF
8. MAPK-pathway inhibition mediates inflammatory reprogramming and sensitizes tumors to targeted activation of innate immunity sensor RIG-I
- Author
-
Johannes Brägelmann, Joshua D’Rozario, Thomas Zillinger, Atsuko Hirabae, Marcel A. Dammert, Jens T. Siveke, Martin L. Sos, Reinhard Büttner, Mareike Haarmann, Julie George, Alexander Quaas, Marcel Schmiel, Kadoaki Ohashi, David F. Ast, Dennis Plenker, Jianing Gu, Julia Wegner, Kazuya Nishii, Sven Borchmann, Sachi Okawa, Martin Schlee, Sebastian Klein, Takamasa Nakasuka, Marija Trajkovic-Arsic, Roman K. Thomas, Lydia Meder, Henry Li, Philipp Lohneis, Emily S. Thomas, Carina Lorenz, Silvia von Karstedt, Roland T. Ullrich, Clemens A. Schmitt, Stefanie Lennartz, Laura Godfrey, Alena Heimsoeth, Jan Forster, Jenny Ostendorp, and Gunther Hartmann
- Subjects
MAPK/ERK pathway ,Cancer Research ,medicine.medical_treatment ,Medizin ,General Physics and Astronomy ,Cancer immunotherapy ,Mice ,Interferon ,Neoplasms ,Receptors, Immunologic ,Multidisciplinary ,Cell Death ,Kinase ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Cytokines ,DEAD Box Protein 58 ,Female ,medicine.drug ,Signal Transduction ,MAP Kinase Signaling System ,Science ,General Biochemistry, Genetics and Molecular Biology ,Article ,Targeted therapies ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Adaptor Proteins, Signal Transducing ,Immune Evasion ,Inflammation ,Innate immune system ,Oncogene ,business.industry ,Cancer ,General Chemistry ,Cell Cycle Checkpoints ,Oncogenes ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Immunity, Innate ,Mice, Inbred C57BL ,Cancer cell ,Cancer research ,business ,Interferon Regulatory Factor-1 - Abstract
Kinase inhibitors suppress the growth of oncogene driven cancer but also enforce the selection of treatment resistant cells that are thought to promote tumor relapse in patients. Here, we report transcriptomic and functional genomics analyses of cells and tumors within their microenvironment across different genotypes that persist during kinase inhibitor treatment. We uncover a conserved, MAPK/IRF1-mediated inflammatory response in tumors that undergo stemness- and senescence-associated reprogramming. In these tumor cells, activation of the innate immunity sensor RIG-I via its agonist IVT4, triggers an interferon and a pro-apoptotic response that synergize with concomitant kinase inhibition. In humanized lung cancer xenografts and a syngeneic Egfr-driven lung cancer model these effects translate into reduction of exhausted CD8+ T cells and robust tumor shrinkage. Overall, the mechanistic understanding of MAPK/IRF1-mediated intratumoral reprogramming may ultimately prolong the efficacy of targeted drugs in genetically defined cancer patients., Kinase inhibitors are widely used to treat cancer, however patients frequently develop resistance. Here, the authors investigate adaption mechanisms during drug persistence and show that stimulation of the innate immunity sensor RIG-I enhances cancer cell death when combined with kinase inhibition.
- Published
- 2021
9. Hepatic FTO is dispensable for the regulation of metabolism but counteracts HCC development in vivo
- Author
-
Jun Zhou, Melanie J. Mittenbühler, Shu-Bing Qian, Hendrik Nolte, Lydia Meder, Roland T. Ullrich, Katarzyna Saedler, F. Thomas Wunderlich, Jens C. Brüning, and Lara Kern
- Subjects
0301 basic medicine ,Male ,Proteomics ,lcsh:Internal medicine ,Carcinoma, Hepatocellular ,endocrine system diseases ,Hepatocellular carcinoma ,Alpha-Ketoglutarate-Dependent Dioxygenase FTO ,030209 endocrinology & metabolism ,Tumor initiation ,Carbohydrate metabolism ,Cul4a ,Brief Communication ,FTO gene ,Polymorphism, Single Nucleotide ,Energy homeostasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Homeostasis ,lcsh:RC31-1245 ,Molecular Biology ,Cell Proliferation ,Mice, Knockout ,Gene knockdown ,biology ,Liver Neoplasms ,nutritional and metabolic diseases ,Cell Biology ,pathological conditions, signs and symptoms ,m6A ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Glucose ,Liver ,Hepatocyte ,Cancer research ,biology.protein ,Demethylase ,CUL4A ,FTO ,Energy Metabolism ,Signal Transduction - Abstract
Objective Single-nucleotide polymorphisms in the FTO gene encoding an m6Am and an m6A demethylase are associated with obesity. Moreover, recent studies have linked a dysregulation of m6A modifications and its machinery, including FTO, to the development of several forms of cancers. However, the functional role of hepatic FTO in metabolism and the development and progression of hepatocellular carcinoma (HCC), a proteotypic obesity-associated cancer, remains unclear. Thus, we aimed to reveal the role of hepatic FTO in metabolism and in the initiation and progression of HCC in vivo. Methods We generated mice with hepatic FTO deficiency (FTOL−KO). The effect of hepatic FTO on metabolism was investigated by extensive metabolic phenotyping. To determine the impact of hepatic FTO on HCC development, FTOL−KO and Ctrl mice were subjected to long-term diethylnitrosamine (DEN)-induced HCC-development and the tumor initiation phase was examined via a short-term DEN protocol. Results In long-term DEN experiments, FTOL−KO mice exhibit increased HCC burden compared to Ctrl mice. In the tumor initiation phase, Ctrl mice display a dynamic regulation of FTO upon induction of liver damage, while this response is abrogated in FTO-deficient mice. Proteomic analyses revealed that liver damage-induced increases in FTO expression reduce CUL4A protein abundance. Functionally, simultaneous knockdown of Cul4a reverses the increased hepatocyte proliferation observed upon loss of FTO. Conclusion Collectively, our study demonstrates that hepatic FTO is dispensable for the control of energy homeostasis and glucose metabolism. However, we show a protective function of FTO in liver carcinogenesis and suggest the FTO-dependent dynamic mRNA demethylation of Cul4a in the initiation of HCC development contributes to this effect., Graphical abstract Image 1, Highlights • Hepatic FTO is dispensable for whole body metabolism. • FTO is dynamically regulated upon acute liver damage and controls proliferation. • Hepatic FTO function protects against the development of hepatocellular carcinoma (HCC). • Cul4a is a downstream target of FTO, and Cul4a knockdown reduces damage-induced proliferation in FTOL−KO livers.
- Published
- 2020
10. Imaging of Non— or Very Subtle Contrast-Enhancing Malignant Gliomas with [C]-Methionine Positron Emission Tomography
- Author
-
Norbert Galldiks, Lutz W. Kracht, Veronika Dunkl, Roland T. Ullrich, Stefan Vollmar, Andreas H. Jacobs, Gereon R. Fink, and Michael Schroeter
- Subjects
Biology (General) ,QH301-705.5 ,Medical technology ,R855-855.5 - Abstract
In patients with World Health Organization (WHO) grade III glioma with a lack of or minimal (< 1 cm 3 ) magnetic resonance imaging (MRI) contrast enhancement, the volume of the metabolically active part of the tumor was assessed by [ 11 C]-methionine positron emission tomography (MET-PET). Eleven patients with WHO grade III gliomas underwent MET-PET and MRI (contrast-enhanced T 1 -and T 2 -weighted images). To calculate the volumes in cubic centimeters, threshold-based volume of interest analyses of the metabolically active tumor (MET uptake index ≥ 1.3), contrast enhancement, and the T 2 lesion were performed after coregistration of all images. In all patients, the metabolically active tumor volume was larger than the volume of gadolinium–diethylenetriamine pentaacetic acid (Gd-DTPA) enhancement (20.8 ± 18.8 vs 0.29 ± 0.25 cm 3 ; p < .001). With the exception of one patient, the volumes of contrast enhancement were located within the metabolically active tumor volume. In contrast, in the majority of patients, MET uptake overlapped with the T2 lesion and reached beyond it (in 10 of 12 MRIs/MET-PET scans). The present data suggest that in patients with WHO grade III glioma with minimal or a lack of contrast enhancement, MET-PET delineates metabolically active tumor tissue. These findings support the use of combined PET-MRI with radiolabeled amino acids (eg, MET) for the delineating of the true extent of active tumor in the diagnosis and treatment planning of patients with gliomas.
- Published
- 2011
- Full Text
- View/download PDF
11. Patient-Tailored, Imaging-Guided, Long-Term Temozolomide Chemotherapy in Patients with Glioblastoma
- Author
-
Norbert Galldiks, Lutz W. Kracht, Lothar Burghaus, Roland T. Ullrich, Heiko Backes, Anna Brunn, Wolf-Dieter Heiss, and Andreas H. Jacobs
- Subjects
Biology (General) ,QH301-705.5 ,Medical technology ,R855-855.5 - Abstract
We present two patients with glioblastoma with an unusually stable clinical course and long-term survival who were treated after surgery and radiotherapy with adjuvant temozolomide (TMZ) chemotherapy for 17 and 20 cycles, respectively. Afterward, adjuvant TMZ chemotherapy was discontinued in one patient and the dosage of TMZ was reduced in the other. In addition to clinical status and magnetic resonance imaging, the biologic activity of the tumors was monitored by repeated methyl- 11 C-l-methionine (MET) and 3′-deoxy-3′- 18 F-fluorothymidine (FLT) positron emission tomography (PET) studies in these patients. In these patients, repeated MET-and FLT-PET imaging documented complete response to the initial treatment regimen, including resection, radiation, and TMZ, and during the course of the disease, recurrent, uncontrollable tumor activity. Continuation or dose escalation of TMZ in both patients was shown to be ineffective to overcome the metabolic activity of the tumor. Our data suggest that repeated MET- and FLT-PET imaging provide information on the biologic activity of a tumor that is highly useful to monitor and detect changes in activity.
- Published
- 2010
- Full Text
- View/download PDF
12. Combined VEGF and PD-L1 Blockade Displays Synergistic Treatment Effects in an Autochthonous Mouse Model of Small Cell Lung Cancer
- Author
-
Sebastian Oberbeck, Juergen Wolf, Reinhard Buettner, H. Christian Reinhardt, Michael von Bergwelt-Baildon, Hans A. Schlößer, Alexandra Florin, Sven Borchmann, Michael Hallek, Margarete Odenthal, Ignacija Vlasic, Roland T. Ullrich, R. Riedel, Marco Herling, Kerstin Wennhold, Felix Dietlein, Anna Schmitt, Martin Thelen, Philipp Schuldt, Kristina Golfmann, Sebastian Klein, and Lydia Meder
- Subjects
0301 basic medicine ,Male ,Vascular Endothelial Growth Factor A ,Cancer Research ,Lung Neoplasms ,T-Lymphocytes ,VEGF ,PD-L1 ,autochthonous mouse model ,SCLC ,Antineoplastic Agents ,Peripheral blood mononuclear cell ,B7-H1 Antigen ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,Medicine ,Animals ,Receptor ,biology ,business.industry ,Cancer ,medicine.disease ,Phenotype ,Small Cell Lung Carcinoma ,Blockade ,Mice, Inbred C57BL ,Vascular endothelial growth factor A ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,business - Abstract
Small cell lung cancer (SCLC) represents the most aggressive pulmonary neoplasm and is often diagnosed at late stage with limited survival, despite combined chemotherapies. We show in an autochthonous mouse model of SCLC that combined anti-VEGF/anti-PD-L1–targeted therapy synergistically improves treatment outcome compared with anti–PD-L1 and anti-VEGF monotherapy. Mice treated with anti–PD-L1 alone relapsed after 3 weeks and were associated with a tumor-associated PD-1/TIM-3 double-positive exhausted T-cell phenotype. This exhausted T-cell phenotype upon PD-L1 blockade was abrogated by the addition of anti-VEGF–targeted treatment. We confirmed a similar TIM-3–positive T-cell phenotype in peripheral blood mononuclear cells of patients with SCLC with adaptive resistance to anti–PD-1 treatment. Mechanistically, we show that VEGFA enhances coexpression of the inhibitory receptor TIM-3 on T cells, indicating an immunosuppressive function of VEGF in patients with SCLC during anti–PD-1-targeted treatment. Our data strongly suggest that a combination of anti-VEGF and anti–PD-L1 therapies can be an effective treatment strategy in patients with SCLC. Significance: Combining VEGF and PD-L1 blockade could be of therapeutic benefit to patients with small cell lung cancer. Cancer Res; 78(15); 4270–81. ©2018 AACR.
- Published
- 2018
13. CD74-NRG1 Fusions in Lung Adenocarcinoma
- Author
-
Dirk Brehmer, Dennis Plenker, Benjamin Solomon, Stefan A. Haas, Mirjam Koker, Denis Moro-Sibilot, Zoe Wainer, Janine Altmüller, Hélène Nagy-Mignotte, Yasushi Yatabe, Gavin M. Wright, Prudence A. Russell, Thomas Zander, Roopika Menon, Annamaria la Torre, Timothy Perera, Sandra Ortiz-Cuaran, Marc Parade, Marc Bos, Elisabeth Brambilla, Erich Stoelben, Ruping Sun, Christian Becker, Vito Michele Fazio, Martin Vingron, Roman K. Thomas, Wenzel Vogel, Jakob Schöttle, Peter Nürnberg, Sylvie Lantuejoul, Idoya Lahortiga, Iver Petersen, Hirotaka Osada, Jürgen Wolf, Roland T. Ullrich, Souichi Ogata, Lucia Anna Muscarella, Jörg Sänger, Lukas C. Heukamp, Joachim H. Clement, Christian Brambilla, Johannes M. Heuckmann, Lynnette Fernandez-Cuesta, Sascha Ansén, Sven Perner, Reinhard Buettner, Martin Peifer, Frauke Leenders, Juliane Daßler, Ilona Dahmen, and Florian Malchers
- Subjects
Adult ,Male ,Lung Neoplasms ,Oncogene Proteins, Fusion ,Neuregulin-1 ,Molecular Sequence Data ,Adenocarcinoma of Lung ,Biology ,Adenocarcinoma ,Transcriptome ,Fusion gene ,Mice ,Cell Line, Tumor ,mental disorders ,medicine ,ROS1 ,Animals ,Humans ,ERBB3 ,Aged ,Aged, 80 and over ,Base Sequence ,Gene Expression Profiling ,Histocompatibility Antigens Class II ,Cancer ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Adenocarcinoma, Mucinous ,3. Good health ,Gene expression profiling ,Antigens, Differentiation, B-Lymphocyte ,Oncology ,Immunology ,Cancer research ,NIH 3T3 Cells ,Ectopic expression ,Female ,Signal Transduction - Abstract
We discovered a novel somatic gene fusion, CD74–NRG1, by transcriptome sequencing of 25 lung adenocarcinomas of never smokers. By screening 102 lung adenocarcinomas negative for known oncogenic alterations, we found four additional fusion-positive tumors, all of which were of the invasive mucinous subtype. Mechanistically, CD74–NRG1 leads to extracellular expression of the EGF-like domain of NRG1 III-β3, thereby providing the ligand for ERBB2–ERBB3 receptor complexes. Accordingly, ERBB2 and ERBB3 expression was high in the index case, and expression of phospho-ERBB3 was specifically found in tumors bearing the fusion (P < 0.0001). Ectopic expression of CD74–NRG1 in lung cancer cell lines expressing ERBB2 and ERBB3 activated ERBB3 and the PI3K–AKT pathway, and led to increased colony formation in soft agar. Thus, CD74–NRG1 gene fusions are activating genomic alterations in invasive mucinous adenocarcinomas and may offer a therapeutic opportunity for a lung tumor subtype with, so far, no effective treatment. Significance: CD74–NRG1 fusions may represent a therapeutic opportunity for invasive mucinous lung adenocarcinomas, a tumor with no effective treatment that frequently presents with multifocal unresectable disease. Cancer Discov; 4(4); 415–22. ©2014 AACR. This article is highlighted in the In This Issue feature, p. 377
- Published
- 2014
- Full Text
- View/download PDF
14. Imaging biological activity of a glioblastoma treated with an individual patient-tailored, experimental therapy regimen
- Author
-
Lutz W. Kracht, Gereon R. Fink, Michael Schroeter, Norbert Galldiks, and Roland T. Ullrich
- Subjects
Oncology ,Experimental glioma therapy ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,l-[methyl-(11)C]-methionine PET ,medicine.drug_class ,Clinical Neurology ,Tyrosine kinase inhibitor ,Case Report ,Tyrosine-kinase inhibitor ,Neurosurgical Procedures ,Piperazines ,Maintenance therapy ,Internal medicine ,Glioma ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Combined Modality Therapy ,Humans ,Hydroxyurea ,medicine.diagnostic_test ,business.industry ,Brain Neoplasms ,Therapeutic effect ,Imatinib ,medicine.disease ,Surgery ,Regimen ,Pyrimidines ,Neurology ,Positron emission tomography ,Positron-Emission Tomography ,Benzamides ,Imatinib Mesylate ,Neurology (clinical) ,Cranial Irradiation ,business ,Glioblastoma ,Platelet-derived growth factor (PDGF) receptor ,medicine.drug - Abstract
Purpose To monitor the metabolic effects of an individual patient-tailored, experimental glioma therapy regimen that included repetitive multiple neurosurgical resections, radiosurgical interventions, and an adjuvant maintenance therapy based on the tyrosine kinase inhibitor imatinib in combination with the chemotherapeutic agent hydroxyurea (HU). Procedures Therapeutic effects were monitored in a 26-year-old male patient with a glioblastoma multiforme by multimodal imaging using sequential l-[methyl-(11)C]-methionine positron emission tomography (MET–PET) and MRI. The normalized MET uptake and volume of the metabolically active tumor were assessed sequentially. Results The individual patient-tailored, experimental glioma therapy caused a continuous decline of metabolically active tumor volume, associated with clinical remission over a period of more than two years. Conclusions MET–PET seems to be useful for monitoring patient-tailored, experimental glioma therapy regimens, especially when patients are treated with a multi-step therapeutic regimen. Monitoring and guidance of those experimental therapy regimens by MET–PET in a larger patient group are needed to confirm its clinical value.
- Published
- 2009
15. A framework for identification of actionable cancer genome dependencies in small cell lung cancer
- Author
-
Christian Müller, Martin L. Sos, Felix Dietlein, Andrey P. Antonchick, Viktor V. Vintonyak, Stefanie Heynck, André Richters, Daniel Rauh, Jakob Schöttle, Reinhard Büttner, Peter M. Schneider, Johannes M. Heuckmann, Martin Peifer, Roland T. Ullrich, Danila Seidel, Florian Malchers, Roman K. Thomas, Takashi Ninomiya, Herbert Waldmann, Mirjam Koker, Hyatt Balke-Want, Lukas C. Heukamp, and Patrick A. Eyers
- Subjects
Cell Survival ,Immunoblotting ,Aurora B kinase ,Apoptosis ,Biology ,Diamines ,Protein Serine-Threonine Kinases ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,Gefitinib ,Aurora kinase ,Growth factor receptor ,Aurora Kinases ,Cell Line, Tumor ,medicine ,Aurora Kinase B ,Humans ,Benzothiazoles ,Kinase activity ,Enzyme Inhibitors ,Organic Chemicals ,neoplasms ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,DNA Primers ,Phosphoinositide-3 Kinase Inhibitors ,0303 health sciences ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,Biological Sciences ,Flow Cytometry ,Small Cell Lung Carcinoma ,humanities ,3. Good health ,respiratory tract diseases ,G2 Phase Cell Cycle Checkpoints ,030220 oncology & carcinogenesis ,Cancer research ,Quinolines ,medicine.drug ,Signal Transduction - Abstract
Small cell lung cancer (SCLC) accounts for about 15% of all lung cancers. The prognosis of SCLC patients is devastating and no biologically targeted therapeutics are active in this tumor type. To develop a framework for development of specific SCLC-targeted drugs we conducted a combined genomic and pharmacological vulnerability screen in SCLC cell lines. We show that SCLC cell lines capture the genomic landscape of primary SCLC tumors and provide genetic predictors for activity of clinically relevant inhibitors by screening 267 compounds across 44 of these cell lines. We show Aurora kinase inhibitors are effective in SCLC cell lines bearing MYC amplification, which occur in 3–7% of SCLC patients. In MYC -amplified SCLC cells Aurora kinase inhibition associates with G2/M-arrest, inactivation of PI3-kinase (PI3K) signaling, and induction of apoptosis. Aurora dependency in SCLC primarily involved Aurora B, required its kinase activity, and was independent of depletion of cytoplasmic levels of MYC. Our study suggests that a fraction of SCLC patients may benefit from therapeutic inhibition of Aurora B. Thus, thorough chemical and genomic exploration of SCLC cell lines may provide starting points for further development of rational targeted therapeutic intervention in this deadly tumor type.
- Published
- 2012
16. Patient-Tailored, Imaging-Guided, Long-Term Temozolomide Chemotherapy in Patients with Glioblastoma
- Author
-
Heiko Backes, Roland T. Ullrich, Andreas H. Jacobs, Anna Brunn, Wolf-Dieter Heiss, Norbert Galldiks, Lutz W. Kracht, and Lothar Burghaus
- Subjects
Oncology ,Adult ,medicine.medical_specialty ,Fluorine Radioisotopes ,lcsh:Medical technology ,medicine.medical_treatment ,Dacarbazine ,Biomedical Engineering ,Methionine ,Internal medicine ,medicine ,Temozolomide ,Humans ,Radiology, Nuclear Medicine and imaging ,Carbon Radioisotopes ,Radionuclide Imaging ,Antineoplastic Agents, Alkylating ,lcsh:QH301-705.5 ,Chemotherapy ,medicine.diagnostic_test ,business.industry ,Brain Neoplasms ,Magnetic resonance imaging ,Middle Aged ,Condensed Matter Physics ,Dideoxynucleosides ,Surgery ,Radiation therapy ,Regimen ,lcsh:Biology (General) ,lcsh:R855-855.5 ,Positron emission tomography ,Molecular Medicine ,Radiopharmaceuticals ,business ,Glioblastoma ,Adjuvant ,Biotechnology ,medicine.drug - Abstract
We present two patients with glioblastoma with an unusually stable clinical course and long-term survival who were treated after surgery and radiotherapy with adjuvant temozolomide (TMZ) chemotherapy for 17 and 20 cycles, respectively. Afterward, adjuvant TMZ chemotherapy was discontinued in one patient and the dosage of TMZ was reduced in the other. In addition to clinical status and magnetic resonance imaging, the biologic activity of the tumors was monitored by repeated methyl- 11 C-l-methionine (MET) and 3′-deoxy-3′- 18 F-fluorothymidine (FLT) positron emission tomography (PET) studies in these patients. In these patients, repeated MET-and FLT-PET imaging documented complete response to the initial treatment regimen, including resection, radiation, and TMZ, and during the course of the disease, recurrent, uncontrollable tumor activity. Continuation or dose escalation of TMZ in both patients was shown to be ineffective to overcome the metabolic activity of the tumor. Our data suggest that repeated MET- and FLT-PET imaging provide information on the biologic activity of a tumor that is highly useful to monitor and detect changes in activity.
- Published
- 2010
17. Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer
- Author
-
Samanthi A. Perera, Aviva Goel, Matthäus Getlik, Daniel Rauh, Lukas C. Heukamp, Martin L. Sos, Lucia Regales, Klaus P. Hoeflich, Katerina Politi, William Pao, Hamid Kashkar, Sascha Ansén, Lori Friedman, Christine Orr, Martin Peifer, Roman K. Thomas, Johannes M. Heuckmann, William R. Sellers, Kevan M. Shokat, Florian Fischer, Stefanie Fischer, Isabel Stückrath, Mirjam Koker, Jonathan Weiss, Kwok-Kin Wong, Rameen Beroukhim, Thomas Zander, Roland T. Ullrich, Stefanie Heynck, and Kathrin Michel
- Subjects
MAPK/ERK pathway ,Multidisciplinary ,biology ,Genotype ,MAP Kinase Signaling System ,Cancer ,Apoptosis ,Biological Sciences ,medicine.disease ,Receptor tyrosine kinase ,Cell biology ,Phosphatidylinositol 3-Kinases ,In vivo ,Cell culture ,Cell Line, Tumor ,Neoplasms ,biology.protein ,medicine ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors - Abstract
In cancer, genetically activated proto-oncogenes often induce “ upstream ” dependency on the activity of the mutant oncoprotein. Therapeutic inhibition of these activated oncoproteins can induce massive apoptosis of tumor cells, leading to sometimes dramatic tumor regressions in patients. The PI3K and MAPK signaling pathways are central regulators of oncogenic transformation and tumor maintenance. We hypothesized that upstream dependency engages either one of these pathways preferentially to induce “ downstream ” dependency. Therefore, we analyzed whether downstream pathway dependency segregates by genetic aberrations upstream in lung cancer cell lines. Here, we show by systematically linking drug response to genomic aberrations in non-small-cell lung cancer, as well as in cell lines of other tumor types and in a series of in vivo cancer models, that tumors with genetically activated receptor tyrosine kinases depend on PI3K signaling, whereas tumors with mutations in the RAS/RAF axis depend on MAPK signaling. However, efficacy of downstream pathway inhibition was limited by release of negative feedback loops on the reciprocal pathway. By contrast, combined blockade of both pathways was able to overcome the reciprocal pathway activation induced by inhibitor-mediated release of negative feedback loops and resulted in a significant increase in apoptosis and tumor shrinkage. Thus, by using a systematic chemo-genomics approach, we identify genetic lesions connected to PI3K and MAPK pathway activation and provide a rationale for combined inhibition of both pathways. Our findings may have implications for patient stratification in clinical trials.
- Published
- 2009
18. RETRACTED: Cytohesins Are Cytoplasmic ErbB Receptor Activators
- Author
-
Michael Famulok, Sampurna Chatterjee, Barbara Albertoni, Peter J. Verveer, Anke Bill, Sebastian Zimmer, Franziska Thorwirth, Roman K. Thomas, Lukas C. Heukamp, David Walrafen, Jin-Na Song, Roland T. Ullrich, Anton Schmitz, Lisa Meffert, Arne Schreiber, and Thorsten Lang
- Subjects
biology ,Kinase ,Biochemistry, Genetics and Molecular Biology(all) ,Autophosphorylation ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,ErbB Receptors ,ErbB ,Cytoplasm ,biology.protein ,Epidermal growth factor receptor ,Receptor ,Intracellular - Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy).This article has been retracted at the request of the authors.In Bill et al., we identified the cytohesin ARNO as an ErbB receptor activator that enhances receptor autophosphorylation in H460 and SkBr3 cells. Cell-free reconstitution with purified proteins showed that ARNO enhances EGF receptor autophosphorylation by direct interaction with the intracellular domain of the receptor (EGFR-ICD). We were recently notified by Cell that the laboratory of Dr. Oreste Segatto was unable to replicate these effects in HeLa cells or under cell-free reconstituted conditions. Although we reproducibly observe 1.6-fold ARNO-dependent stimulation of cellular EGFR autophosphorylation and partial colocalization of ARNO with EGFR in plasma membrane sheets of HeLa cells, we cannot reproduce the data showing direct stimulation of EGFR-ICD autophosphorylation by ARNO-Sec7 in a cell-free reconstitution system (shown in Figures 5D and S5D). Thus, the conclusion that ARNO enhances receptor activation by direct interaction with EGFR-ICD is no longer supported by experimental evidence. We maintain the view that cellular EGFR autophosphorylation is enhanced by ARNO, that ARNO and EGFR colocalize at the plasma membrane, and that ARNO binds to the EGFR-ICD. Nevertheless, our inability to reproduce the data in Figures 5D and S5D compromises our confidence in the proposed mechanism of this regulation. Therefore, we wish to retract the paper. We apologize for any inconvenience that may have resulted from its publication. Franziska Thorwith could not be reached about this retraction.
- Full Text
- View/download PDF
19. Somatic rearrangements causing oncogenic ectodomain deletions of FGFR1 in squamous cell lung cancer
- Author
-
Florian Malchers, Lucia Nogova, Martijn H.A. van Attekum, Lukas Maas, Johannes Brägelmann, Christoph Bartenhagen, Luc Girard, Graziella Bosco, Ilona Dahmen, Sebastian Michels, Clare E. Weeden, Andreas H. Scheel, Lydia Meder, Kristina Golfmann, Philipp Schuldt, Janna Siemanowski, Jan Rehker, Sabine Merkelbach-Bruse, Roopika Menon, Oliver Gautschi, Johannes M. Heuckmann, Elisabeth Brambilla, Marie-Liesse Asselin-Labat, Thorsten Persigehl, John D. Minna, Henning Walczak, Roland T. Ullrich, Matthias Fischer, Hans Christian Reinhardt, Jürgen Wolf, Reinhard Büttner, Martin Peifer, Julie George, and Roman K. Thomas
- Subjects
Genetics ,Oncology ,Medicine - Abstract
The discovery of frequent 8p11-p12 amplifications in squamous cell lung cancer (SQLC) has fueled hopes that FGFR1, located inside this amplicon, might be a therapeutic target. In a clinical trial, only 11% of patients with 8p11 amplification (detected by FISH) responded to FGFR kinase inhibitor treatment. To understand the mechanism of FGFR1 dependency, we performed deep genomic characterization of 52 SQLCs with 8p11-p12 amplification, including 10 tumors obtained from patients who had been treated with FGFR inhibitors. We discovered somatically altered variants of FGFR1 with deletion of exons 1–8 that resulted from intragenic tail-to-tail rearrangements. These ectodomain-deficient FGFR1 variants (ΔEC-FGFR1) were expressed in the affected tumors and were tumorigenic in both in vitro and in vivo models of lung cancer. Mechanistically, breakage-fusion-bridges were the source of 8p11-p12 amplification, resulting from frequent head-to-head and tail-to-tail rearrangements. Generally, tail-to-tail rearrangements within or in close proximity upstream of FGFR1 were associated with FGFR1 dependency. Thus, the genomic events shaping the architecture of the 8p11-p12 amplicon provide a mechanistic explanation for the emergence of FGFR1-driven SQLC. Specifically, we believe that FGFR1 ectodomain–deficient and FGFR1-centered amplifications caused by tail-to-tail rearrangements are a novel somatic genomic event that might be predictive of therapeutically relevant FGFR1 dependency.
- Published
- 2023
- Full Text
- View/download PDF
20. Tripartite antigen-agnostic combination immunotherapy cures established poorly immunogenic tumors
- Author
-
Reinhard Büttner, Khosro Hekmat, David Kirsch, Michael Hallek, Birgit Gathof, Jana Fassunke, Asmae Gassa, Hakan Alakus, Sven Borchmann, Carolin Selenz, Mia Lohmann, Hanna Ludwig, Johannes Brägelmann, Philipp Lohneis, Lydia Meder, Julia Mattlener, Sara Breid, Marieke Nill, Amy J. Wisdom, Anik Compes, H. Christian Reinhardt, and Roland T. Ullrich
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Single-agent immunotherapy has shown remarkable efficacy in selected cancer entities and individual patients. However, most patients fail to respond. This is likely due to diverse immunosuppressive mechanisms acting in a concerted way to suppress the host anti-tumor immune response. Combination immunotherapy approaches that are effective in such poorly immunogenic tumors mostly rely on precise knowledge of antigenic determinants on tumor cells. Creating an antigen-agnostic combination immunotherapy that is effective in poorly immunogenic tumors for which an antigenic determinant is not known is a major challenge.Methods We use multiple cell line and poorly immunogenic syngeneic, autochthonous, and autologous mouse models to evaluate the efficacy of a novel combination immunotherapy named tripartite immunotherapy (TRI-IT). To elucidate TRI-ITs mechanism of action we use immune cell depletions and comprehensive tumor and immune infiltrate characterization by flow cytometry, RNA sequencing and diverse functional assays.Results We show that combined adoptive cellular therapy (ACT) with lymphokine-activated killer cells, cytokine-induced killer cells, Vγ9Vδ2-T-cells (γδ-T-cells) and T-cells enriched for tumor recognition (CTLs) display synergistic antitumor effects, which are further enhanced by cotreatment with anti-PD1 antibodies. Most strikingly, the full TRI-IT protocol, a combination of this ACT with anti-PD1 antibodies, local immunotherapy of agonists against toll-like receptor 3, 7 and 9 and pre-ACT lymphodepletion, eradicates and induces durable anti-tumor immunity in a variety of poorly immunogenic syngeneic, autochthonous, as well as autologous humanized patient-derived models. Mechanistically, we show that TRI-IT coactivates adaptive cellular and humoral, as well as innate antitumor immune responses to mediate its antitumor effect without inducing off-target toxicity.Conclusions Overall, TRI-IT is a novel, highly effective, antigen-agnostic, non-toxic combination immunotherapy. In this study, comprehensive insights into its preclinical efficacy, even in poorly immunogenic tumors, and mode of action are given, so that translation into clinical trials is the next step.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.