10 results on '"Tobias Gutting"'
Search Results
2. Expression of the EGFR-RAS Inhibitory Proteins DOK1 and MTMR7 and its Significance in Colorectal Adenoma and Adenoma Recurrence
- Author
-
Tobias, Gutting, Andreas, Merkel, David J, Fink, Svetlana, Hetjens, Philip, Weidner, Yanxiong, Yu, Georg, Kähler, Matthias P, Ebert, Timo, Gaiser, Elke, Burgermeister, and Sebastian, Belle
- Subjects
Adenoma ,Male ,Gastroenterology ,Colonic Polyps ,RNA-Binding Proteins ,Phosphoproteins ,Protein Tyrosine Phosphatases, Non-Receptor ,DNA-Binding Proteins ,ErbB Receptors ,ras Proteins ,Humans ,Female ,Neoplasm Recurrence, Local ,Colorectal Neoplasms - Abstract
Background and Aims: Colorectal adenomas are precursor lesions for colorectal cancer (CRC), a major cause of cancer-related death. Despite all molecular insights, there are still unknown variables in the development of CRC as well as uncertainties regarding adenoma recurrence after resection. We aimed to characterize the expression of docking protein 1 (DOK1) and myotubularin-related protein 7 (MTMR7), which share inhibiting functions on EGFR-RAS-signalling, a major oncogenic driver in CRC, and their association with clinical variables and adenoma recurrence. Methods: This observational study is based on clinical data obtained from patients who underwent routine endoscopy and consecutive follow-up examinations. Immunohistochemistry was conducted both in dysplastic tissue and adjacent non-dysplastic mucosa followed by microscopical assessment. Recurrence was differentiated between local, segmental and distant relapse. Results: A total of 56 patients (23 females) gathering 96 adenomas/polyps were included. 36 patients experienced a metachronous lesion, 23 patients had simultaneous lesions in their index endoscopy. Female patients showed lower levels of MTMR7 in adenomas (p=0.0318). Adenomas of young patients showed lower DOK1 than those of older patients (p=0.0469). Big adenomas showed a higher expression of DOK1 than small lesions (p=0.0044). In serrated lesions, DOK1 was reduced (p=0.0026) and correlated with the quantity of lesions (p < 0.001). MTMR7 was significantly reduced in distant (p=0.05) and local segmental recurrence (p=0.0362), while DOK1 showed higher expression in recurrence (p=0.0291). Conclusions: We found ambivalent results regarding the role of the markers as potential tumor suppressors, implying a context-dependent function of these molecules which might change in the course of time. DOK1 may play an inhibiting role in the serrated pathway. Remarkably, molecular markers have the potential to predict recurrence, since a combined expression analysis of high DOK1 and low MTMR7 correlated with the likelihood of segmental adenoma recurrence.
- Published
- 2021
- Full Text
- View/download PDF
3. The drug-induced phenotypic landscape of colorectal cancer organoids
- Author
-
Johannes Betge, Niklas Rindtorff, Jan Sauer, Benedikt Rauscher, Clara Dingert, Haristi Gaitantzi, Frank Herweck, Kauthar Srour-Mhanna, Thilo Miersch, Erica Valentini, Kim E. Boonekamp, Veronika Hauber, Tobias Gutting, Larissa Frank, Sebastian Belle, Timo Gaiser, Inga Buchholz, Ralf Jesenofsky, Nicolai Härtel, Tianzuo Zhan, Bernd Fischer, Katja Breitkopf-Heinlein, Elke Burgermeister, Matthias P. Ebert, and Michael Boutros
- Subjects
Organoids ,Multidisciplinary ,Phenotype ,General Physics and Astronomy ,Humans ,General Chemistry ,Colorectal Neoplasms ,General Biochemistry, Genetics and Molecular Biology ,Signal Transduction - Abstract
Patient-derived organoids resemble the biology of tissues and tumors, enabling ex vivo modeling of human diseases. They have heterogeneous morphologies with unclear biological causes and relationship to treatment response. Here, we use high-throughput, image-based profiling to quantify phenotypes of over 5 million individual colorectal cancer organoids after treatment with >500 small molecules. Integration of data using multi-omics modeling identifies axes of morphological variation across organoids: Organoid size is linked to IGF1 receptor signaling, and cystic vs. solid organoid architecture is associated with LGR5 + stemness. Treatment-induced organoid morphology reflects organoid viability, drug mechanism of action, and is biologically interpretable. Inhibition of MEK leads to cystic reorganization of organoids and increases expression ofLGR5, while inhibition of mTOR induces IGF1 receptor signaling. In conclusion, we identify shared axes of variation for colorectal cancer organoid morphology, their underlying biological mechanisms, and pharmacological interventions with the ability to move organoids along them.
- Published
- 2021
4. Detection of mutational patterns in cell‐free DNA of colorectal cancer by custom amplicon sequencing
- Author
-
Tianzuo Zhan, Nicolai Härtel, Nadine Schulte, Simon Herrmann, Benedikt Rauscher, Ralf Jesenofsky, Sebastian Belle, Matthias P. Ebert, Michael Boutros, Timo Gaiser, Tobias Gutting, Johannes Betge, and Ralf Hofheinz
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,next‐generation sequencing ,colorectal cancer ,Computational biology ,Drug resistance ,Disease ,Biology ,lcsh:RC254-282 ,DNA sequencing ,03 medical and health sciences ,0302 clinical medicine ,Multiplex polymerase chain reaction ,Genetics ,medicine ,Humans ,Liquid biopsy ,cfDNA ,Research Articles ,liquid biopsy ,High-Throughput Nucleotide Sequencing ,Reproducibility of Results ,General Medicine ,Amplicon ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Amplicon sequencing ,Molecular Medicine ,Colorectal Neoplasms ,Cell-Free Nucleic Acids ,Research Article - Abstract
Monitoring the mutational patterns of solid tumors during cancer therapy is a major challenge in oncology. Analysis of mutations in cell-free (cf) DNA offers a noninvasive approach to detect mutations that may be prognostic for disease survival or predictive for primary or secondary drug resistance. A main challenge for the application of cfDNA as a diagnostic tool is the diverse mutational landscape of cancer. Here, we developed a flexible end-to-end experimental and bioinformatic workflow to analyze mutations in cfDNA using custom amplicon sequencing. Our approach relies on open-software tools to select primers suitable for multiplex PCR using minimal cfDNA as input. In addition, we developed a robust linear model to identify specific genetic alterations from sequencing data of cfDNA. We used our workflow to design a custom amplicon panel suitable for detection of hotspot mutations relevant for colorectal cancer and analyzed mutations in serial cfDNA samples from a pilot cohort of 34 patients with advanced colorectal cancer. Using our method, we could detect recurrent and patient-specific mutational patterns in the majority of patients. Furthermore, we show that dynamic changes of mutant allele frequencies in cfDNA correlate well with disease progression. Finally, we demonstrate that sequencing of cfDNA can reveal mechanisms of resistance to anti-Epidermal Growth Factor Receptor(EGFR) antibody treatment. Thus, our approach offers a simple and highly customizable method to explore genetic alterations in cfDNA.
- Published
- 2019
5. Checkpoints and beyond – Immunotherapy in colorectal cancer
- Author
-
Tobias Gutting, Elke Burgermeister, Nicolai Härtel, and Matthias P. Ebert
- Subjects
0301 basic medicine ,Cancer Research ,Adoptive cell transfer ,Colorectal cancer ,medicine.medical_treatment ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,medicine ,Humans ,Precision Medicine ,Cytokine-induced killer cell ,business.industry ,Tumor-infiltrating lymphocytes ,Immunogenicity ,Melanoma ,Cell Cycle Checkpoints ,Dendritic Cells ,Immunotherapy ,medicine.disease ,Combined Modality Therapy ,Chimeric antigen receptor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Colorectal Neoplasms ,business - Abstract
Immunotherapy is the latest revolution in cancer therapy. It continues to show impressive results in malignancies like melanoma and others. At least so far, effects are modest in colorectal cancer (CRC) and only a subset of patients benefits from already approved checkpoint inhibitors. In this review, we discuss major hurdles of immunotherapy like the immunosuppressive niche and low immunogenicity of CRC next to current achievements of checkpoint inhibitors, interleukin treatment and adoptive cell transfer (dendritic cells/cytokine induced killer cells, tumor infiltrating lymphocytes, chimeric antigen receptor cells, T cell receptor transfer) in pre-clinical models and clinical trials. We intensively examine approaches to overcome low immunogenicity by combination of different therapies and address future strategies of therapy as well as the need of predictive factors in this emerging field of precision medicine.
- Published
- 2019
- Full Text
- View/download PDF
6. A multicenter open-label phase II trial to evaluate nivolumab and ipilimumab for 2nd line therapy in elderly patients with advanced esophageal squamous cell cancer (RAMONA)
- Author
-
Nadja M, Meindl-Beinker, Johannes, Betge, Tobias, Gutting, Elke, Burgermeister, Sebastian, Belle, Tianzuo, Zhan, Nadine, Schulte, Martin, Maenz, Matthias P, Ebert, and Nicolai, Haertel
- Subjects
Aged, 80 and over ,Esophageal Neoplasms ,Programmed Cell Death 1 Receptor ,Esophageal squamous cell cancer ,Comprehensive geriatric assessment ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Ipilimumab ,Survival Analysis ,Personalized medicine ,lcsh:RC254-282 ,B7-H1 Antigen ,Study Protocol ,Nivolumab ,Treatment Outcome ,610 Medical sciences Medicine ,Elderly ,Antineoplastic Combined Chemotherapy Protocols ,Carcinoma, Squamous Cell ,Humans ,Geriatric oncology ,Checkpoint inhibitors ,Aged - Abstract
Background Advanced esophageal squamous cell cancer (ESCC) is frequently diagnosed in elderly patients. The impact of 2nd line chemotherapy is poorly defined. Recent data demonstrated effectiveness of checkpoint inhibitors in different squamous cell carcinomas. Therefore, we assess combined nivolumab/ipilimumab as 2nd line therapy in elderly ESCC patients. Methods RAMONA is a multicenter open-label phase II trial. The primary objective is to demonstrate a significant survival benefit of nivolumab/ipilimumab in advanced ESCC compared to historical data of standard chemotherapy. Primary endpoint is therefore overall survival (OS). Major secondary objective is the evaluation of tolerability. Time to QoL deterioration will thus be determined as key secondary endpoint. Further secondary endpoints are tumor response, PFS and safety. We aim to recruit a total of n = 75 subjects that have to be > 65 years old. Eligibility is determined by the geriatric status (G8 screening and Deficit Accumulation Frailty Index (DAFI)). A safety assessment will be performed after a 3 cycle run-in phase of nivolumab (240 mg Q2W) to justify escalation for eligible patients to combined nivolumab (240 mg Q2W) and ipilimumab (1 mg/kg Q6W), while the other patients will remain on nivolumab only. RAMONA also includes translational research sub-studies to identify predictive biomarkers, including PD-1 and PD-L1 evaluation at different time points, establishment of organoid cultures and microbiome analyses for response prediction. Discussion The RAMONA trial aims to implement checkpoint inhibitors for elderly patients with advanced ESCC as second line therapy. Novel biomarkers for checkpoint-inhibitor response are analyzed in extensive translational sub-studies. Trial registration EudraCT Number: 2017–002056-86; NCT03416244, registered: 31.1.2018. Electronic supplementary material The online version of this article (10.1186/s12885-019-5446-2) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
7. PPARγ induces PD-L1 expression in MSS+ colorectal cancer cells
- Author
-
Maximilian Eckardt, Kay Klapproth, Veronika Hauber, Philip Weidner, Michael Boutros, Tianzuo Zhan, Matthias P. Ebert, Beifang Li, Timo Gaiser, Wenyue Wu, Jens Pahl, Sebastian Belle, Frank Herweck, Elke Burgermeister, Adelheid Cerwenka, Torsten Schroeder, Juliane Reichling, Laura Helm, Ioana Dobrota, Carsten Sticht, Tobias Gutting, and Johannes Betge
- Subjects
0301 basic medicine ,mss ,medicine.medical_treatment ,Immunology ,Peroxisome proliferator-activated receptor ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,pd-l1 ,PD-L1 ,Tumor Microenvironment ,medicine ,Humans ,cancer ,Immunology and Allergy ,RC254-282 ,Original Research ,colorectal ,chemistry.chemical_classification ,Tumor microenvironment ,biology ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,Microsatellite instability ,Immunotherapy ,RC581-607 ,medicine.disease ,Immune checkpoint ,PPAR gamma ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Microsatellite Instability ,immunotherapy ,Immunologic diseases. Allergy ,Colorectal Neoplasms ,ppar ,business ,Research Article - Abstract
Only a small subset of colorectal cancer (CRC) patients benefits from immunotherapies, comprising blocking antibodies (Abs) against checkpoint receptor “programmed-cell-death-1” (PD1) and its ligand (PD-L1), because most cases lack the required mutational burden and neo-antigen load caused by microsatellite instability (MSI) and/or an inflamed, immune cell-infiltrated PD-L1+ tumor microenvironment. Peroxisome proliferator-activated-receptor-gamma (PPARγ), a metabolic transcription factor stimulated by anti-diabetic drugs, has been previously implicated in pre/clinical responses to immunotherapy. We therefore raised the hypothesis that PPARγ induces PD-L1 on microsatellite stable (MSS) tumor cells to enhance Ab-target engagement and responsiveness to PD-L1 blockage. We found that PPARγ-agonists upregulate PD-L1 mRNA/protein expression in human gastrointestinal cancer cell lines and MSS+ patient-derived tumor organoids (PDOs). Mechanistically, PPARγ bound to and activated DNA-motifs similar to cognate PPARγ-responsive-elements (PPREs) in the proximal −2 kb promoter of the human PD-L1 gene. PPARγ-agonist reduced proliferation and viability of tumor cells in co-cultures with PD-L1 blocking Ab and lymphokine-activated killer cells (LAK) derived from the peripheral blood of CRC patients or healthy donors. Thus, metabolic modifiers improved the antitumoral response of immune checkpoint Ab, proposing novel therapeutic strategies for CRC.
- Published
- 2021
- Full Text
- View/download PDF
8. Insulin-like growth factor 2 expression in prostate cancer is regulated by promoter-specific methylation
- Author
-
Maurice Stephan Michel, Alexander Marx, Tobias Gutting, Djeda Belharazem, Stefan Küffer, Christian Sauer, Philipp Ströbel, and Lutz Trojan
- Subjects
Male ,0301 basic medicine ,Cancer Research ,endocrine system diseases ,medicine.medical_treatment ,Prostate cancer ,0302 clinical medicine ,Prostate ,Gene expression ,Promoter Regions, Genetic ,Research Articles ,biology ,IGF2 ,General Medicine ,Methylation ,Middle Aged ,prostate cancer ,targeted therapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,female genital diseases and pregnancy complications ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Molecular Medicine ,imprinting ,Research Article ,animal structures ,Down-Regulation ,promoter methylation ,lcsh:RC254-282 ,Genomic Imprinting ,03 medical and health sciences ,Insulin-Like Growth Factor II ,Genetics ,medicine ,Humans ,RNA, Messenger ,Aged ,Neoplasm Staging ,Growth factor ,Prostatic Neoplasms ,Promoter ,DNA Methylation ,medicine.disease ,030104 developmental biology ,Insulin-like growth factor 2 ,Cancer research ,biology.protein ,Neoplasm Grading - Abstract
Deregulation of the insulin-like growth factor (IGF) axis and dysbalance of components of the IGF system as potential therapeutic targets have been described in different tumor types. IGF2 is a major embryonic growth factor and an important activator of IGF signaling. It is regulated by imprinting in a development- and tissue-dependent manner and has been implicated in a broad range of malignancies including prostate cancer (PCa). Loss of imprinting (LOI) usually results in bi-allelic gene expression and increased levels of IGF2. However, the regulatory mechanisms and the pathophysiological impact of altered IGF2 expression in PCa remain elusive. Here, we show that in contrast to many other tumors, IGF2 mRNA and protein levels were decreased in 80% of PCa in comparison with non-neoplastic adjacent prostate and were independent of LOI status. Instead, IGF2 expression in both tumors and adjacent prostate depended on preferential usage of the IGF2 promoters P3 and P4. Decreased IGF2 expression in tumors was strongly related to hypermethylation of these two promoters. Methylation of the A region in promoter P4 correlated specifically with IGF2 expression in the 20% of PCa where IGF2 was higher in tumors than in adjacent prostate. We conclude that IGF2 is downregulated in most PCa and may be particularly relevant during early stages of tumor development or during chemotherapy and androgen deprivation. PCa differs from other tumors in that IGF2 expression is mainly regulated through methylation of promoter-specific and not by imprinting. Targeting of promoter-specific regions may have relevance for the adjuvant treatment of PCa.
- Published
- 2018
- Full Text
- View/download PDF
9. Myotubularin-related protein 7 inhibits insulin signaling in colorectal cancer
- Author
-
Peter Kienle, Tobias Gutting, Christoph Röcken, Carsten Hopf, Timo Gaiser, Hans-Michael Behrens, Elke Burgermeister, Rony Seger, Julia Magdeburg, Teresa Friedrich, Matthias P. Ebert, Michaela Söhn, Philip Weidner, Hermelindis Ruh, and Tamar Hanoch
- Subjects
0301 basic medicine ,Gerontology ,Oncology ,Male ,Myotubularin ,Colorectal cancer ,Mice ,0302 clinical medicine ,Tissue Distribution ,Neoplasm Metastasis ,biology ,Middle Aged ,Protein Tyrosine Phosphatases, Non-Receptor ,myotubularin ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,Biological regulation ,Colorectal Neoplasms ,Research Paper ,Signal Transduction ,medicine.medical_specialty ,Poor prognosis ,insulin ,colorectal cancer ,phosphatase ,MTMR7 ,03 medical and health sciences ,Genomic Imprinting ,Growth factor receptor ,Insulin-Like Growth Factor II ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,In patient ,Gene Silencing ,Cell Proliferation ,business.industry ,Gene Expression Profiling ,medicine.disease ,Insulin receptor ,030104 developmental biology ,Increased risk ,Diabetes Mellitus, Type 2 ,Tissue Array Analysis ,biology.protein ,business - Abstract
// Philip Weidner 1, * , Michaela Sohn 1, * , Tobias Gutting 1 , Teresa Friedrich 1 , Timo Gaiser 2 , Julia Magdeburg 3 , Peter Kienle 3 , Hermelindis Ruh 4 , Carsten Hopf 4 , Hans-Michael Behrens 5 , Christoph Rocken 5 , Tamar Hanoch 6 , Rony Seger 6 , Matthias P.A. Ebert 1 , Elke Burgermeister 1 1 Department of Medicine II, Universitatsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, D-68167 Mannheim, Germany 2 Institute of Pathology, Universitatsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, D-68167 Mannheim, Germany 3 Department of Surgery, Universitatsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, D-68167 Mannheim, Germany 4 ABIMAS Research Center, Mannheim University of Applied Sciences, D-68163 Mannheim, Germany 5 Institute of Pathology, Christian Albrecht University, D-24105 Kiel, Germany 6 Department of Biological Regulation, Weizmann Institute of Science, I-7610001 Rehovot, Israel * These authors have contributed equally to this work Correspondence to: Elke Burgermeister, email: elke.burgermeister@medma.uni-heidelberg.de Keywords: colorectal cancer, insulin, MTMR7, phosphatase, myotubularin Received: January 13, 2016 Accepted: June 16, 2016 Published: July 07, 2016 ABSTRACT Phosphoinositide (PIP) phosphatases such as myotubularins (MTMs) inhibit growth factor receptor signaling. However, the function of myotubularin-related protein 7 (MTMR7) in cancer is unknown. We show that MTMR7 protein was down-regulated with increasing tumor grade (G), size (T) and stage (UICC) in patients with colorectal cancer (CRC) (n=1786). The presence of MTMR7 in the stroma correlated with poor prognosis, whereas MTMR7 expression in the tumor was not predictive for patients’ survival. Insulin reduced MTMR7 protein levels in human CRC cell lines, and CRC patients with type 2 diabetes mellitus (T2DM) or loss of imprinting (LOI) of insulin-like growth factor 2 ( IGF2) had an increased risk for MTMR7 loss. Mechanistically, MTMR7 lowered PIPs and inhibited insulin-mediated AKT-ERK1/2 signaling and proliferation in human CRC cell lines. MTMR7 provides a novel link between growth factor signaling and cancer, and may thus constitute a potential marker or drug target for human CRC.
- Published
- 2016
10. Therapeutic drug monitoring (TDM) of 5-fluorouracil (5-FU): new preanalytic aspects
- Author
-
Sihem Aida, Ralf-Dieter Hofheinz, Sonani Mindt, Michael Neumaier, Annette Müller, Maren Hedtke, Kirsten Merx, and Tobias Gutting
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Continuous infusion ,Clinical Biochemistry ,Urology ,Pre-Analytical Phase ,03 medical and health sciences ,0302 clinical medicine ,Port (medical) ,Neoplasms ,medicine ,Humans ,Vein ,Aged ,Body surface area ,Aged, 80 and over ,Immunoassay ,Venipuncture ,medicine.diagnostic_test ,business.industry ,Biochemistry (medical) ,Palliative Care ,Area under the curve ,General Medicine ,Middle Aged ,030104 developmental biology ,medicine.anatomical_structure ,ROC Curve ,Fluorouracil ,Therapeutic drug monitoring ,030220 oncology & carcinogenesis ,Area Under Curve ,Female ,Drug Monitoring ,business ,medicine.drug - Abstract
Background 5-Fluorouracil (5-FU) is frequently used for the treatment of gastrointestinal tumors. The pharmacological effect of 5-FU is influenced by genetic polymorphisms as well as differently dosed regimens. Currently, 5-FU is generally administered as a continuous infusion via an implanted port system using a body surface area (BSA)-based dose calculation. In order to optimize treatment, the area under the curve (AUC) can be estimated to allow for individual dose adjustment. A 5-FU AUC range between 20 and 30 [mg×h×L] is recommended. The aim of the current study was to assess if blood for AUC analysis could also be drawn at the side where the port system had been placed. Methods We collected EDTA blood samples of patients receiving infusional 5-FU simultaneously from different sampling points (right/left cubital vein). 5-FU concentrations were measured in a steady-state equilibrium based on nanoparticle immunoassay (My5-FU; Saladax). Results A total of 39 patients took part in this study. About half of the patients did not reach the target 5-FU concentration window (37% were under- and 16% of the patients were overdosed). Calculated median AUC was 23.3 for the right arm (range 5.8–59.4) and a median of 23.4 for the left arm (range 5.3–61.0). AUC values showed no difference between right compared to left arms (p=0.99). Conclusions In all, these results confirm that a high percentage of patients are not treated with 5-FU doses reaching suggested AUC levels of 20–30. The location of venepuncture, however, had no impact on the results of plasma 5-FU concentration.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.