Network


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

Hotspot


Dive into the research topics where Katharina Ochs is active.

Publication


Featured researches published by Katharina Ochs.


Frontiers in Immunology | 2015

Cancer Immunotherapy by Targeting IDO1/TDO and Their Downstream Effectors.

Michael Platten; Nikolaus von Knebel Doeberitz; Iris Oezen; Wolfgang Wick; Katharina Ochs

The tryptophan (TRP) to kynurenine (KYN) metabolic pathway is now firmly established as a key regulator of innate and adaptive immunity. A plethora of preclinical models suggests that this immune tolerance pathway – driven by the key and rate-limiting enzymes indoleamine-2,3-dioxygenase and TRP-2,3-dioxygenase – is active in cancer immunity, autoimmunity, infection, transplant rejection, and allergy. Drugs targeting this pathway, specifically indoleamine-2,3-dioxygenase, are already in clinical trials with the aim at reverting cancer-induced immunosuppression. In the past years, there has been an increase in our understanding of the regulation and downstream mediators of TRP metabolism, such as the aryl hydrocarbon receptor as a receptor for KYN and kynurenic acid. This more detailed understanding will expand our opportunities to interfere with the pathway therapeutically on multiple levels. Here, we discuss the perspective of targeting TRP metabolism at these different levels based on reviewing recent insight into the regulation of TRP metabolism and its downstream effectors.


PLOS ONE | 2011

The Indoleamine-2,3-Dioxygenase (IDO) Inhibitor 1-Methyl-D-tryptophan Upregulates IDO1 in Human Cancer Cells

Christiane A. Opitz; Ulrike Litzenburger; Uta Opitz; Felix Sahm; Katharina Ochs; Christian Lutz; Wolfgang Wick; Michael Platten

1-methyl-D-tryptophan (1-D-MT) is currently being used in clinical trials in patients with relapsed or refractory solid tumors with the aim of inhibiting indoleamine-2,3-dioxygenase (IDO)-mediated tumor immune escape. IDO is expressed in tumors and tumor-draining lymph nodes and degrades tryptophan (trp) to create an immunsuppressive micromilieu both by depleting trp and by accumulating immunosuppressive metabolites of the kynurenine (kyn) pathway. Here we show that proliferation of alloreactive T-cells cocultured with IDO1-positive human cancer cells paradoxically was inhibited by 1-D-MT. Surprisingly incubation with 1-D-MT increased kyn production of human cancer cells. Cell-free assays revealed that 1-D-MT did not alter IDO1 enzymatic activity. Instead, 1-D-MT induced IDO1 mRNA and protein expression through pathways involving p38 MAPK and JNK signalling. Treatment of cancer patients with 1-D-MT has transcriptional effects that may promote rather than suppress anti-tumor immune escape by increasing IDO1 in the cancer cells. These off-target effects should be carefully analyzed in the ongoing clinical trials with 1-D-MT.


Journal of Neuroimmunology | 2013

Immature mesenchymal stem cell-like pericytes as mediators of immunosuppression in human malignant glioma

Katharina Ochs; Felix Sahm; Christiane A. Opitz; Tobias V. Lanz; Iris Oezen; Pierre Olivier Couraud; Andreas von Deimling; Wolfgang Wick; Michael Platten

Malignant gliomas are primary brain tumors characterized by profound local immunosuppression. While the remarkable plasticity of perivascular cells - resembling mesenchymal stem cells (MSC) - in malignant gliomas and their contribution to angiogenesis is increasingly recognized, their role as potential mediators of immunosuppression is unknown. Here we demonstrate that FACS-sorted malignant glioma-derived pericytes (HMGP) were characterized by the expression of CD90, CD248, and platelet-derived growth factor receptor-β (PDGFR-β). HMGP shared this expression profile with human brain vascular pericytes (HBVP) and human MSC (HMSC) but not human cerebral microvascular endothelial cells (HCMEC). CD90+PDGFR-β+perivascular cells distinct from CD31+ endothelial cells accumulated in human gliomas with increasing degree of malignancy and negatively correlated with the presence of blood vessel-associated leukocytes and CD8+ T cells. Cultured CD90+PDGFR-β+HBVP were equally capable of suppressing allogeneic or mitogen-activated T cell responses as human MSC. HMGP, HBVP and HMSC expressed prostaglandin E synthase (PGES), inducible nitric oxide synthase (iNOS), human leukocyte antigen-G (HLA-G), hepatocyte growth factor (HGF) and transforming growth factor-β (TGF-β). These factors but not indoleamine 2,3-dioxygenase-mediated conversion of tryptophan to kynurenine functionally contributed to immunosuppression of immature pericytes. Our data provide evidence that human cerebral CD90+ perivascular cells possess T cell inhibitory capability comparable to human MSC and suggest that these cells, besides their critical role in tumor vascularization, also promote local immunosuppression in malignant gliomas and possibly other brain diseases.


Current Neurology and Neuroscience Reports | 2014

Microenvironmental Clues for Glioma Immunotherapy

Michael Platten; Katharina Ochs; Dieter Lemke; Christiane A. Opitz; Wolfgang Wick

Gliomas have been viewed for decades as inaccessible for a meaningful antitumor immune response as they grow in a sanctuary site protected from infiltrating immune cells. Moreover, the glioma microenvironment constitutes a hostile environment for an efficient antitumor immune response as glioma-derived factors such as transforming growth factor β and catabolites of the essential amino acid tryptophan paralyze T-cell function. There is growing evidence from preclinical and clinical studies that a meaningful antitumor immunity exists in glioma patients and that it can be activated by vaccination strategies. As a consequence, the concept of glioma immunotherapy appears to be experiencing a renaissance with the first phase 3 randomized immunotherapy trials entering the clinical arena. On the basis of encouraging results from other tumor entities using immunostimulatory approaches by blocking endogenous T-cell suppressive pathways mediated by cytotoxic T-lymphocyte antigen 4 or programmed cell death protein 1/programmed cell death protein 1 ligand 1 with humanized antibodies, there is now a realistic and promising option to combine active immunotherapy with agents blocking the immunosuppressive microenvironment in patients with gliomas to allow a peripheral antitumor immune response induced by vaccination to become effective. Here we review the current clinical and preclinical evidence of antimicroenvironment immunotherapeutic strategies in gliomas.


Glia | 2015

Suppression of TDO-mediated tryptophan catabolism in glioblastoma cells by a steroid-responsive FKBP52-dependent pathway

Martina Ott; Ulrike Litzenburger; Katharina J. Rauschenbach; Lukas Bunse; Katharina Ochs; Felix Sahm; Stefan Pusch; Christiane A. Opitz; Jonas Blaes; Andreas von Deimling; Wolfgang Wick; Michael Platten

Tryptophan catabolism is increasingly recognized as a key and druggable molecular mechanism active in cancer, immune, and glioneural cells and involved in the modulation of antitumor immunity, autoimmunity and glioneural function. In addition to the pivotal rate limiting enzyme indoleamine‐2,3‐dioxygenase, expression of tryptophan‐2,3‐dioxygenase (TDO) has recently been described as an alternative pathway responsible for constitutive tryptophan degradation in malignant gliomas and other types of cancer. In addition, TDO has been implicated as a key regulator of neurotoxicity involved in neurodegenerative diseases and ageing. The pathways regulating TDO expression, however, are largely unknown. Here, a siRNA‐based transcription factor profiling in human glioblastoma cells revealed that the expression of human TDO is suppressed by endogenous glucocorticoid signaling. Similarly, treatment of glioblastoma cells with the synthetic glucocorticoid dexamethasone led to a reduction of TDO expression and activity in vitro and in vivo. TDO inhibition was dependent on the immunophilin FKBP52, whose FK1 domain physically interacted with the glucocorticoid receptor as demonstrated by bimolecular fluorescence complementation and in situ proximity ligation assays. Accordingly, gene expression profile analyses revealed negative correlation of FKBP52 and TDO in glial and neural tumors and in normal brain. Knockdown of FKBP52 and treatment with the FK‐binding immunosuppressant FK506 enhanced TDO expression and activity in glioblastoma cells. In summary, we identify a novel steroid‐responsive FKBP52‐dependent pathway suppressing the expression and activity of TDO, a central and rate‐limiting enzyme in tryptophan metabolism, in human gliomas. GLIA 2015;63:78–90


eLife | 2016

Correlated magnetic resonance imaging and ultramicroscopy (MR-UM) is a tool kit to assess the dynamics of glioma angiogenesis

Michael O. Breckwoldt; Julia Bode; Felix T. Kurz; Angelika Hoffmann; Katharina Ochs; Martina Ott; Katrin Deumelandt; Thomas Krüwel; Daniel Schwarz; Xavier Helluy; David Milford; Klara Kirschbaum; Gergely Solecki; Sara Chiblak; Amir Abdollahi; Frank Winkler; Wolfgang Wick; Michael Platten; Sabine Heiland; Martin Bendszus; Björn Tews

Neoangiogenesis is a pivotal therapeutic target in glioblastoma. Tumor monitoring requires imaging methods to assess treatment effects and disease progression. Until now mapping of the tumor vasculature has been difficult. We have developed a combined magnetic resonance and optical toolkit to study neoangiogenesis in glioma models. We use in vivo magnetic resonance imaging (MRI) and correlative ultramicroscopy (UM) of ex vivo cleared whole brains to track neovascularization. T2* imaging allows the identification of single vessels in glioma development and the quantification of neovessels over time. Pharmacological VEGF inhibition leads to partial vascular normalization with decreased vessel caliber, density, and permeability. To further resolve the tumor microvasculature, we performed correlated UM of fluorescently labeled microvessels in cleared brains. UM resolved typical features of neoangiogenesis and tumor cell invasion with a spatial resolution of ~5 µm. MR-UM can be used as a platform for three-dimensional mapping and high-resolution quantification of tumor angiogenesis. DOI: http://dx.doi.org/10.7554/eLife.11712.001


Journal of Neurochemistry | 2016

Tryptophan‐2,3‐dioxygenase is regulated by prostaglandin E2 in malignant glioma via a positive signaling loop involving prostaglandin E receptor‐4

Katharina Ochs; Martina Ott; Katharina J. Rauschenbach; Katrin Deumelandt; Felix Sahm; Christiane A. Opitz; Andreas von Deimling; Wolfgang Wick; Michael Platten

Malignant gliomas and other types of tumors generate a local immunosuppressive microenvironment, which prohibits an effective anti‐tumor immune response and promotes tumor growth. Along with others, we have recently demonstrated that catabolism of the essential amino acid tryptophan via tryptophan‐2,3‐dioxygenase (TDO) is an important mechanism mediating tumor‐associated immunosuppression particularly in gliomas. The pathways regulating TDO in tumors, however, are poorly understood. Here, we show that prostaglandins enhance TDO expression and enzymatic activity in malignant gliomas via activation of prostaglandin E receptor‐4 (EP4). Stimulation with prostaglandin E2 (PGE2) up‐regulated TDO‐mediated kynurenine release in human glioma cell lines, whereas knockdown of the PGE2 receptor EP4 inhibited TDO expression and activity. In human malignant glioma tissue expression of the PGE2‐producing enzyme cyclooxygenase‐2 (COX2) and its receptor EP4 were associated with TDO expression both on transcript and protein level. High expression of EP4 correlated with poor survival in malignant glioma patients WHO III‐IV. Importantly, treatment of glioma cells with an EP4 inhibitor decreased TDO expression and activity. Moreover, TDO‐over‐expressing murine gliomas showed increased COX2 and EP4 expression suggesting a positive feedback mechanism in vivo. In summary, targeting EP4 may inhibit – in addition to immunosuppressive COX2 signaling – tryptophan degradation as another important immunosuppressive pathway and thus, could provide a dual clinically relevant immunotherapeutic avenue for the treatment of malignant gliomas.


Scientific Reports | 2018

Fourier Transform Infrared Microscopy Enables Guidance of Automated Mass Spectrometry Imaging to Predefined Tissue Morphologies

Jan Hinrich Rabe; Denis A. Sammour; Sandra Schulz; Bogdan Munteanu; Martina Ott; Katharina Ochs; Peter Hohenberger; Alexander Marx; Michael Platten; Christiane A. Opitz; Daniel S. Ory; Carsten Hopf

Multimodal imaging combines complementary platforms for spatially resolved tissue analysis that are poised for application in life science and personalized medicine. Unlike established clinical in vivo multimodality imaging, automated workflows for in-depth multimodal molecular ex vivo tissue analysis that combine the speed and ease of spectroscopic imaging with molecular details provided by mass spectrometry imaging (MSI) are lagging behind. Here, we present an integrated approach that utilizes non-destructive Fourier transform infrared (FTIR) microscopy and matrix assisted laser desorption/ionization (MALDI) MSI for analysing single-slide tissue specimen. We show that FTIR microscopy can automatically guide high-resolution MSI data acquisition and interpretation without requiring prior histopathological tissue annotation, thus circumventing potential human-annotation-bias while achieving >90% reductions of data load and acquisition time. We apply FTIR imaging as an upstream modality to improve accuracy of tissue-morphology detection and to retrieve diagnostic molecular signatures in an automated, unbiased and spatially aware manner. We show the general applicability of multimodal FTIR-guided MALDI-MSI by demonstrating precise tumor localization in mouse brain bearing glioma xenografts and in human primary gastrointestinal stromal tumors. Finally, the presented multimodal tissue analysis method allows for morphology-sensitive lipid signature retrieval from brains of mice suffering from lipidosis caused by Niemann-Pick type C disease.


Scientific Reports | 2018

Author Correction: Fourier Transform Infrared Microscopy Enables Guidance of Automated Mass Spectrometry Imaging to Predefined Tissue Morphologies

Jan-Hinrich Rabe; Denis A. Sammour; Sandra Schulz; Bogdan Munteanu; Martina Ott; Katharina Ochs; Peter Hohenberger; Alexander Marx; Michael Platten; Christiane A. Opitz; Daniel S. Ory; Carsten Hopf

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.


Oncotarget | 2014

Constitutive IDO expression in human cancer is sustained by an autocrine signaling loop involving IL-6, STAT3 and the AHR

Ulrike Litzenburger; Christiane A. Opitz; Felix Sahm; Katharina J. Rauschenbach; Saskia Trump; Marcus Winter; Martina Ott; Katharina Ochs; Christian Lutz; Xiangdong Liu; Natasa Anastasov; Irina Lehmann; Thomas Höfer; Andreas von Deimling; Wolfgang Wick; Michael Platten

Collaboration


Dive into the Katharina Ochs's collaboration.

Top Co-Authors

Avatar

Wolfgang Wick

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Christiane A. Opitz

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Martina Ott

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Michael Platten

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Platten

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Andreas von Deimling

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lukas Bunse

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

A von Deimling

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge