Network


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

Hotspot


Dive into the research topics where Bernhard Reis is active.

Publication


Featured researches published by Bernhard Reis.


PLOS ONE | 2012

Expression profiling of human immune cell subsets identifies miRNA-mRNA regulatory relationships correlated with cell type specific expression

Florence Allantaz; Donavan T. Cheng; Tobias Bergauer; Palanikumar Ravindran; Michel F. Rossier; Martin Ebeling; Laura Badi; Bernhard Reis; Hans Bitter; Matilde D'Asaro; Alberto Chiappe; Sriram Sridhar; Gonzalo Durán Pacheco; Michael E. Burczynski; Denis F. Hochstrasser; Jacky Vonderscher; Thomas Matthes

Blood consists of different cell populations with distinct functions and correspondingly, distinct gene expression profiles. In this study, global miRNA expression profiling was performed across a panel of nine human immune cell subsets (neutrophils, eosinophils, monocytes, B cells, NK cells, CD4 T cells, CD8 T cells, mDCs and pDCs) to identify cell-type specific miRNAs. mRNA expression profiling was performed on the same samples to determine if miRNAs specific to certain cell types down-regulated expression levels of their target genes. Six cell-type specific miRNAs (miR-143; neutrophil specific, miR-125; T cells and neutrophil specific, miR-500; monocyte and pDC specific, miR-150; lymphoid cell specific, miR-652 and miR-223; both myeloid cell specific) were negatively correlated with expression of their predicted target genes. These results were further validated using an independent cohort where similar immune cell subsets were isolated and profiled for both miRNA and mRNA expression. miRNAs which negatively correlated with target gene expression in both cohorts were identified as candidates for miRNA/mRNA regulatory pairs and were used to construct a cell-type specific regulatory network. miRNA/mRNA pairs formed two distinct clusters in the network corresponding to myeloid (nine miRNAs) and lymphoid lineages (two miRNAs). Several myeloid specific miRNAs targeted common genes including ABL2, EIF4A2, EPC1 and INO80D; these common targets were enriched for genes involved in the regulation of gene expression (p<9.0E-7). Those miRNA might therefore have significant further effect on gene expression by repressing the expression of genes involved in transcriptional regulation. The miRNA and mRNA expression profiles reported in this study form a comprehensive transcriptome database of various human blood cells and serve as a valuable resource for elucidating the role of miRNA mediated regulation in the establishment of immune cell identity.


Cell | 2017

An Immune Atlas of Clear Cell Renal Cell Carcinoma

Stéphane Chevrier; Jacob H. Levine; Vito Riccardo Tomaso Zanotelli; Karina Silina; Daniel Schulz; Marina Bacac; Carola Ries; Laurie Ailles; Michael Alexander Spencer Jewett; Holger Moch; Maries van den Broek; Christian Beisel; Michael B. Stadler; Craig Gedye; Bernhard Reis; Dana Pe’er; Bernd Bodenmiller

Summary Immune cells in the tumor microenvironment modulate cancer progression and are attractive therapeutic targets. Macrophages and T cells are key components of the microenvironment, yet their phenotypes and relationships in this ecosystem and to clinical outcomes are ill defined. We used mass cytometry with extensive antibody panels to perform in-depth immune profiling of samples from 73 clear cell renal cell carcinoma (ccRCC) patients and five healthy controls. In 3.5 million measured cells, we identified 17 tumor-associated macrophage phenotypes, 22 T cell phenotypes, and a distinct immune composition correlated with progression-free survival, thereby presenting an in-depth human atlas of the immune tumor microenvironment in this disease. This study revealed potential biomarkers and targets for immunotherapy development and validated tools that can be used for immune profiling of other tumor types.


Journal of Hepatology | 2016

Randomized phase II placebo controlled study of codrituzumab in previously treated patients with advanced hepatocellular carcinoma

Ghassan K. Abou-Alfa; Oscar Puig; Bruno Daniele; Masatoshi Kudo; Philippe Merle; Joong Won Park; Paul Ross; Jean Marie Peron; Oliver Ebert; Stephen L. Chan; Tung Ping Poon; Massimo Colombo; Takuji Okusaka; Baek Yeol Ryoo; Beatriz Minguez; Takayoshi Tanaka; Toshihiko Ohtomo; Stacey Ukrainskyj; Frederic Boisserie; Olga Rutman; Ya Chi Chen; Chao Xu; Eliezer Shochat; Lori Jukofsky; Bernhard Reis; Gong Chen; Laura Di Laurenzio; Ray Lee; Chia Jui Yen

BACKGROUND & AIMS Codrituzumab, a humanized monoclonal antibody against Glypican-3 (GPC3) that is expressed in hepatocellular carcinoma (HCC), interacts with CD16/FcγRIIIa and triggers antibody-dependent cytotoxicity. Codrituzumab was studied vs. placebo in a randomized phase II trial in advanced HCC patients who had failed prior systemic therapy. METHODS Patients with advanced HCC who had failed prior systemic therapy, ⩾18years, Eastern cooperative oncology group (ECOG) 0-1, Child-Pugh A were randomized 2:1 to biweekly codrituzumab 1600mg vs. placebo. Patients were stratified based on GPC3 immunohistochemical expression: 2+/3+, 1+, and 0. Primary endpoint was progression free survival. Secondary endpoints include overall survival (OS), tolerability, pharmacokinetics, and an exploratory endpoint in biomarkers analysis. RESULTS 185 patients were enrolled: 125 received codrituzumab and 60 placebo: Median age 64/63, 85/75% male, 46/42% Asian, ECOG 0 65/63%, 74/77% having vascular invasion and/or extra-hepatic metastasis. 84%/70% had prior sorafenib. Drug exposure was 98.4% of planned dose, with an identical adverse events profile between the 2 groups. The median progression free survival and overall survival in the codrituzumab vs. placebo groups in months were: 2.6 vs. 1.5 (hazard ratios 0.97, p=0.87), and 8.7 vs. 10 (hazard ratios 0.96, p=0.82). Projected Ctrough at cycle 3day 1 based exposure, high CD16/FcγRIIIa on peripheral immune cells, and GPC3 expression in the tumor, were all associated with prolonged progression free survival and overall survival. CONCLUSIONS Codrituzumab did not show clinical benefit in this previously treated HCC population. Whether higher codrituzumab drug exposure or the use of CD16 and GPC3 as potential biomarkers would improve outcome remain unanswered questions. LAY SUMMARY Codrituzumab is a manufactured antibody against a liver cancer protein called glypican-3. In this clinical trial, codrituzumab was not found be effective against liver cancer. It was suggested though that a higher dose of codrituzumab or selecting patients with high level of glypican-3 or its mediator CD16 might improve outcome. CLINICAL TRIAL REGISTRATION This trial is registered at Clinicaltrials.gov (NCT01507168).


Haematologica | 2016

Acute myeloid leukemia patients' clinical response to idasanutlin (RG7388) is associated with pre-treatment MDM2 protein expression in leukemic blasts.

Bernhard Reis; Lori Jukofsky; Gong Chen; Giovanni Martinelli; Hua Zhong; W. Venus So; Michael Dickinson; Mark W. Drummond; Sarit Assouline; Maneja Hashemyan; Michel Theron; Steven Blotner; Je-Hwan Lee; Margaret Kasner; Sung-Soo Yoon; Ruediger Rueger; Karen Seiter; Steven Middleton; Kevin R. Kelly; Norbert Vey; Karen Yee; Gwen Nichols; Lin-Chi Chen; William E. Pierceall

In translational research described, we investigated biomarker expression by flow cytometry for MDM2 antagonist clinical response association in relapsed/refractory AML patients treated with idasanutlin-based therapy ( [Clinicaltrials.gov][1] identifier: [NCT01773408][2] ). As MDM2 targets p53 for


Frontiers in Immunology | 2017

Pharmacodynamic Monitoring of RO5459072, a Small Molecule Inhibitor of Cathepsin S

Michel Theron; Darren Bentley; Sandra Nagel; Marianne Manchester; Michael Gerg; Thomas Schindler; Ana Silva; Barbara Ecabert; P. C. Teixeira; Camille Perret; Bernhard Reis

Major histocompatibility complex class II (MHCII)-restricted antigen priming of CD4+ T cells is both involved in adaptive immune responses and the pathogenesis of autoimmune diseases. Degradation of invariant chain Ii, a protein that prevents premature peptide loading, is a prerequisite for nascent MHCII–peptide complex formation. A key proteolytic step in this process is mediated by cathepsin S. Inhibition of this cysteine protease is known to result in the intracellular accumulation of Lip10 in B cells. Here, we describe the development and application of a neoepitope-based flow cytometry assay measuring accumulation of Lip10. This novel method enabled the investigation of cathepsin S-dependent MHCII maturation in professional antigen-presenting cell (APC) subsets. Inhibition of cathepsin S by a specific inhibitor, RO5459072, in human PBMC ex vivo resulted in accumulation of Lip10 in B cells and myeloid dendritic cells, but not in plasmacytoid dendritic cells and only to a minor degree in monocytes. We qualified Lip10 as a pharmacodynamic biomarker by showing the cathepsin S inhibitor-dependent accumulation of Lip10 in vivo in cynomolgus monkeys treated with RO5459072. Finally, dosing of RO5459072 in a first-in-human clinical study (www.ClinicalTrials.gov, identifier NCT02295332) exhibited a dose-dependent increase in Lip10, confirming target engagement and demonstrating desired pharmacologic inhibition in vivo. The degree of cathepsin S antagonist-induced maximum Lip10 accumulation in APCs varied significantly between individuals both in vitro and in vivo. This finding has not been reported previously using alternative, less sensitive methods and demands further investigation as to the potential of this biomarker to predict response to treatment. These results will help guide subsequent clinical studies investigating the pharmacokinetic and pharmacodynamic relationship of cathepsin S inhibitor RO5459072 after multiple dosing.


Oncotarget | 2018

Combining expression of GPC3 in tumors and CD16 on NK cells from peripheral blood to identify patients responding to codrituzumab

Gong Chen; Ya-Chi Chen; Bernhard Reis; Anton Belousov; Lori Jukofsky; Christine Rossin; Axel Muehlig; Chao Xu; Laurent Essioux; Toshihiko Ohtomo; Laura Di Laurenzio; Oscar Puig; Ray Lee

Background Codrituzumab, a monoclonal antibody targeting an oncofetal protein glypican-3 (GPC3) expressed on cell surface of hepatocellular carcinoma (HCC) induces antibody-dependent cellular cytotoxicity (ADCC) and inhibits tumor growth in preclinical studies. Based on this mechanism, tumor GPC3 expression and CD16 expression on NK cells, which are the effector cells of ADCC, were investigated to correlate with codrituzumabs clinical efficacy in patients with advanced HCC. Results Joint analyses of the two biomarkers revealed that both high levels of GPC3 and CD16 were required for patients to benefit from codrituzumab; lack of either one of them would lead to a loss of the therapeutic effect. Conclusions These results suggest the combination of tumor GPC3 expression and CD16 expression on NK cells from peripheral blood at baseline as a composite biomarker to select HCC patients for codrituzumab. Impact The conclusion warrants a future study in an HCC population with both high GPC3 expression and high levels of CD16 at baseline to establish codrituzumabs therapeutic benefit in HCC. Methods Data from a phase II clinical trial of codrituzumab were used for the analyses. GPC3 expression in baseline tumor biopsies was determined by immunohistochemistry (IHC) analysis, and baseline CD16 expression on NK cells were quantified by peripheral blood lymphocyte immunophenotyping. According to high or low expression of GPC3 and CD16, different patient subgroups were formed; for each subgroup, overall survival of patients having high codrituzumab exposure was compared to that of patients receiving placebo.


Biochemical Pharmacology | 2017

Cathepsin S inhibition suppresses autoimmune-triggered inflammatory responses in macrophages

Sophia Thanei; Michel Theron; Ana Silva; Bernhard Reis; Leonore Branco; Lucia Schirmbeck; Fabrice A. Kolb; Wolfgang Haap; Thomas Schindler; Marten Trendelenburg

Graphical abstract Figure. No Caption available. ABSTRACT In several types of antigen‐presenting cells (APCs), Cathepsin S (CatS) plays a crucial role in the regulation of MHC class II surface expression and consequently influences antigen (Ag) presentation of APCs to CD4+ T cells. During the assembly of MHC class II‐Ag peptide complexes, CatS cleaves the invariant chain p10 (Lip10) – a fragment of the MHC class II‐associated invariant chain peptide. In this report, we used a selective, high‐affinity CatS inhibitor to suppress the proteolytic activity of CatS in lymphoid and myeloid cells. CatS inhibition resulted in a concentration‐dependent Lip10 accumulation in B cells from both healthy donors and patients with systemic lupus erythematosus (SLE). Furthermore, CatS inhibition led to a decreased MHC class II expression on B cells, monocytes, and proinflammatory macrophages. In SLE patient‐derived peripheral blood mononuclear cells, CatS inhibition led to a suppressed secretion of IL‐6, TNF&agr;, and IL‐10. In a second step, we tested the effect of CatS inhibition on macrophages being exposed to patient‐derived autoantibodies against C1q (anti‐C1q) that are known to be associated with severe lupus nephritis. As shown previously, those SLE patient‐derived high‐affinity anti‐C1q bound to immobilized C1q induce a proinflammatory phenotype in macrophages. Using this human in vitro model of autoimmunity, we found that CatS inhibition reduces the inflammatory responses of macrophages as demonstrated by a decreased secretion of proinflammatory cytokines, the downregulation of MHC class II and CD80. In summary, we can show that the used CatS inhibitor is able to block Lip10 degradation in healthy donor‐ and SLE patient‐derived B cells and inhibits the induction of proinflammatory macrophages. Thus, CatS inhibition seems to be a promising future treatment of SLE.


Cancer Research | 2016

Abstract 3944: Combining tumor glypican-3 expression and CD16 Expression on NK cells from peripheral blood to identify patients responding to codrituzumab/GC33/RO5137382

Gong Chen; Ya-Chi Chen; Bernhard Reis; Anton Belousov; Lori Jukofsky; Christine Rossin; Axel Muehlig; Chao Xu; Laurent Essioux; Toshihiko Ohtomo; Laura Di Laurenzio; Oscar Puig; Ray Lee

Background & Aims: Codrituzumab (GC33 or RO5137382), a monoclonal antibody targeting an oncofetal protein glypican-3 (GPC3) expressed on the cell surface of hepatocellular carcinoma (HCC), induces antibody-dependent cellular cytotoxicity (ADCC) and inhibits tumor growth in preclinical studies. Based on this mechanism of codrituzumab, two biomarkers GPC3 and CD16 of the NK cells, which are the effector cells of ADCC, were investigated to correlate with clinical efficacy of codrituzumab in patients with advanced HCC. Methods: Data from a phase II clinical trial of codrituzumab were used for this analysis. GPC3 expression was determined by immunohistochemistry (IHC) analysis of tumor biopsies, and CD16 expression on NK cells were quantified by a flow cytometry analysis of peripheral blood mononuclear cells. Overall survival of patients with high exposure of codrituzumab or placebo (n = 113) was used to compare different patient subgroups stratified by high or low expression of GPC3 and CD16. Results: In individual-biomarker analyses, longer survival after codrituzumab treatment correlates with either high expression level of GPC3 in tumors (n = 97, HR = 0.39, 95%CI = 0.21 - 0.70, p = 0.00081) or a relatively high level of CD16 expressed on NK cells (n = 80, HR = 0.44, 95%CI = 0.23 - 0.83, p = 0.0056) at baseline. Furthermore, joint analyses of the two biomarkers reveal that both high levels of GPC3 and CD16 are required for patients to benefit from codrituzumab treatment (n = 67, HR = 0.29, 95%CI = 0.13 - 0.62, p = 0.00074), and lack of either one of them leads to a loss of codrituzumab therapeutic effect. Conclusions: The retrospective analysis supports the mechanism of ADCC, in which the combination of high GPC3 expression in tumors and high CD16 expression in NK cells from peripheral blood is associated with prolonged overall survival given treatment of codrituzumab. This result supports the usage of both GPC3 and CD16 as potential biomarkers to select HCC patients for codrituzumab treatment. Citation Format: Gong Chen, Ya-Chi Chen, Bernhard Reis, Anton Belousov, Lori Jukofsky, Christine Rossin, Axel Muehlig, Chao Xu, Laurent Essioux, Toshihiko Ohtomo, Laura Di Laurenzio, Oscar Puig, Ray Lee. Combining tumor glypican-3 expression and CD16 Expression on NK cells from peripheral blood to identify patients responding to codrituzumab/GC33/RO5137382. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3944.


Cancer Discovery | 2018

Tissue-Specific Immunoregulation: A Call for Better Understanding of the “Immunostat” in the Context of Cancer

W. Pao; Chia-Huey Ooi; Fabian Birzele; Astrid Ruefli-Brasse; Michael Cannarile; Bernhard Reis; Sebastian H. Scharf; David A. Schubert; Klas Hatje; Nadege Pelletier; Olivia Spleiss; John C. Reed


Blood | 2016

Minimal Residual Disease (MRD) Assessment By Multiparametric Flow Cytometry Is Prognostic for Progression-Free Survival in Phase 1/1b Relapsed/Refractory Acute Myeloid Leukemia (AML) Patients Treated with Idasanutlin MDM2 Antagonist

Benjamin Lanza; Giovanni Martinelli; Karen Yee; Lori Jukofsky; Bernhard Reis; Steven Blotner; Mark W. Drummond; Norbert Vey; Karen Seiter; Michael Dickinson; Kevin R. Kelly; Margaret Kasner; Michel Theron; Jeffrey M. Venstrom; Steven Middleton; Lin-Chi Chen; Nelson Kinnersley; Gwen Nichols; William E. Pierceall

Collaboration


Dive into the Bernhard Reis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge