Network


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

Hotspot


Dive into the research topics where Binje Vick is active.

Publication


Featured researches published by Binje Vick.


Hepatology | 2009

Knockout of myeloid cell leukemia‐1 induces liver damage and increases apoptosis susceptibility of murine hepatocytes

Binje Vick; Achim Weber; Toni Urbanik; Thorsten Maass; Andreas Teufel; Peter H. Krammer; Joseph T. Opferman; Marcus Schuchmann; Peter R. Galle; Henning Schulze-Bergkamen

Myeloid cell leukemia‐1 (Mcl‐1) is an antiapoptotic member of the Bcl‐2 protein family. It interacts with proapoptotic Bcl‐2 family members, thereby inhibiting mitochondrial activation and induction of apoptosis. Mcl‐1 is essential for embryonal development and the maintenance of B cells, T cells, and hematopoietic stem cells. We have recently shown that induction of Mcl‐1 by growth factors rescues primary human hepatocytes from CD95‐mediated apoptosis. This prompted us to further analyze the relevance of Mcl‐1 for hepatocellular homeostasis. Therefore, we generated a hepatocyte‐specific Mcl‐1 knockout mouse (Mcl‐1flox/flox‐AlbCre). Deletion of Mcl‐1 in hepatocytes results in liver cell damage caused by spontaneous induction of apoptosis. Livers of Mcl‐1flox/flox‐AlbCre mice are smaller compared to control littermates, due to higher apoptosis rates. As a compensatory mechanism, proliferation of hepatocytes is enhanced in the absence of Mcl‐1. Importantly, hepatic pericellular fibrosis occurs in Mcl‐1 negative livers in response to chronic liver damage. Furthermore, Mcl‐1flox/flox‐AlbCre mice are more susceptible to hepatocellular damage induced by agonistic anti‐CD95 antibodies or concanavalin A. Conclusion: The present study provides in vivo evidence that Mcl‐1 is a crucial antiapoptotic factor for the liver, contributing to hepatocellular homeostasis and protecting hepatocytes from apoptosis induction. (HEPATOLOGY 2009.)


Hepatology | 2010

Hepatocyte‐specific deletion of the antiapoptotic protein myeloid cell leukemia‐1 triggers proliferation and hepatocarcinogenesis in mice

Achim Weber; Rj Boger; Binje Vick; Toni Urbanik; Johannes Haybaeck; Stefan Zoller; Andreas Teufel; Peter H. Krammer; Joseph T. Opferman; Peter R. Galle; Marcus Schuchmann; Mathias Heikenwalder; Henning Schulze-Bergkamen

Regulation of hepatocellular apoptosis is crucial for liver homeostasis. Increased sensitivity of hepatocytes toward apoptosis results in chronic liver injury, whereas apoptosis resistance is linked to hepatocarcinogenesis and nonresponsiveness to therapy‐induced cell death. Recently, we have demonstrated an essential role of the antiapoptotic Bcl‐2 family member Myeloid cell leukemia‐1 (Mcl‐1) in hepatocyte survival. In mice lacking Mcl‐1 specifically in hepatocytes (Mcl‐1Δhep), spontaneous apoptosis caused severe liver damage. Here, we demonstrate that chronically increased apoptosis of hepatocytes coincides with strong hepatocyte proliferation resulting in hepatocellular carcinoma (HCC). Liver cell tumor formation was observed in >50% of Mcl‐1Δhep mice already by the age of 8 months, whereas 12‐month‐old wild‐type (wt) and heterozygous Mcl‐1flox/wt mice lacked tumors. Tumors revealed a heterogenous spectrum ranging from small dysplastic nodules to HCC. The neoplastic nature of the tumors was confirmed by histology, expression of the HCC marker glutamine synthetase and chromosomal aberrations. Liver carcinogenesis in Mcl‐1Δhep mice was paralleled by markedly increased levels of Survivin, an important regulator of mitosis which is selectively overexpressed in common human cancers. Conclusion: This study provides in vivo evidence that increased apoptosis of hepatocytes not only impairs liver homeostasis but is also accompanied by hepatocyte proliferation and hepatocarcinogenesis. Our findings might have implications for understanding apoptosis‐related human liver diseases. (HEPATOLOGY 2010.)


Leukemia | 2016

Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell-induced immune escape mechanism

Christina Krupka; Peter Kufer; Roman Kischel; Gerhard Zugmaier; Felix S. Lichtenegger; Thomas Köhnke; Binje Vick; Irmela Jeremias; Klaus H. Metzeler; Torben Altmann; Stephanie Schneider; Michael Fiegl; Karsten Spiekermann; P A Bauerle; Wolfgang Hiddemann; Gert Riethmüller; Marion Subklewe

Bispecific T-cell engagers (BiTEs) are very effective in recruiting and activating T cells. We tested the cytotoxicity of the CD33/CD3 BiTE antibody construct AMG 330 on primary acute myeloid leukemia (AML) cells ex vivo and characterized parameters contributing to antileukemic cytolytic activity. The E:T ratio and the CD33 expression level significantly influenced lysis kinetics in long-term cultures of primary AML cells (n=38). AMG 330 induced T-cell-mediated proinflammatory conditions, favoring the upregulation of immune checkpoints on target and effector cells. Although not constitutively expressed at the time of primary diagnosis (n=123), PD-L1 was strongly upregulated on primary AML cells upon AMG 330 addition to ex vivo cultures (n=27, P<0.0001). This phenomenon was cytokine-driven as the sole addition of interferon (IFN)-γ and tumor necrosis factor-α also induced expression. Through blockade of the PD-1/PD-L1 interaction, AMG 330-mediated lysis (n=9, P=0.03), T-cell proliferation (n=9, P=0.01) and IFN-γ secretion (n=8, P=0.008) were significantly enhanced. The combinatorial approach was most beneficial in settings of protracted AML cell lysis. Taken together, we have characterized a critical resistance mechanism employed by primary AML cells under AMG 330-mediated proinflammatory conditions. Our results support the evaluation of checkpoint molecules in upcoming clinical trials with AMG 330 to enhance BiTE antibody construct-mediated cytotoxicity.


Science | 2017

The target landscape of clinical kinase drugs

Susan Klaeger; Stephanie Heinzlmeir; Mathias Wilhelm; Harald Polzer; Binje Vick; Paul-Albert Koenig; Maria Reinecke; Benjamin Ruprecht; Svenja Petzoldt; Chen Meng; Jana Zecha; Katrin Reiter; Huichao Qiao; Dominic Helm; Heiner Koch; Melanie Schoof; Giulia Canevari; Elena Casale; Stefania Re Depaolini; Annette Feuchtinger; Zhixiang Wu; Tobias Schmidt; Lars Rueckert; Wilhelm Becker; Jan Huenges; Anne-Kathrin Garz; Bjoern-Oliver Gohlke; Daniel Paul Zolg; Gian Kayser; Tõnu Vooder

An atlas for drug interactions Kinase inhibitors are an important class of drugs that block certain enzymes involved in diseases such as cancer and inflammatory disorders. There are hundreds of kinases within the human body, so knowing the kinase “target” of each drug is essential for developing successful treatment strategies. Sometimes clinical trials can fail because drugs bind more than one target. Yet sometimes off-target effects can be beneficial, and drugs can be repurposed for treatment of additional diseases. Klaeger et al. performed a comprehensive analysis of 243 kinase inhibitors that are either approved for use or in clinical trials. They provide an open-access resource of target summaries that could help researchers develop better drugs, understand how existing drugs work, and design more effective clinical trials. Science, this issue p. eaan4368 The druggable kinome is unraveled. INTRODUCTION Molecularly targeted drugs such as imatinib and crizotinib have revolutionized the treatment of certain blood and lung cancers because of their remarkable clinical success. Over the past 20 years, protein kinases have become a major class of drug targets because these signaling biomolecules are often deregulated in disease, particularly in cancer. Today, 37 small kinase inhibitors (KIs) are approved medicines worldwide and more than 250 drug candidates are undergoing clinical evaluation. RATIONALE Although it is commonly accepted that most KIs target more than one protein, the extent to which this information is available to the public varies greatly between drugs. It would seem important to thoroughly characterize the target spectrum of any drug because additional off-targets may offer opportunities, not only for repurposing but also to explain undesired side effects. To this end, we used a chemical proteomic approach (kinobeads) and quantitative mass spectrometry to characterize the target space of 243 clinical KIs that are approved drugs or have been tested in humans. RESULTS The number of targets for a given drug differed substantially. Whereas some compounds showed exquisite selectivity, others targeted more than 100 kinases simultaneously, making it difficult to attribute their biological effects to any particular mode of action. Also of note is that recently developed irreversible KIs can address more kinases than their intended targets epidermal growth factor receptor (EGFR) and Bruton’s tyrosine kinase (BTK). Collectively, the evaluated KIs targeted 220 kinases with submicromolar affinity, offering a view of the druggable kinome and enabling the development of a universal new selectivity metric termed CATDS (concentration- and target-dependent selectivity). All drug profiles can be interactively explored in ProteomicsDB and a purpose-built shinyApp. Many uses of this unique data and analysis resource by the scientific community can be envisaged, of which we can only highlight a few. The profiles identified many new targets for established drugs, thus improving our understanding of how these drugs might exert their phenotypic effects. For example, we evaluated novel salt-inducible kinase 2 (SIK2) inhibitors for their ability to modulate tumor necrosis factor–α (TNFα) and interleukin-10 (IL-10) production, which may allow repurposing these drugs for inflammatory conditions. Integrating target space information with phosphoproteomic analysis of several EGFR inhibitors enabled the identification of drug response markers inside and outside the canonical EGFR signaling pathway. Off-target identification may also inform drug discovery projects using high-value clinical molecules as lead compounds. We illustrate such a case by a novel structure-affinity relationship analysis of MELK inhibitors based on target profiles and cocrystal structures. To assess the repurposing potential of approved or clinically advanced compounds, we used cell-based assays and mouse xenografts to show that golvatinib and cabozantinib may be used for the treatment of acute myeloid leukemia (AML) based on their FLT3 inhibitory activity. CONCLUSION This work provides a rich data resource describing the target landscape of 243 clinically tested KIs. It is the most comprehensive study to date and illustrates how the information may be used in basic research, drug discovery, or clinical decision-making. Schematic representation of identifying the druggable kinome. A chemical proteomic approach revealed quantitative interaction profiles of 243 clinically evaluated small-molecule KIs covering half of the human kinome. Results can be interactively explored in ProteomicsDB and inform basic biology, drug discovery, and clinical decision-making. Kinase inhibitors are important cancer therapeutics. Polypharmacology is commonly observed, requiring thorough target deconvolution to understand drug mechanism of action. Using chemical proteomics, we analyzed the target spectrum of 243 clinically evaluated kinase drugs. The data revealed previously unknown targets for established drugs, offered a perspective on the “druggable” kinome, highlighted (non)kinase off-targets, and suggested potential therapeutic applications. Integration of phosphoproteomic data refined drug-affected pathways, identified response markers, and strengthened rationale for combination treatments. We exemplify translational value by discovering SIK2 (salt-inducible kinase 2) inhibitors that modulate cytokine production in primary cells, by identifying drugs against the lung cancer survival marker MELK (maternal embryonic leucine zipper kinase), and by repurposing cabozantinib to treat FLT3-ITD–positive acute myeloid leukemia. This resource, available via the ProteomicsDB database, should facilitate basic, clinical, and drug discovery research and aid clinical decision-making.


Nature Medicine | 2017

Loss of the histone methyltransferase EZH2 induces resistance to multiple drugs in acute myeloid leukemia

Stefanie Göllner; Thomas Oellerich; Shuchi Agrawal-Singh; Tino Schenk; Hans-Ulrich Klein; Christian Rohde; Caroline Pabst; Tim Sauer; Mads Lerdrup; Sigal Tavor; Friedrich Stölzel; Sylvia Herold; Gerhard Ehninger; Gabriele Köhler; Kuan Ting Pan; Henning Urlaub; Hubert Serve; Martin Dugas; Karsten Spiekermann; Binje Vick; Irmela Jeremias; Wolfgang E. Berdel; Klaus Hansen; Arthur Zelent; Claudia Wickenhauser; Lutz P. Müller; Christian Thiede; Carsten Müller-Tidow

In acute myeloid leukemia (AML), therapy resistance frequently occurs, leading to high mortality among patients. However, the mechanisms that render leukemic cells drug resistant remain largely undefined. Here, we identified loss of the histone methyltransferase EZH2 and subsequent reduction of histone H3K27 trimethylation as a novel pathway of acquired resistance to tyrosine kinase inhibitors (TKIs) and cytotoxic drugs in AML. Low EZH2 protein levels correlated with poor prognosis in AML patients. Suppression of EZH2 protein expression induced chemoresistance of AML cell lines and primary cells in vitro and in vivo. Low EZH2 levels resulted in derepression of HOX genes, and knockdown of HOXB7 and HOXA9 in the resistant cells was sufficient to improve sensitivity to TKIs and cytotoxic drugs. The endogenous loss of EZH2 expression in resistant cells and primary blasts from a subset of relapsed AML patients resulted from enhanced CDK1-dependent phosphorylation of EZH2 at Thr487. This interaction was stabilized by heat shock protein 90 (HSP90) and followed by proteasomal degradation of EZH2 in drug-resistant cells. Accordingly, inhibitors of HSP90, CDK1 and the proteasome prevented EZH2 degradation, decreased HOX gene expression and restored drug sensitivity. Finally, patients with reduced EZH2 levels at progression to standard therapy responded to the combination of bortezomib and cytarabine, concomitant with the re-establishment of EZH2 expression and blast clearance. These data suggest restoration of EZH2 protein as a viable approach to overcome treatment resistance in this AML patient population.


PLOS ONE | 2015

An Advanced Preclinical Mouse Model for Acute Myeloid Leukemia Using Patients' Cells of Various Genetic Subgroups and In Vivo Bioluminescence Imaging

Binje Vick; Maja Rothenberg; Nadine Sandhöfer; Michela Carlet; Cornelia Finkenzeller; Christina Krupka; Michaela Grunert; Andreas Trumpp; Selim Corbacioglu; Martin Ebinger; Maya C. André; Wolfgang Hiddemann; Stephanie Schneider; Marion Subklewe; Klaus H. Metzeler; Karsten Spiekermann; Irmela Jeremias

Acute myeloid leukemia (AML) is a clinically and molecularly heterogeneous disease with poor outcome. Adequate model systems are required for preclinical studies to improve understanding of AML biology and to develop novel, rational treatment approaches. Xenografts in immunodeficient mice allow performing functional studies on patient-derived AML cells. We have established an improved model system that integrates serial retransplantation of patient-derived xenograft (PDX) cells in mice, genetic manipulation by lentiviral transduction, and essential quality controls by immunophenotyping and targeted resequencing of driver genes. 17/29 samples showed primary engraftment, 10/17 samples could be retransplanted and some of them allowed virtually indefinite serial transplantation. 5/6 samples were successfully transduced using lentiviruses. Neither serial transplantation nor genetic engineering markedly altered sample characteristics analyzed. Transgene expression was stable in PDX AML cells. Example given, recombinant luciferase enabled bioluminescence in vivo imaging and highly sensitive and reliable disease monitoring; imaging visualized minimal disease at 1 PDX cell in 10000 mouse bone marrow cells and facilitated quantifying leukemia initiating cells. We conclude that serial expansion, genetic engineering and imaging represent valuable tools to improve the individualized xenograft mouse model of AML. Prospectively, these advancements enable repetitive, clinically relevant studies on AML biology and preclinical treatment trials on genetically defined and heterogeneous subgroups.


Oncotarget | 2015

Anti-leukemic effects of the V-ATPase inhibitor Archazolid A.

Siwei Zhang; Lina S. Schneider; Binje Vick; Michaela Grunert; Irmela Jeremias; Dirk Menche; Rolf Müller; Angelika M. Vollmar; Johanna Liebl

Prognosis for patients suffering from T-ALL is still very poor and new strategies for T-ALL treatment are urgently needed. Our study shows potent anti-leukemic effects of the myxobacterial V-ATPase inhibitor Archazolid A. Archazolid A reduced growth and potently induced death of leukemic cell lines and human leukemic samples. By inhibiting lysosomal acidification, Archazolid A blocked activation of the Notch pathway, however, this was not the mechanism of V-ATPase inhibition relevant for cell death induction. In fact, V-ATPase inhibition by Archazolid A decreased the anti-apoptotic protein survivin. As underlying mode of action, this work is in line with recent studies from our group demonstrating that Archazolid A induced S-phase cell cycle arrest by interfering with the iron metabolism in leukemic cells. Our study provides evidence for V-ATPase inhibition as a potential new therapeutic option for T-ALL.


Theranostics | 2018

Dual Targeting of Acute Leukemia and Supporting Niche by CXCR4-Directed Theranostics

Stefan Habringer; Constantin Lapa; Peter Herhaus; Margret Schottelius; Rouzanna Istvanffy; Katja Steiger; Julia Slotta-Huspenina; Andreas Schirbel; Heribert Hänscheid; Stefan Kircher; Andreas K. Buck; Katharina Götze; Binje Vick; Irmela Jeremias; Markus Schwaiger; Christian Peschel; Robert A.J. Oostendorp; Hans-Jürgen Wester; Götz-Ulrich Grigoleit; Ulrich auf dem Keller

C-X-C chemokine receptor 4 (CXCR4) is a transmembrane receptor with pivotal roles in cell homing and hematopoiesis. CXCR4 is also involved in survival, proliferation and dissemination of cancer, including acute lymphoblastic and myeloid leukemia (ALL, AML). Relapsed/refractory ALL and AML are frequently resistant to conventional therapy and novel highly active strategies are urgently needed to overcome resistance. Methods: We used patient-derived (PDX) and cell line-based xenograft mouse models of ALL and AML to evaluate the efficacy and toxicity of a CXCR4-targeted endoradiotherapy (ERT) theranostic approach. Results: The positron emission tomography (PET) tracer 68Ga-Pentixafor enabled visualization of CXCR4 positive leukemic burden. In xenografts, CXCR4-directed ERT with 177Lu-Pentixather distributed to leukemia harboring organs and resulted in efficient reduction of leukemia. Despite a substantial in vivo cross-fire effect to the leukemia microenvironment, mesenchymal stem cells (MSCs) subjected to ERT were viable and capable of supporting the growth and differentiation of non-targeted normal hematopoietic cells ex vivo. Finally, three patients with refractory AML after first allogeneic hematopoietic stem cell transplantation (alloSCT) underwent CXCR4-directed ERT resulting in leukemia clearance, second alloSCT, and successful hematopoietic engraftment. Conclusion: Targeting CXCR4 with ERT is feasible and provides a highly efficient means to reduce refractory acute leukemia for subsequent cellular therapies. Prospective clinical trials testing the incorporation of CXCR4 targeting into conditioning regimens for alloSCT are highly warranted.


Leukemia | 2018

Tyrosine kinase inhibition increases the cell surface localization of FLT3-ITD and enhances FLT3-directed immunotherapy of acute myeloid leukemia

Katrin Reiter; Harald Polzer; Christina Krupka; A Maiser; Binje Vick; Maja Rothenberg-Thurley; Klaus H. Metzeler; Daniela Dörfel; H R Salih; G Jung; E Nößner; Irmela Jeremias; Wolfgang Hiddemann; Heinrich Leonhardt; Karsten Spiekermann; Marion Subklewe; Philipp A. Greif

The fms-related tyrosine kinase 3 (FLT3) receptor has been extensively studied over the past two decades with regard to oncogenic alterations that do not only serve as prognostic markers but also as therapeutic targets in acute myeloid leukemia (AML). Internal tandem duplications (ITDs) became of special interest in this setting as they are associated with unfavorable prognosis. Because of sequence-dependent protein conformational changes FLT3-ITD tends to autophosphorylate and displays a constitutive intracellular localization. Here, we analyzed the effect of tyrosine kinase inhibitors (TKIs) on the localization of the FLT3 receptor and its mutants. TKI treatment increased the surface expression through upregulation of FLT3 and glycosylation of FLT3-ITD and FLT3-D835Y mutants. In T cell-mediated cytotoxicity (TCMC) assays, using a bispecific FLT3 × CD3 antibody construct, the combination with TKI treatment increased TCMC in the FLT3-ITD-positive AML cell lines MOLM-13 and MV4-11, patient-derived xenograft cells and primary patient samples. Our findings provide the basis for rational combination of TKI and FLT3-directed immunotherapy with potential benefit for FLT3-ITD-positive AML patients.


Leukemia & Lymphoma | 2018

Frequent and reliable engraftment of certain adult primary acute lymphoblastic leukemias in mice

Birgitta Christine Heckl; Michela Carlet; Binje Vick; Catrin Roolf; Ameera Alsadeq; Michaela Grunert; Wen-Hsin Liu; Andrea Liebl; Wolfgang Hiddemann; Rolf Marschalek; Denis M. Schewe; Karsten Spiekermann; Christian Junghanss; Irmela Jeremias

Birgitta Christine Heckl , Michela Carlet , Binje Vick, Catrin Roolf, Ameera Alsadeq, Michaela Grunert, Wen-Hsin Liu, Andrea Liebl, Wolfgang Hiddemann, Rolf Marschalek, Denis Martin Schewe , Karsten Spiekermann, Christian Junghanss and Irmela Jeremias Research Unit Apoptosis in Hematopoietic Stem Cells (AHS), Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany; German Cancer Consortium (DKTK), Partner Site, Munich, Germany; Department of Medicine III – Hematology, Oncology and Palliative Care, Rostock University Medical Center, Rostock, Germany; Pediatric Hematology/Oncology, ALL-BFM Study Group, Christian Albrechts University Kiel and University Hospital Schleswig-Holstein, Kiel, Germany; Department of Medicine III, University Hospital, LMU, Munich, Germany; Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany; Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University (LMU), Munich, Germany

Collaboration


Dive into the Binje Vick'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

Anne-Kathrin Garz

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Katrin Reiter

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Mathias Heikenwalder

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge