Network


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

Hotspot


Dive into the research topics where Oliver Keil is active.

Publication


Featured researches published by Oliver Keil.


Cancer Research | 2008

Atu027, a Liposomal Small Interfering RNA Formulation Targeting Protein Kinase N3, Inhibits Cancer Progression

Manuela Aleku; Petra Schulz; Oliver Keil; Ansgar Santel; Ute Schaeper; Britta Dieckhoff; Oliver Janke; Jens Endruschat; Birgit Durieux; Nadine Röder; Kathrin Löffler; Christian Lange; Melanie Fechtner; Kristin Möpert; Gerald Fisch; Sibylle Dames; Wolfgang Arnold; Karin Jochims; Klaus Giese; Bertram Wiedenmann; Arne Scholz; Jörg Kaufmann

We have previously described a small interfering RNA (siRNA) delivery system (AtuPLEX) for RNA interference (RNAi) in the vasculature of mice. Here we report preclinical data for Atu027, a siRNA-lipoplex directed against protein kinase N3 (PKN3), currently under development for the treatment of advanced solid cancer. In vitro studies revealed that Atu027-mediated inhibition of PKN3 function in primary endothelial cells impaired tube formation on extracellular matrix and cell migration, but is not essential for proliferation. Systemic administration of Atu027 by repeated bolus injections or infusions in mice, rats, and nonhuman primates results in specific, RNAi-mediated silencing of PKN3 expression. We show the efficacy of Atu027 in orthotopic mouse models for prostate and pancreatic cancers with significant inhibition of tumor growth and lymph node metastasis formation. The tumor vasculature of Atu027-treated animals showed a specific reduction in lymph vessel density but no significant changes in microvascular density.


principles and practice of constraint programming | 2012

Phase I clinical development of Atu027, a siRNA formulation targeting PKN3 in patients with advanced solid tumors

Dirk Strumberg; Beate Schultheis; Ulrich Traugott; Christiane Vank; Ansgar Santel; Oliver Keil; Klaus Giese; Jörg Kaufmann; Joachim Drevs

Chemically synthesized, small interfering RNAs (siRNA) are currently used as a new class of therapeutic molecules, allowing the controlled down-regulation of pathologically relevant gene expression e.g., oncogenes and other similar targets in cancer [1, 2, 3].However, the overall negative charge of siRNA molecules (up to 40 negative charg-es) and the relatively high molecular weight (12,000 – 14,000 Da) prevent the functional uptake of these novel therapeutic molecules in vivo. Besides the inefficient uptake and the degradation in endosomal compartments at the cellular level, non-formulated siRNAs are rapidly cleared by renal excretion from the blood stream when administered i.v. [4].To overcome these limitations, a variety of non-viral nanoparticles (50 – 200 nm) have been recently developed enabling chemically synthesized siRNA to be used therapeuti-cally for inhibition of RNAi-mediated tumor growth. Atu027 is a novel RNAi therapeutic agent based on cationic lipoplexes containing chemically stabilized siRNAs, which target Protein Kinase N3 (PKN3) gene expression in the vascular endothelium (Figure 1) [5]. PKN3, a member of the AGC kinase fam-ily, has been identified as a promising, novel therapeutic target in cancer cells for inhibiting tumor progression and lymph node metasta-sis formation [6]. These studies have revealed that PKN3 mediates malignant cell growth downstream of the chronically activated phosphoinositide 3-kinase (PI3K) pathway [6]. Recently, PKN3 has also been considered as a suitable therapeutic target for modulating tumor-associated angiogenesis. Preclinical data, obtained in various cancer mouse mod-els, revealed target-specific, RNAi-mediated silencing of PKN3 expression and significant inhibition of tumor progression and metasta-sis formation [


Journal of Clinical Oncology | 2014

First-in-Human Phase I Study of the Liposomal RNA Interference Therapeutic Atu027 in Patients With Advanced Solid Tumors

Beate Schultheis; Dirk Strumberg; Ansgar Santel; Christiane Vank; Frank Gebhardt; Oliver Keil; Christian Lange; Klaus Giese; Jörg Kaufmann; Michael Khan; Joachim Drevs

PURPOSE Atu027 is a novel liposomal RNA interference therapeutic that includes a short-interfering RNA (siRNA), which silences expression of protein kinase N3 in the vascular endothelium. Atu027 has previously been shown to inhibit local tumor invasion as well as lymph node and pulmonary metastasis in mouse cancer models. This first-in-human study aimed to assess the safety, tolerability, and pharmacokinetics of Atu027 while evaluating therapeutic effects on both primary tumors and metastatic lesions. PATIENTS AND METHODS Thirty-four patients with advanced solid tumors received 10 escalating doses of Atu027 without premedication, as one single followed by eight intravenous infusions twice per week during a 28-day cycle. Response was monitored by computed tomography/magnetic resonance imaging at baseline, at the end of treatment (EoT), and at final follow-up (EoS), and was assessed according to RECIST. RESULTS Atu027 was well tolerated up to dose levels of 0.336 mg/kg; most adverse events (AEs) were low-grade toxicities (grade 1 or 2). No maximum tolerated dose was reached. Plasma levels of siRNA strands and lipids were dose proportional, peaking during 4-hour infusion. Disease stabilization was achieved in 41% of patients at EoT (n = 14 of 34 treated patients); eight patients had stable disease at EoS, and some experienced complete or partial regression of metastases. sFLT1 (soluble variant of vascular endothelial growth factor receptor-1) decreased from pretreatment levels in most patients after dose levels 04 to 10. CONCLUSION Atu027 was safe in patients with advanced solid tumors, with 41% of patients having stable disease for at least 8 weeks. In view of these results, further clinical trials have been initiated, and sFLT1 will be investigated as a potential biomarker.


Clinical Cancer Research | 2010

Atu027 Prevents Pulmonary Metastasis in Experimental and Spontaneous Mouse Metastasis Models

Ansgar Santel; Manuela Aleku; Nadine Röder; Kristin Möpert; Birgit Durieux; Oliver Janke; Oliver Keil; Jens Endruschat; Sibylle Dames; Christian Lange; Mona Eisermann; Kathrin Löffler; Melanie Fechtner; Gerald Fisch; Christiane Vank; Ute Schaeper; Klaus Giese; Jörg Kaufmann

Purpose: Atu027, a novel RNA interference therapeutic, has been shown to inhibit lymph node metastasis in orthotopic prostate cancer mouse models. The aim of this study is to elucidate the pharmacologic activity of Atu027 in inhibiting hematogenous metastasis to the target organ lung in four different preclinical mouse models. Experimental Design: Atu027 compared with vehicle or control small interfering RNA lipoplexes was tested in two experimental lung metastasis models (Lewis lung carcinoma, B16V) and spontaneous metastasis mouse models (MDA-MB-435, MDA-MB-231, mammary fat pad). Different dosing schedules (repeated low volume tail vein injections) were applied to obtain insight into effective Atu027 treatment. Primary tumor growth and lung metastasis were measured, and tissues were analyzed by immunohistochemistry and histology. In vitro studies in human umbilical vein endothelial cells were carried out to provide an insight into molecular changes on depletion of PKN3, in support of efficacy results. Results: Intravenous administration of Atu027 prevents pulmonary metastasis. In particular, formation of spontaneous lung metastasis was significantly inhibited in animals with large tumor grafts as well as in mice with resected primary mammary fat pad tumors. In addition, we provide evidence that an increase in VE-cadherin protein levels as a downstream result of PKN3 target gene inhibition may change endothelial function, resulting in reduced colonization and micrometastasis formation. Conclusion: Atu027 can be considered as a potent drug for preventing lung metastasis formation, which might be suitable for preventing hematogenous metastasis in addition to standard cancer therapy. Clin Cancer Res; 16(22); 5469–80. ©2010 AACR.


Molecular Therapy | 2014

Delivery of Therapeutic siRNA to the Lung Endothelium via Novel Lipoplex Formulation DACC

Volker Fehring; Ute Schaeper; Katharina Ahrens; Ansgar Santel; Oliver Keil; M Eisermann; Klaus Giese; Jörg Kaufmann

Posttranscriptional gene silencing by RNA interference can be therapeutically exploited to inhibit pathophysiological gene expression. However, in contrast to the established effectiveness of RNAi in vitro, safe and effective delivery of siRNAs to specific organs and cell types in vivo remains the major hurdle. Here, we report the development and in vivo characterization of a novel siRNA delivery system (DACC lipoplex) suitable for modulating target gene expression specifically in the lung vasculature. Systemic administration of DACC in mice delivered siRNA cargo functionally to the lung pulmonary endothelium. A single dose of DACC lipoplexes administered by bolus injection or by infusion was sufficient to specifically silence genes expressed in pulmonary endothelial cells such as CD31, Tie-2, VE-cadherin, or BMP-R2. When tested in a mouse model for lung cancer, repeated treatment with DACC/siRNA(CD31) reduced formation of lung metastases and increased life span in a mouse model of experimental lung metastasis.


Microvascular Research | 2008

Intracellular localization of lipoplexed siRNA in vascular endothelial cells of different mouse tissues

Manuela Aleku; Gerald Fisch; Kristin Möpert; Oliver Keil; Wolfgang Arnold; Jörg Kaufmann; Ansgar Santel

Liposomally formulated siRNA can be used for RNAi applications in vivo. Intravenous bolus administration of lipoplexed siRNA has been shown to reduce gene expression in the vascular endothelium. Here, we applied immunofluorescence staining for different endothelial markers (PECAM-1, CD34, laminin) on paraffin sections to compare the respective expression pattern with the intracellular localization of intravenously administered, fluorescently labeled siRNA (siRNA-Cy3-lipoplex). By confocal microscopy, lipoplexed siRNA-Cy3 was detected inside vascular endothelial cells in vivo, which where identified with co-staining of endothelial markers. Consequently, the finding of intracellular siRNA uptake by vascular endothelial cells correlated with RNAi based specific protein reduction in situ as revealed by PECAM-1 specific immunofluorescence staining in lung tissue sections. Therefore, by using a cell biological approach these in situ data emphasize the functional uptake of liposomal siRNA molecules in vascular endothelial cells of different mouse tissues as indicated in our previous molecular study.


Archive | 2007

Lipoplex formulations for specific delivery to vascular endothelium

Jörg Kaufmann; Oliver Keil; Ansgar Santel


Archive | 2007

Means for inhibiting the expression of protein kinase 3

Joerg Kaufmann; Oliver Keil; Ansgar Santel


Journal of Clinical Oncology | 2011

First-in-human phase I study of Atu027, a liposomal small interfering RNA formulation, targeting protein kinase N3 (PKN3) in patients with advanced solid tumors.

Dirk Strumberg; Beate Schultheis; U. Traugott; Christiane Vank; Ansgar Santel; Oliver Keil; K. Giese; Jörg Kaufmann; Joachim Drevs


ASCO Meeting Abstracts | 2012

Antimetastatic activity of Atu027, a liposomal small interfering RNA formulation, targeting protein kinase N3 (PKN3): Final results of a phase I study in patients with advanced solid tumors.

Dirk Strumberg; Beate Schultheis; W Meyer-Sabellek; Christiane Vank; Frank Gebhardt; Ansgar Santel; Oliver Keil; Klaus Giese; Jörg Kaufmann; Joachim Drevs

Collaboration


Dive into the Oliver Keil's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dirk Strumberg

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wolfgang Arnold

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arne Scholz

Humboldt University of Berlin

View shared research outputs
Researchain Logo
Decentralizing Knowledge