Network


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

Hotspot


Dive into the research topics where Hans-Ulrich Siegmund is active.

Publication


Featured researches published by Hans-Ulrich Siegmund.


Frontiers in Physiology | 2011

A Computational Systems Biology Software Platform for Multiscale Modeling and Simulation: Integrating Whole-Body Physiology, Disease Biology, and Molecular Reaction Networks

Thomas Eissing; Lars Kuepfer; Corina Becker; Michael Block; Katrin Coboeken; Thomas Gaub; Linus Goerlitz; Juergen Jaeger; Roland Loosen; Bernd Ludewig; Michaela Meyer; Christoph Niederalt; Michael Sevestre; Hans-Ulrich Siegmund; Juri Solodenko; Kirstin Thelen; Ulrich Telle; Wolfgang Weiss; Thomas Wendl; Stefan Willmann; Joerg Lippert

Today, in silico studies and trial simulations already complement experimental approaches in pharmaceutical R&D and have become indispensable tools for decision making and communication with regulatory agencies. While biology is multiscale by nature, project work, and software tools usually focus on isolated aspects of drug action, such as pharmacokinetics at the organism scale or pharmacodynamic interaction on the molecular level. We present a modeling and simulation software platform consisting of PK-Sim® and MoBi® capable of building and simulating models that integrate across biological scales. A prototypical multiscale model for the progression of a pancreatic tumor and its response to pharmacotherapy is constructed and virtual patients are treated with a prodrug activated by hepatic metabolization. Tumor growth is driven by signal transduction leading to cell cycle transition and proliferation. Free tumor concentrations of the active metabolite inhibit Raf kinase in the signaling cascade and thereby cell cycle progression. In a virtual clinical study, the individual therapeutic outcome of the chemotherapeutic intervention is simulated for a large population with heterogeneous genomic background. Thereby, the platform allows efficient model building and integration of biological knowledge and prior data from all biological scales. Experimental in vitro model systems can be linked with observations in animal experiments and clinical trials. The interplay between patients, diseases, and drugs and topics with high clinical relevance such as the role of pharmacogenomics, drug–drug, or drug–metabolite interactions can be addressed using this mechanistic, insight driven multiscale modeling approach.


PLOS ONE | 2011

Evaluation of the Efficacy and Safety of Rivaroxaban Using a Computer Model for Blood Coagulation

Rolf Burghaus; Katrin Coboeken; Thomas Gaub; Lars Kuepfer; Anke Sensse; Hans-Ulrich Siegmund; Wolfgang Weiss; Wolfgang Mueck; Joerg Lippert

Rivaroxaban is an oral, direct Factor Xa inhibitor approved in the European Union and several other countries for the prevention of venous thromboembolism in adult patients undergoing elective hip or knee replacement surgery and is in advanced clinical development for the treatment of thromboembolic disorders. Its mechanism of action is antithrombin independent and differs from that of other anticoagulants, such as warfarin (a vitamin K antagonist), enoxaparin (an indirect thrombin/Factor Xa inhibitor) and dabigatran (a direct thrombin inhibitor). A blood coagulation computer model has been developed, based on several published models and preclinical and clinical data. Unlike previous models, the current model takes into account both the intrinsic and extrinsic pathways of the coagulation cascade, and possesses some unique features, including a blood flow component and a portfolio of drug action mechanisms. This study aimed to use the model to compare the mechanism of action of rivaroxaban with that of warfarin, and to evaluate the efficacy and safety of different rivaroxaban doses with other anticoagulants included in the model. Rather than reproducing known standard clinical measurements, such as the prothrombin time and activated partial thromboplastin time clotting tests, the anticoagulant benchmarking was based on a simulation of physiologically plausible clotting scenarios. Compared with warfarin, rivaroxaban showed a favourable sensitivity for tissue factor concentration inducing clotting, and a steep concentration–effect relationship, rapidly flattening towards higher inhibitor concentrations, both suggesting a broad therapeutic window. The predicted dosing window is highly accordant with the final dose recommendation based upon extensive clinical studies.


Frontiers in Physiology | 2014

Computational investigation of potential dosing schedules for a switch of medication from warfarin to rivaroxaban—an oral, direct Factor Xa inhibitor

Rolf Burghaus; Katrin Coboeken; Thomas Gaub; Christoph Niederalt; Anke Sensse; Hans-Ulrich Siegmund; Wolfgang Weiss; Wolfgang Mueck; Takahiko Tanigawa; Jörg Lippert

The long-lasting anticoagulant effect of vitamin K antagonists can be problematic in cases of adverse drug reactions or when patients are switched to another anticoagulant therapy. The objective of this study was to examine in silico the anticoagulant effect of rivaroxaban, an oral, direct Factor Xa inhibitor, combined with the residual effect of discontinued warfarin. Our simulations were based on the recommended anticoagulant dosing regimen for stroke prevention in patients with atrial fibrillation. The effects of the combination of discontinued warfarin plus rivaroxaban were simulated using an extended version of a previously validated blood coagulation computer model. A strong synergistic effect of the two distinct mechanisms of action was observed in the first 2–3 days after warfarin discontinuation; thereafter, the effect was close to additive. Nomograms for the introduction of rivaroxaban therapy after warfarin discontinuation were derived for Caucasian and Japanese patients using safety and efficacy criteria described previously, together with the coagulation model. The findings of our study provide a mechanistic pharmacologic rationale for dosing schedules during the therapy switch from warfarin to rivaroxaban and support the switching strategies as outlined in the Summary of Product Characteristics and Prescribing Information for rivaroxaban.


Thin Solid Films | 1992

Characterization of Langmuir-Blodgett overlayers by time-of-flight secondary ion mass spectrometry

Birgit Hagenhoff; A. Benninghoven; Hans-Ulrich Siegmund; Dieter Holtkamp

Abstract Time-of-flight secondary ion mass spectrometry (TOF-SIMS) in combination with ion imaging (lateral resolution 1 μm) has been applied to the analysis and characterization of Langmuir-Blodgett (LB) films. Silver, evaporated onto polycarbonate slices, was used as substrate material. Whereas films made from monomeric amphiphiles show defect structures of a diameter less than 1 μm, polymeric amphiphiles were found to form closed and homogeneous layers. Binary mixtures consisting of monomeric species in a polymeric matrix show a vertical diffusion of the monomeric component towards the surface. An additional application of imaging TOF-SIMS is demonstrated by the localization of contaminants.


Thin Solid Films | 1992

An optical biosensor principle based on fluorescence energy transfer

Hans-Ulrich Siegmund; Arno Becker; Holger Dipl Chem Dr Ohst; Klaus Sommer

Abstract The specific binding of a fluorescence-labelled mannoside to immobilized concanavalin A is detected by Forster energy transfer, where the donor fluorophore is polymer-bound in a Langmuir-Blodgett film. The applicability of this method to biosensing is discussed.


Sensors and Actuators B-chemical | 1993

A new way of biosensing using fluorescence energy transfer and Langmuir-Blodgett films

Hans-Ulrich Siegmund; Arno Becker

Abstract A method of detecting the binding of ligand molecules (mannosides) to a biological receptor (Con A) by Forster energy transfer from Langmuir-Blodgett (LB) films is presented. The LB films consist of pre-polymerized amphiphilic polymethacrylates containing a coumarin fluorophore (donor) and reactive groups to which biological receptors can be attached. Studies determining the stability of the system are described. The specific binding of rhodamine (acceptor)-labelled ligands and their competitive replacement by unlablled analytes is demonstrated. A simple dual-channel fluorescence detector optimized for this purpose is described as well.


Journal of Pharmacokinetics and Pharmacodynamics | 2018

A generic whole body physiologically based pharmacokinetic model for therapeutic proteins in PK-Sim

Christoph Niederalt; Lars Kuepfer; Juri Solodenko; Thomas Eissing; Hans-Ulrich Siegmund; Michael Block; Stefan Willmann; Jörg Lippert

Proteins are an increasingly important class of drugs used as therapeutic as well as diagnostic agents. A generic physiologically based pharmacokinetic (PBPK) model was developed in order to represent at whole body level the fundamental mechanisms driving the distribution and clearance of large molecules like therapeutic proteins. The model was built as an extension of the PK-Sim model for small molecules incorporating (i) the two-pore formalism for drug extravasation from blood plasma to interstitial space, (ii) lymph flow, (iii) endosomal clearance and (iv) protection from endosomal clearance by neonatal Fc receptor (FcRn) mediated recycling as especially relevant for antibodies. For model development and evaluation, PK data was used for compounds with a wide range of solute radii. The model supports the integration of knowledge gained during all development phases of therapeutic proteins, enables translation from pre-clinical species to human and allows predictions of tissue concentration profiles which are of relevance for the analysis of on-target pharmacodynamic effects as well as off-target toxicity. The current implementation of the model replaces the generic protein PBPK model available in PK-Sim since version 4.2 and becomes part of the Open Systems Pharmacology Suite.


British Journal of Clinical Pharmacology | 2015

Contribution of rivaroxaban to the international normalized ratio when switching to warfarin for anticoagulation as determined by simulation studies.

Hans-Ulrich Siegmund; Rolf Burghaus; Dagmar Kubitza; Katrin Coboeken

AIM This study evaluated the influence of rivaroxaban 20 mg once daily on international normalized ratio (INR) during the co-administration period when switching from rivaroxaban to warfarin. METHODS We developed a calibrated coagulation model that was qualified with phase I clinical data. Prothrombin time and INR values were simulated by use of phospholipid concentrations that matched Neoplastin Plus® and Innovin® reagents. To simulate the combined effects of rivaroxaban and warfarin on INR during switching, warfarin initiation was simulated by adjusting the magnitude of the warfarin effect to reach the desired target INRs over the course of 21 days. The warfarin effect values (obtained every 6 h) and the desired rivaroxaban plasma concentrations were used. Nomograms were generated from rivaroxaban induced increases in INR. RESULTS The simulation had good prediction quality. Rivaroxaban induced increases in the total INR from the warfarin attributed INR were seen, which increased with rivaroxaban plasma concentration. When the warfarin only INR was 2.0-3.0, the INR contribution of rivaroxaban with Neoplastin Plus® was 0.5-1.2, decreasing to 0.3-0.6 with Innovin® at median trough rivaroxaban plasma concentrations (38 μg l(-1) ). CONCLUSIONS The data indicate that measuring warfarin induced changes in INR are best performed at trough rivaroxaban concentrations (24 h after rivaroxaban dosing) during the co-administration period when switching from rivaroxaban to warfarin. Furthermore, Innovin® is preferable to Neoplastin Plus® because of its substantially lower sensitivity to rivaroxaban, thereby reducing the influence of rivaroxaban on the measured INR.


Archive | 1990

Optical biosensor and method of use

Herbert Hugl; Eberhard Kuckert; Dietmar Möbius; Holger Dipl Chem Dr Ohst; Meinhard Rolf; Hans J. Rosenkranz; Heinrich Christian Schopper; Hans-Ulrich Siegmund; Klaus Sommer; Rolf Wehrmann


Archive | 1997

Electrochemical sensors having improved selectivity and enhanced sensitivity

Karlheinz Hildenbrand; Hans-Ulrich Siegmund

Collaboration


Dive into the Hans-Ulrich Siegmund's collaboration.

Researchain Logo
Decentralizing Knowledge