Network


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

Hotspot


Dive into the research topics where Andrea Schweinitz is active.

Publication


Featured researches published by Andrea Schweinitz.


Bioorganic & Medicinal Chemistry Letters | 1999

3-Amidinophenylalanine-based inhibitors of urokinase.

Jörg Stürzebecher; Helmut Vieweg; Torsten Steinmetzer; Andrea Schweinitz; Milton T. Stubbs; Martin Renatus; Peter Wikström

Synthesis and anti-uPA activity of a series of Nalpha-triisopropyl-phenylsulfonyl-protected 3-amidinophenylalanine amides are described. We have explored SAR around the C-terminal amide part for inhibition of uPA, plasmin and trypsin. Modification of the amide part has been found to affect potency but not selectivity. With a Ki of 0.41 microM 2r-L is one of the most potent uPA inhibitors described so far. The X-ray crystal structure of 2r-L was solved in complex with trypsin, superimposed with uPA and the results suggest an unique binding mode of this inhibitor type.


Medicinal Chemistry | 2006

New Substrate Analogue Inhibitors of Factor Xa Containing 4-Amidinobenzylamide as P1 Residue: Part 1

Torsten Steinmetzer; Jörg Stürzebecher; Oliver Schuster; Uta Stürzebecher; Andrea Schweinitz; Anne Stürzebecher

The trypsin-like serine protease factor Xa (fXa) is located at the convergence point of the intrinsic and extrinsic coagulation cascade, and therefore has emerged as an attractive target for the design of novel anticoagulants. During the development of substrate-analogue urokinase inhibitors we have found that the protection of the P3-dSer side chain leads to a scaffold of potent fXa inhibitors with the general structure R1-SO2-dSer(R2)-Gly-4-amidinobenzylamide. The first lead (3) with an N-terminal benzylsulfonyl group and dSer(tBu) as P3 residue inhibits human fXa with a Ki of 14 nM. A variety of derivatives with modified P4, P3, and P2 residues have been investigated in terms of inhibition of fXa and related proteases and for their anticoagulant potency and elimination behaviour. Most inhibitors were rapidly cleared from the circulation of rats. However, compound 48 (Ki= 3.5 nM), one of the most potent and selective inhibitors containing a dArg as P3 residue was relatively slowly eliminated (t1/2 approximately 1 h). Inhibitor 48 doubled clotting times in human plasma at 0.32 microM (aPTT) and 0.28 microM (PT), and is approximately 10-fold more potent than the reference fXa inhibitor DX-9065a in the inhibition of the prothrombinase complex. The structures of two inhibitors in complex with human fXa were solved by X-ray crystallography.


Bioorganic & Medicinal Chemistry Letters | 2002

4-Amidinobenzylamine-Based Inhibitors of Urokinase

Sebastian Künzel; Andrea Schweinitz; Siegmund Reißmann; Jörg Stürzebecher; Torsten Steinmetzer

A series of 4-amidinobenzylamine-based peptidomimetic inhibitors of urokinase was synthesized. The most potent one, benzylsulfonyl-D-Ser-Ala-4-amidinobenzylamide 16, inhibits uPA with a K(i) of 7.7 nM but is less selective than 10 with a Gly as P2 residue. Hydroxyamidine and carbonate prodrugs were prepared, which are rapidly converted into the active inhibitors in rats after subcutaneous application.


Journal of Molecular Biology | 2003

Crystals of urokinase type plasminogen activator complexes reveal the binding mode of peptidomimetic inhibitors

Ewa Zeslawska; Uwe Jacob; Andrea Schweinitz; Gary S. Coombs; Wolfram Bode; Edwin L. Madison

Urokinase type plasminogen activator (uPA), a trypsin-like serine proteinase, plays an important role in normal tissue re-modelling, cell adhesion, and cell motility. In addition, studies utilizing normal animals and potent, selective uPA inhibitors or genetically modified mice that lack functional uPA genes have demonstrated that uPA can significantly enhance tumor initiation, growth, progression and metastasis, strongly suggesting that this enzyme may be a promising anti-cancer target. We have investigated the structure-activity relationship (SAR) of peptidomimetic inhibitors of uPA and solved high resolution X-ray structures of key, lead small molecule inhibitors (e.g. phenethylsulfonamidino(P4)-D-seryl(P3)-L-alanyl(P2)-L-argininal(P1) and derivatives thereof) in complex with the uPA proteinase domain. These potent inhibitors are highly selective for uPA. The non-natural D-seryl residue present at the P3 position in these inhibitors contributes substantially to both potency and selectivity because, due to its D-configuration, its side-chain binds in the S4 pocket to interact with the uPA unique residues Leu97b and His99. Additional potency and selectivity can be achieved by optimizing the inhibitor P4 residue to bind a pocket, known as S1sub or S1beta, that is adjacent to the primary specificity pocket of uPA.


Bioorganic & Medicinal Chemistry Letters | 2009

Modification of the N-terminal sulfonyl residue in 3-amidinophenylalanine-based matriptase inhibitors.

Torsten Steinmetzer; Daniel Dönnecke; Martin Korsonewski; Claudia Neuwirth; Peter Steinmetzer; Alexander Schulze; Sebastian M. Saupe; Andrea Schweinitz

Replacement of the N-terminal beta-alanyl-amide moiety in previously identified matriptase inhibitors by non-charged aryl groups caused a slightly decreased potency and partially reduced selectivity, especially towards thrombin. However, some of these analogues are still potent matriptase inhibitors with K(i)-values <10nM. In contrast, improved activity was observed for newly designed tribasic analogues, especially for compound 21, which inhibits matriptase with an K(i)-value of 80pM.


ChemMedChem | 2007

Highly Potent and Selective Substrate Analogue Factor Xa Inhibitors Containing D‐Homophenylalanine Analogues as P3 Residue: Part 2

Anne Stürzebecher; Daniel Dönnecke; Andrea Schweinitz; Oliver Schuster; Peter Steinmetzer; Uta Stürzebecher; Joscha Kotthaus; Bernd Clement; Jörg Stürzebecher; Torsten Steinmetzer

A series of highly potent substrate‐analogue factor Xa inhibitors containing D‐homophenylalanine analogues as the P3 residue has been identified by systematic optimization of a previously described inhibitor structure. An initial lead, benzylsulfonyl‐d‐hPhe‐Gly‐4‐amidinobenzylamide (3), inhibits fXa with an inhibition constant of 6.0 nM. Most modifications of the P2 amino acid and P4 benzylsulfonyl group did not improve the affinity and selectivity of the compounds as fXa inhibitors. In contrast, further variation at the P3 position led to inhibitors with significantly enhanced potency and selectivity. Inhibitor 27, benzylsulfonyl‐D‐homo‐2‐pyridylalanyl(N‐oxide)‐Gly‐4‐amidinobenzylamide, inhibits fXa with a Ki value of 0.32 nM. The inhibitor has strong anticoagulant activity in plasma and doubles the activated partial thromboplastin time and prothrombin time at concentrations of 280 nM and 170 nM, respectively. Compound 27 inhibits the prothrombinase complex with an IC50 value of 5 nM and is approximately 50 times more potent than the reference inhibitor DX‐9065a in this assay.


Bioorganic & Medicinal Chemistry Letters | 2009

Incorporation of neutral C-terminal residues in 3-amidinophenylalanine-derived matriptase inhibitors

Andrea Schweinitz; Daniel Dönnecke; Alexander Ludwig; Peter Steinmetzer; Alexander Schulze; Joscha Kotthaus; Silvia Wein; Bernd Clement; Torsten Steinmetzer

A novel series of matriptase inhibitors based on previously identified tribasic 3-amidinophenylalanine derivatives was prepared. The C-terminal basic group was replaced by neutral residues to reduce the hydrophilicity of the inhibitors. The most potent compound 22 inhibits matriptase with a K(i) value of 0.43 nM, but lacks selectivity towards factor Xa. By combination with neutral N-terminal sulfonyl residues several potent thrombin inhibitors were identified, which had reduced matriptase affinity.


Biological Chemistry | 2003

Increase of anti-metastatic efficacy by selectivity- but not affinity-optimization of synthetic serine protease inhibitors.

Ingo J. Banke; Matthias J. E. Arlt; Caroline J. Pennington; Charlotte Kopitz; Torsten Steinmetzer; Andrea Schweinitz; Bernd Gansbacher; James P. Quigley; Dylan R. Edwards; Jörg Stürzebecher; Achim Krüger

Abstract Although tumors frequently show elevated protease activities, the concept of anti-proteolytic cancer therapy has lost momentum after failure of clinical trials with broad-spectrum matrix metalloproteinase inhibitors. Thus we need to adapt our design strategies for protease inhibitors. Here, we employed a series of seven structurally fine-modulated and pharmacokinetically closely related synthetic 4-amidinobenzylamine based inhibitors with distinct selectivity for prototypical serine proteases in a murine T cell lymphoma liver metastasis model. This in vivo screening revealed efficacy of urokinase inhibitors but no correlation between urokinase selectivity or affinity and antimetastatic effect. In contrast, factor Xa-selective inhibitors were more potent, demonstrating factor Xa or a factor Xa-like serine protease likely to be more determinant in this model. Factor Xa selectivity, but not affinity, significantly improved antimetastatic efficacy. For example, factor Xa inhibitors CJ-504 and CJ-510 exert similar affinity for factor Xa (Ki=14 nM versus 8.8 nM) but CJ-504 was 70-fold more selective for factor Xa. This correlated with higher antimetastatic efficacy (58.8% with CJ-504; 28.2% with CJ-510). Our results show that among the protease inhibitors employed that have affinities in the nanomolar range, the strategy of selectivity-optimization is superior to further improvement of affinity to significantly enhance anti-metastatic efficacy. This appreciation may be important for the future rational design of new anti-proteolytic agents for cancer therapy.


Biological Chemistry | 2014

Correlating structure and ligand affinity in drug discovery: a cautionary tale involving second shell residues

Anastasia Tziridis; Daniel Rauh; Piotr Neumann; Petr Kolenko; Anja Menzel; Ulrike Bräuer; Christian Ursel; Peter Steinmetzer; Jörg Stürzebecher; Andrea Schweinitz; Torsten Steinmetzer; Milton T. Stubbs

Abstract A high-resolution crystallographic structure determination of a protein–ligand complex is generally accepted as the ‘gold standard’ for structure-based drug design, yet the relationship between structure and affinity is neither obvious nor straightforward. Here we analyze the interactions of a series of serine proteinase inhibitors with trypsin variants onto which the ligand-binding site of factor Xa has been grafted. Despite conservative mutations of only two residues not immediately in contact with ligands (second shell residues), significant differences in the affinity profiles of the variants are observed. Structural analyses demonstrate that these are due to multiple effects, including differences in the structure of the binding site, differences in target flexibility and differences in inhibitor binding modes. The data presented here highlight the myriad competing microscopic processes that contribute to protein–ligand interactions and emphasize the difficulties in predicting affinity from structure.


Letters in Peptide Science | 2000

Novel non-peptide lead structures for bradykinin B2-receptor antagonists

L. Felipe Pineda; Claus Liebmann; Sabine Hensellek; Inge Paegelow; Torsten Steinmetzer; Andrea Schweinitz; Jörg Stürzebecher; Siegmund Reissmann

A series of new non-peptide Bradykinin (BK) B2-receptor antagonists is reported. These new leads belong to a larger set including both commercially or otherwise available potential candidates found by proprietary database searches using 3D-pharmacophore models as query, and several bis-benzamidino compounds selected from our tryptase-like protease inhibitor library on the basis of topological considerations, derived from the same models. Some of these compounds show functional competitive antagonistic activity, inhibiting in vitro the BK-induced contraction of isolated guinea-pig ileum (GPI) and rat uterus with a pA2 up to 5.3 and 7.0, respectively. They display also binding affinity (IC50 up to 0.56 μM) to the BK B2-receptor in radioligand binding assays on GPI membrane preparations and on human IMR-90 fetal lung fibroblast cells expressing this receptor subtype. Furthermore, the results with the commercially available compounds, in some cases developed as therapeutics, show that the used 3D-pharmacophore model allows to predict to some certainty possible side actions of potential drugs.

Collaboration


Dive into the Andrea Schweinitz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Torsten Steinmetzer

Schiller International University

View shared research outputs
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