Network


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

Hotspot


Dive into the research topics where Beatrix Stelte-Ludwig is active.

Publication


Featured researches published by Beatrix Stelte-Ludwig.


Bioorganic & Medicinal Chemistry Letters | 2003

Biphenyls as Potent Vitronectin Receptor Antagonists. Part 2: Biphenylalanine Ureas

Klaus Urbahns; Michael Härter; Andrea Vaupel; Markus Albers; Delf Schmidt; Ulf Brüggemeier; Beatrix Stelte-Ludwig; Christoph Gerdes; Hideki Tsujishita

Vitronectin receptor (alpha(V)beta(3)) antagonism has been implicated in a variety of disease states, like restenosis, osteoporosis and cancer. In this work, we present the development of a novel class of biphenyl vitronectin receptor antagonists. Identified from a focused combinatorial library based on para-bromo phenylalanine, these compounds show nanomolar affinity to the vitronectin receptor and display unprecedented SAR. Their binding mode can be rationalized by computational docking studies using the X-ray structure of alpha(V)beta(3).


Molecular Cancer Therapeutics | 2017

Preclinical Antitumor Efficacy of BAY 1129980—a Novel Auristatin-Based Anti-C4.4A (LYPD3) Antibody–Drug Conjugate for the Treatment of Non–Small Cell Lung Cancer

Jörg Willuda; Lars Linden; Hans-Georg Lerchen; Charlotte Kopitz; Beatrix Stelte-Ludwig; Carol Pena; Claudia Lange; Sven Golfier; Christoph Kneip; Patricia E. Carrigan; Kirk Mclean; Joachim Schuhmacher; Oliver von Ahsen; Jörg Müller; Frank Dittmer; Rudolf Beier; Sherif El Sheikh; Jan Tebbe; Gabriele Leder; Heiner Apeler; Rolf Jautelat; Karl Ziegelbauer; Bertolt Kreft

C4.4A (LYPD3) has been identified as a cancer- and metastasis-associated internalizing cell surface protein that is expressed in non–small cell lung cancer (NSCLC), with particularly high prevalence in the squamous cell carcinoma (SCC) subtype. With the exception of skin keratinocytes and esophageal endothelial cells, C4.4A expression is scarce in normal tissues, presenting an opportunity to selectively treat cancers with a C4.4A-directed antibody–drug conjugate (ADC). We have generated BAY 1129980 (C4.4A-ADC), an ADC consisting of a fully human C4.4A-targeting mAb conjugated to a novel, highly potent derivative of the microtubule-disrupting cytotoxic drug auristatin via a noncleavable alkyl hydrazide linker. In vitro, C4.4A-ADC demonstrated potent antiproliferative efficacy in cell lines endogenously expressing C4.4A and inhibited proliferation of C4.4A-transfected A549 lung cancer cells showing selectivity compared with a nontargeted control ADC. In vivo, C4.4A-ADC was efficacious in human NSCLC cell line (NCI-H292 and NCI-H322) and patient-derived xenograft (PDX) models (Lu7064, Lu7126, Lu7433, and Lu7466). C4.4A expression level correlated with in vivo efficacy, the most responsive being the models with C4.4A expression in over 50% of the cells. In the NCI-H292 NSCLC model, C4.4A-ADC demonstrated equal or superior efficacy compared to cisplatin, paclitaxel, and vinorelbine. Furthermore, an additive antitumor efficacy in combination with cisplatin was observed. Finally, a repeated dosing with C4.4A-ADC was well tolerated without changing the sensitivity to the treatment. Taken together, C4.4A-ADC is a promising therapeutic candidate for the treatment of NSCLC and other cancers expressing C4.4A. A phase I study (NCT02134197) with the C4.4A-ADC BAY 1129980 is currently ongoing. Mol Cancer Ther; 16(5); 893–904. ©2017 AACR.


Angewandte Chemie | 2018

Antibody‐drug conjugates with pyrrole‐based KSP inhibitors as novel payload class

Hans-Georg Lerchen; Sven Wittrock; Beatrix Stelte-Ludwig; Anette Sommer; Sandra Berndt; Nils Griebenow; Anne-Sophie Rebstock; Sarah Johannes; Yolanda Cancho-Grande; Christoph Mahlert; Simone Greven; Carsten Terjung

The number of cytotoxic payload classes successfully employed in antibody-drug conjugates (ADCs) is still rather limited. The identification of ADC payloads with a novel mode of action will increase therapeutic options and potentially increase the therapeutic window. Herein, we describe the utilization of kinesin spindle protein inhibitors (KSPi) as a novel payload class providing highly potent ADCs against different targets, for instance HER-2 or TWEAKR/Fn14. Aspects of technical optimization include the development of different linker attachment sites, the stabilization of ADC linkage to avoid payload deconjugation and finally, the tailor-made design of active metabolites with a long lasting intracellular exposure in the tumor matching the mode of action of KSP inhibition. These KSPi-ADCs are highly potent and selective inu2005vitro and demonstrate inu2005vivo efficacy in a broad panel of tumor models including complete regressions in a patient-derived urothelial cancer model.


Cancer Research | 2011

Abstract 1754: Identification of BAY 94-9343, a mesothelin antibody-drug conjugate (ADC): Characterization and anti-tumor activity in mesothelin-positive preclinical tumor models

Sven Golfier; Antje Kahnert; Iring Heisler; Charlotte Kopitz; Kerstin Berhörster; Beatrix Stelte-Ludwig; Anke Mayer-Bartschmid; Sandra Bruder; Lars Linden; Axel Harrenga; Christoph A. Schatz; Bertolt Kreft; Beate Müller-Tiemann; Karl Ziegelbauer

Monoclonal antibodies have proven to be very effective in the treatment of various cancers, including solid tumors. For example, HERCEPTIN® and Erbitux® are successfully used to treat HER2-positive breast cancer and EGFR-positive colorectal cancer, respectively. Conjugation of cytotoxic drugs to antibodies represents a promising approach to improve cancer therapy. Antibody-drug conjugates (ADCs) are able to deliver highly potent toxophores to tumors while at the same time reducing systemic toxicity. Promising efficacy and tolerability profiles of ADCs have been observed in clinical trials including Hodgkin lymphoma (brentuximab vedotin) and breast cancer (trastuzumab-DM1), thus, development of new ADCs targeting tumor- associated antigens has potential to identifiy novel cancer therapeutics. Mesothelin, a glycoprotein expressed in mesothelial cells found in the membrane lining of the peritoneal and pleural cavities, is overexpressed in all mesotheliomas as well as many ovarian and pancreatic cancers. Due to its limited expression on normal tissues and higher expression in a number of tumor types, mesothelin represents an attractive ADC target. BAY 94-9343 consists of a fully human anti-mesothelin IgG1 antibody conjugated to the potent tubulin-binding drug DM4 with an average of 3.2 drug molecules per antibody. The resulting ADC bound to human recombinant mesothelin with high affinity (Kd = 15nM) leading to antigen-dependent internalization and potent cytotoxicity (nanomolar range in vitro IC50) in tumor cells that express mesothelin either endogenously or exogenously, but not in mesothelin-negative cells. In vivo, BAY 94-9343 demonstrated dose-dependent, mesothelin-specific anti-tumor efficacy in subcutaneous and orthotopic xenograft models at doses between 2.5 and 10 mg/kg using a Q3Dx3 schedule. Endogenously expressing mesothelin tumor models included sc and orthotopic OVCAR3 (ovarian), sc BxPC-3 (pancreatic) and sc NCI-H226 (mesothelioma). Furthermore, in mesothelin-positive patient-derived preclinical tumor models of both platinum-resistant ovarian cancer and gemcitabine-resistant pancreatic cancer, BAY 94-9343 exhibited high anti-tumor efficacy leading to partial and complete tumor regressions with a 10mg/kg Q3Dx3 dosing schedule. This ADC was well tolerated in mice at 10mg/kg (Q3Dx3) without any evidence of body weight loss, compared to either cisplatin or gemcitabine treatments. In summary, BAY 94-9343 is a mesothelin-targeted ADC with promising preclinical anti-tumor activity for mesothelin-positive tumors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1754. doi:10.1158/1538-7445.AM2011-1754


Tetrahedron | 2004

Solid phase synthesis of an extensively focused library of thiadiazole ethers

Josef Pernerstorfer; Michael Brands; Hartmut Schirok; Beatrix Stelte-Ludwig; Elisabeth Woltering


Archive | 2001

β-PHENYLALANINE DERIVATIVES AS INTEGRIN ANTAGONISTS

Andreas Schoop; Gerhard Mueller; Ulf Brueggemeier; Delf Schmidt; Beatrix Stelte-Ludwig; Joerg Keldenich


Archive | 1999

1,4-benzodiazepinone derivatives and their use as integrin antagonists

Klaus Urbahns; Delf Schmidt; Ulf Brüggemeier; Christoph Gerdes; Beatrix Stelte-Ludwig; Jörg Keldenich; Elke Stahl


Archive | 2003

Stents containing pyridine-substituted pyrazolopyridine derivatives

Achim Feurer; Stefan Weigand; Beatrix Stelte-Ludwig; Jeffry-Lynn Grunkemeyer; Jeffrey Low; Johannes-Peter Stasch


Angewandte Chemie | 2018

Antikörper-Wirkstoff-Konjugate mit Pyrrol-basierten KSP-Inhibitoren als Payload-Klasse

Hans-Georg Lerchen; Sven Wittrock; Beatrix Stelte-Ludwig; Anette Sommer; Sandra Berndt; Nils Griebenow; Anne-Sophie Rebstock; Sarah Johannes; Yolanda Cancho-Grande; Christoph Mahlert; Simone Greven; Carsten Terjung


Archive | 2004

New N-alkyl-dibenzoxazepinone derivatives, are aminopeptidase N inhibitors useful for treating cardiovascular, inflammatory, autoimmune or cancer diseases or chronic pain, especially arteriosclerosis

Claudia Hirth-Dietrich; Jaques Dumas; Elisabeth Rank; Beatrix Stelte-Ludwig; Dmitry Zubov

Researchain Logo
Decentralizing Knowledge