Network


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

Hotspot


Dive into the research topics where Hans-Georg Lerchen is active.

Publication


Featured researches published by Hans-Georg Lerchen.


Cancer Research | 2016

Preclinical efficacy of the auristatin-based antibody-drug conjugate BAY 1187982 for the treatment of FGFR2-positive solid tumors.

Anette Sommer; Charlotte Kopitz; Christoph Schatz; Carl Friedrich Nising; Christoph Mahlert; Hans-Georg Lerchen; Beatrix Stelte-Ludwig; Stefanie Hammer; Simone Greven; Joachim Schumacher; Manuela Braun; Ruprecht Zierz; Sabine Wittemer-Rump; Axel Harrenga; Frank Dittmer; Frank Reetz; Heiner Apeler; Rolf Jautelat; Hung Huynh; Karl Ziegelbauer; Bertolt Kreft

The fibroblast growth factor receptor FGFR2 is overexpressed in a variety of solid tumors, including breast, gastric, and ovarian tumors, where it offers a potential therapeutic target. In this study, we present evidence of the preclinical efficacy of BAY 1187982, a novel antibody-drug conjugate (ADC). It consists of a fully human FGFR2 monoclonal antibody (mAb BAY 1179470), which binds to the FGFR2 isoforms FGFR2-IIIb and FGFR2-IIIc, conjugated through a noncleavable linker to a novel derivative of the microtubule-disrupting cytotoxic drug auristatin (FGFR2-ADC). In FGFR2-expressing cancer cell lines, this FGFR2-ADC exhibited potency in the low nanomolar to subnanomolar range and was more than 100-fold selective against FGFR2-negative cell lines. High expression levels of FGFR2 in cells correlated with efficient internalization, efficacy, and cytotoxic effects in vitro Pharmacokinetic analyses in mice bearing FGFR2-positive NCI-H716 tumors indicated that the toxophore metabolite of FGFR2-ADC was enriched more than 30-fold in tumors compared with healthy tissues. Efficacy studies demonstrated that FGFR2-ADC treatment leads to a significant tumor growth inhibition or tumor regression of cell line-based or patient-derived xenograft models of human gastric or breast cancer. Furthermore, FGFR2 amplification or mRNA overexpression predicted high efficacy in both of these types of in vivo model systems. Taken together, our results strongly support the clinical evaluation of BAY 1187982 in cancer patients and a phase I study (NCT02368951) has been initiated. Cancer Res; 76(21); 6331-9. ©2016 AACR.


Cancer Research | 2014

Abstract 4491: FGFR2-ADC potently and selectively inhibits growth of gastric and breast cancer xenograft models

Anette Sommer; Carl Friedrich Nising; Christoph Mahlert; Charlotte Kopitz; Hans-Georg Lerchen; Simone Greven; Beatrix Stelte-Ludwig; Joachim Schuhmacher; Ruprecht Zierz; Sabine Wittemer-Rump; Christoph Schatz; Frank Reetz; Heiner Apeler; Rolf Jautelat; Bertolt Kreft; Karl Ziegelbauer

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Antibody-drug conjugates (ADCs) represent a promising therapeutic approach for treatment of cancer. We have developed a novel ADC directed against fibroblast growth factor receptor 2 (FGFR2). FGFR2 is overexpressed in several cancer indications, such as gastric cancer and breast cancer, thus representing an interesting therapeutic target for the treatment of FGFR2 positive cancer patients with an ADC-based therapy. The FGFR2-ADC consists of the fully human anti-FGFR2-mAb BAY 1179470 conjugated via a stable linker to a novel auristatin cytotoxic agent (technology licensed from Seattle Genetics). The FGFR2-mAb BAY 1179470, which is cross-reactive with human, mouse, rat and monkey, induces internalization of FGFR2. Quantitative data on FGFR2 antibody bound per cell (ABC) were determined with the QuantiBrite assay using BAY 1179470. FGFR2-ADC has a potency in the single digit nM to sub nM range in a panel of FGFR2-positive cells lines (e.g., SNU-16, KatoIII, SUM52-PE, MFM-223) and shows more than 100-fold selectivity versus FGFR2-negative cell lines. High levels of FGFR2 on cancer cells correlate with internalization efficacy and cytotoxic activity in vitro. FGFR2-ADC is highly efficacious in monotherapy and results in tumor growth inhibition in the gastric cancer xenograft model SNU-16 and tumor regression in the breast cancer xenograft model MFM-223. At doses efficacious in mice, FGFR2-ADC is well tolerated. The pre-clinical efficacy and tolerability data obtained for FGFR2-ADC suggest a therapeutic index and support clinical testing. Citation Format: Anette Sommer, Carl F. Nising, Christoph Mahlert, Charlotte C. Kopitz, Hans-Georg Lerchen, Simone Greven, Beatrix Stelte-Ludwig, Joachim Schuhmacher, Ruprecht Zierz, Sabine Wittemer-Rump, Christoph Schatz, Frank Reetz, Heiner Apeler, Rolf Jautelat, Bertolt Kreft, Karl Ziegelbauer. FGFR2-ADC potently and selectively inhibits growth of gastric and breast cancer xenograft models. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4491. doi:10.1158/1538-7445.AM2014-4491


Cancer Research | 2014

Abstract 5445: Preclinical anti-tumor efficacy of an anti-C4.4a (LYPD3) antibody drug conjugate for the treatment of lung squamous cell carcinoma

Joerg Willuda; Lars Linden; Hans-Georg Lerchen; Charlotte Kopitz; Sven Golfier; Ute Bach; Joachim Schumacher; Beatrix Stelte-Ludwig; Oliver von Ahsen; Claudia Schneider; Frank Dittmer; Rudolf Beier; Sherif El-Sheik; Jan Tebbe; Gabriele Leder; Heiner Apeler; Rolf Jautelat; Bertolt Kreft; Karl Ziegelbauer

C4.4a (LYPD3) has been identified previously as a cancer- and metastasis-associated internalizing cell surface protein. Targeting C4.4a with a specific antibody-drug conjugate (ADC) represents an unique opportunity to treat tumors with high unmet medical need such as squamous cell carcinomas SCC, in particular lung SCC. We have generated an anti-C4.4a ADC consisting of a fully human monoclonal antibody linked to a non cell-permeable tubulin-binding auristatin cytotoxic agent (technology licensed from Seattle Genetics). In vitro, anti-C4.4a ADC showed an anti-proliferative efficacy (IC50) in the nanomolar range in cell lines endogenously expressing C4.4a (e.g. human lung cancer cell lines NCI-H292 and NCI-H322). High ADC stability and selectivity was observed in transfected A549 lung cancer cells over-expressing C4.4a compared to mock-transfected cells. In vivo, anti-C4.4a ADC exhibited a potent and selective antitumor activity in various human xenograft models (NCI-H292, NCI-H322, SCC-4) as well as in two SCC (Lu7433, Lu7343) and one pleomorphic (Lu7064) patient-derived lung cancer xenograft models. The in vivo efficacy is strictly target-dependent and selective as no efficacy was observed in C4.4a negative models (Fadu, Lu 7700) or using a non-specific isotype antibody ADC (NCI-H292, NCI-H322). A minimal effective dose (MED) as low as 1.9 mg/kg, response rates of up to 100%, and additive anti-tumor efficacy in combination with cisplatin were observed in the NCI-H292 xenograft model. Furthermore, it has been demonstrated that NCI-H292 were still sensitive to ADC treatment when tumors were allowed to regrow after the initial treatment cycle(). The anti-C4.4a ADC, which is fully cross-reactive with the mouse orthologue of C4.4a, was well tolerated at efficacious doses. Reversible skin reddening was observed only at doses markedly higher than the MED. In summary, anti-C4.4a ADC is a promising therapeutic candidate for the treatment of C4.4a-expressing squamous cell carcinomas, andpreclinical development has been initiated. Citation Format: Joerg Willuda, Lars Linden, Hans-Georg Lerchen, Charlotte Kopitz, Sven Golfier, Ute Bach, Joachim Schumacher, Beatrix Stelte-Ludwig, Oliver Von Ahsen, Claudia Schneider, Frank Dittmer, Rudolf Beier, Sherif El-Sheik, Jan Tebbe, Gabriele Leder, Heiner Apeler, Rolf Jautelat, Bertolt Kreft, Karl Ziegelbauer. Preclinical anti-tumor efficacy of an anti-C4.4a (LYPD3) antibody drug conjugate for the treatment of lung squamous cell carcinoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5445. doi:10.1158/1538-7445.AM2014-5445


Archive | 2009

AMINO ACID ESTER PRODRUGS AND THE USE THEREOF

Daniel Meibom; Hans-Georg Lerchen; Alexandros Vakalopoulos; Barbara Albrech-Küpper; Peter Nell; Jörg Keldenich; Katja Zimmermann; Ursula Krenz


Archive | 2008

Prodrugs of 2-amino-6- ( { [2- (4-chlorphenyl) -1, 3-thiaz0l-4-yl] methyl}thio) -4- [4- ( 2 -hydroxyethoxy) phenyl] pyridin-3, 5-dicarbonitrile

Nicole Diedrichs; Thomas Krahn; Ursula Krenz; Joerg Keldenich; Hanna Tinel; Claudia Hirth-Dietrich; Hans-Georg Lerchen


Archive | 2016

ANTIBODY DRUG CONJUGATES (ADCS) WITH KINESIN SPINDEL PROTEIN (KSP)

Hans-Georg Lerchen; Sven Wittrock; Nils Griebenow; Beatrix Stelte-Ludwig; Anette Sommer; Sandra Berndt; Christoph Mahlert; Mario Lobell; Carsten Terjung; Simone Greven; Yolanda Cancho Grande


Archive | 2010

AMINOACYL PRODRUGS AS AN ACTIVE PHARMACEUTICAL INGREDIENT FOR THROMBOEMBOLIC DISORDERS

Hans-Georg Lerchen; Ursula Krenz; Michael Härter; Mark Jean Gnoth; Georges von Degenfeld; Elke Dittrich-wengenroth; Anja Buchmüller; Susanne Röhrig; Swen Allerheiligen; Elisabeth Perzborn; Christoph Gerdes; Karl-Heinz Schlemmer; Metin Akbaba


Archive | 2015

ANTIBODY-DRUG CONJUGATES (ADCS) OF KSP INHIBITORS WITH AGLYCOSYLATED ANTI-TWEAKR ANTIBODIES

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


Archive | 2012

POLYETHYLENE GLYCOL BASED PRODRUG OF ADRENOMEDULLIN AND USE THEREOF

Ingo Flamme; Johannes Köbberling; Hans-Georg Lerchen; Nils Griebenow; Rudolf Schohe-Loop; Sven Wittrock; Maria Köllnberger; Frank Wunder; Gorden Redlich; Andreas Knorr; Julie Marley; Iain Pritchard


Archive | 2012

TYROSINE BASED LINKERS FOR THE RELEASABLE CONNECTION OF PEPTIDES

Ingo Flamme; Johannes Köbberling; Hans-Georg Lerchen; Nils Griebenow; Rudolf Schohe-Loop; Sven Wittrock; Ursula Krenz

Collaboration


Dive into the Hans-Georg Lerchen's collaboration.

Top Co-Authors

Avatar

Ursula Krenz

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Beatrix Stelte-Ludwig

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Alexandros Vakalopoulos

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Anette Sommer

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Daniel Meibom

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Gerhard Siemeister

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Joerg Keldenich

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Katja Zimmermann

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Peter Nell

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Simone Greven

Bayer HealthCare Pharmaceuticals

View shared research outputs
Researchain Logo
Decentralizing Knowledge