Network


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

Hotspot


Dive into the research topics where Adelbert Grossmann is active.

Publication


Featured researches published by Adelbert Grossmann.


Journal of Separation Science | 2012

Mechanism of improved antibody aggregate separation in polyethylene glycol-modulated cation exchange chromatography.

Simon Kluters; Sebastian Neumann; Thomas von Hirschheydt; Adelbert Grossmann; Andreas Schaubmar; Christian Frech

Ion-exchange chromatography is used in biopharmaceutical downstream processes to reduce product-related impurity levels. Because protein aggregate levels can be considered as a critical quality attribute, the removal of aggregated protein species is of primary importance. The addition of polyethylene glycol (PEG) to the mobile phase in ion-exchange chromatography was found to significantly improve the chromatographic separation of monomers from aggregates. In this work, linear gradient elution experiments with monomeric and aggregated samples of a monoclonal antibody were performed on a strong cation exchange resin at different PEG concentrations to investigate the underlying effects responsible for the observed selectivity improvement. PEG is well known to be excluded from a surface layer volume around the protein and the stationary phase; thus, enhancing adsorption of the preferentially hydrated protein to the hydrated stationary phase. The exclusion volume depends on the accessible surface area of the protein leading to a stronger influence of PEG on larger protein species and thus an improved separation of monomer and aggregates. This hypothesis could be consolidated comparing the distribution equilibrium in PEG solution to that in water by calculating equilibrium constants and transfer free energies using the chromatographic data from the linear gradient elution experiments performed at different pH values.


Cancer Research | 2013

Abstract 486: Tumor-targeted, engineered IL-2 variant (IL-2v)-based immunocytokines for the immunotherapy of cancer.

Christian Klein; Inja Waldhauer; Valeria Nicolini; Claire Dunn; Anne Freimoser-Grundschober; Sylvia Herter; Edwin J. W. Geven; Otto C. Boerman; Tapan Nayak; Erwin van Puijenbroek; David Wittig; Samuel Moser; Oliver Ast; Peter Bruenker; Ralf Hosse; Sabine Lang; Sebastian Neumann; Hubert Kettenberger; Adelbert Grossmann; Ingo H. Gorr; Stefan Evers; Pavel Pisa; Jennifer Fretland; Victor Levitsky; Christian Gerdes; Marina Bacac; Ekkehard Moessner; Pablo Umana

IL-2 therapy can lead to durable responses in cancer patients, but is associated with significant toxicity. None of the described IL-2-based immunocytokines has progressed beyond Phase II trials due to various constraints in their design: 1) pM affinity for IL-2Rαβγ on immune cells and pulmonary vascular endothelium compromising tumor targeting due to the fusion of two wildtype IL-2 moieties to the antibody, together with FcγR binding on the same cells; 2) Rapid systemic clearance and short half-life due to high affinity IL-2Rαβγ binding; 3) Preferential activation of Tregs over immune effectors by wt IL-2. Here, we describe a novel monomeric tumor-targeted immunocytokine where a single, engineered IL-2 variant (IL-2v) with abolished IL-2Rα (CD25) binding is fused to the C-terminus of an antibody with a heterodimeric Fc-part. FcγR and C1q binding is completely abolished by a novel Fc mutation. For targeting, human(-ized) high affinity antibodies against CEA (GA504, CEA-IL2v) or FAP (GA501, FAP-IL2v) were selected. CEA- and FAP-IL2v were recombinantly produced and induction of P-STAT5, proliferation, activation induced cell death (AICD), activation markers and cytokines were determined on effector cells. Safety, pharmacokinetics (PK), tumor targeting, pharmacodynamics and anti-tumor efficacy were analyzed in SCID and immunocompetent C57Bl/6 mice. FAP- and CEA-IL-2v completely lack binding to CD25, but retain IL-Rβγ binding. They do not bind to CD25 or preferentially activate Tregs, and induce lower degree of AICD. However, IL-2Rβγ bioactivity is retained and they activate NK, CD4+ and CD8+ T cells as shown by induction of activation markers and proliferation. In particular, CEA- and FAP-IL2v expand and activate NK cells and skew the CD4:CD8 ratio towards CD8+ T cells in vivo. In C57Bl/6 mice, CEA- and FAP-IL2v demonstrate improved safety despite of higher exposure and circulatory half-life than the corresponding wt IL-2 immunocytokine. MicroSPECT/CT imaging revealed FAP-mediated tumor targeting of FAP-IL2v with low normal tissue uptake with FAP-IL2v tumor targeting being similar to the parental FAP antibody with low accumulation in lymphoid tissues and clearly superior to an FAP-targeted wt IL-2 immunocytokine that shows preferential spleen targeting. Studies in tumor-bearing mice showed dose dependent anti-tumor efficacy of CEA- and FAP-IL2v in established xenograft and syngeneic mouse models. Thus, CEA- and FAP-IL2v demonstrate superior safety, PK and tumor targeting, while lacking preferential induction of Tregs due to abolished CD25 and FcγR binding, monovalency and high-affinity tumor-targeting as compared to classical immunocytokines. They retain capacity to activate NK and T‐effector cells through IL‐2Rβγ; in particular once targeted to the tumor microenvironment. These data support their investigation for the immunotherapy of CEA/FAP-positive tumors. Citation Format: Christian Klein, Inja Waldhauer, Valeria Nicolini, Claire Dunn, Anne Freimoser-Grundschober, Sylvia Herter, Edwin Geven, Otto Boerman, Tapan Nayak, Erwin van Puijenbroek, David Wittig, Samuel Moser, Oliver Ast, Peter Bruenker, Ralf Hosse, Sabine Lang, Sebastian Neumann, Hubert Kettenberger, Adelbert Grossmann, Ingo Gorr, Stefan Evers, Pavel Pisa, Jennifer Fretland, Victor Levitsky, Christian Gerdes, Marina Bacac, Ekkehard Moessner, Pablo Umana. Tumor-targeted, engineered IL-2 variant (IL-2v)-based immunocytokines for the immunotherapy of cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 486. doi:10.1158/1538-7445.AM2013-486


Cancer Research | 2013

Abstract PR8: Novel tumor-targeted, engineered IL-2 variant (IL-2v)-based immunocytokines for immunotherapy of cancer.

Christian Klein; Waldhauer Inja; Valeria Nicolini; Dunn Claire; Anne Freimoser; Sylvia Herter; Edwin J. W. Geven; Otto C. Boerman; Erwin van Puijenbroek; David Wittig; Samuel Moser; Oliver Ast; Ralf Hosse; Sabine Lang; Sebastian Neumann; Adelbert Grossmann; Ingo H. Gorr; Stefan Evers; Pavel Pisa; Jennifer Fretland; Victor Levitsky; Christian Gerdes; Marina Bacac; Ekkehard Moessner; Pablo Umana

Introduction: IL-2 therapy can lead to durable responses in a modest proportion of cancer patients, but the treatment is associated with significant toxicity. Over the last decades, various IL-2-based immunocytokines have been generated by fusing IL-2 to tumor-targeting antibodies. However, none of these molecules have progressed beyond Phase II trials and they are hampered by various liabilities: 1) High functional affinity (low pM) for IL-2Rabg on immune cells and on pulmonary vascular endothelium (Krieg et al., PNAS, 2010) compromising preferential tumor targeting due to fusion of two IL-2 moieties to the antibody. This is further compounded when the immunocytokine binds to FcgRs on the same cells. 2) Rapid systemic clearance and short half-life due to high affinity IL-2Rabg binding. 3) Preferential activation of Tregs over immune effectors due to use of wildtype IL-2. Here, we describe a novel monomeric tumor-targeted immunocytokine where a single, engineered IL-2 variant (IL-2v) with abolished IL-2Ra (CD25) binding is fused to the C-terminus of a tumor-specific hIgG1 antibody with a heterodimeric Fc-part. FcgR and C1q binding is completely abolished by a novel Fc mutation. For targeting, human(-ized) high affinity antibodies against CEA (GA504, CEA-IL2v) or FAP (GA501, FAP-IL2v) were chosen. Experimental procedures: CEA- and FAP-IL2v were recombinantly produced and characterized by surface plasmon resonance. Induction of P-STAT5, proliferation, activation induced cell death (AICD), various activation markers and cytokine release were determined on effector cells. Safety, pharmacokinetics (PK), tumor targeting by imaging, immune-pharmacodynamics and anti-tumor efficacy were analyzed in immunocompromised Scid and immunocompetent C57BL/6 mice. Results: IL-2v completely lacks binding to CD25, but retains IL-Rbg binding. In line with this, FAP- and CEA-IL2v do not bind to CD25 or preferentially activate Tregs, and do not cause AICD. However, IL-2Rbg bioactivity is retained and they are still able to activate NK, CD4 and CD8 T cells as shown by concentration dependent increase in activation markers and induction of proliferation. In particular, CEA- and FAP-IL2v expand and activate NK cells and skew the CD4:CD8 ratio towards activated CD8 T cells in vivo. In C57BL/6 mice CEA- and FAP-IL2v demonstrate improved safety despite of ca. 2-fold higher exposure and t1/2 than a wildtype IL-2-based IgG immunocytokine. SPECT/CT imaging revealed FAP-mediated tumor targeting and accumulation of FAP-IL2v with low normal tissue uptake. Notably, FAP-IL2v tumor targeting was similar to the parental FAP antibody with low accumulation in lymphoid tissues; clearly superior to an FAP-targeted wt IL-2 immunocytokine that showed preferential homing to the spleen. Studies in tumor bearing mice showed dose dependent efficacy of CEA- and FAP-IL2v in established xenograft and immunocompetent syngeneic mouse models in terms of survival. Conclusion: CEA- and FAP-IL2v demonstrate superior safety, PK and tumor targeting, while lacking preferential induction of Tregs due to abolished CD25 and FcgR binding, monovalency and high-affinity tumor-targeting as compared to conventional immunocytokines. They retain the capacity to activate NK and T effector cells through IL 2Rbg; in particular once targeted and immobilized in the tumor microenvironment. These preclinical properties support further investigation for the immunotherapy of CEA/FAP-positive tumors. This abstract is also presented as Poster A23. Citation Format: Christian Klein, Waldhauer Inja, Valeria Nicolini, Dunn Claire, Anne Freimoser, Anne Freimoser, Sylvia Herter, Edwin Geven, Otto Boerman, Erwin van Puijenbroek, David Wittig, Samuel Moser, Oliver Ast, Ralf Hosse, Sabine Lang, Sebastian Neumann, Adelbert Grossmann, Ingo Gorr, Stefan Evers, Pavel Pisa, Jennifer Fretland, Victor Levitsky, Christian Gerdes, Marina Bacac, Ekkehard Moessner, Ekkehard Moessner, Pablo Umana. Novel tumor-targeted, engineered IL-2 variant (IL-2v)-based immunocytokines for immunotherapy of cancer. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology: Multidisciplinary Science Driving Basic and Clinical Advances; Dec 2-5, 2012; Miami, FL. Philadelphia (PA): AACR; Cancer Res 2013;73(1 Suppl):Abstract nr PR8.


FEBS Journal | 2001

The pro-sequence facilitates folding of human nerve growth factor from Escherichia coli inclusion bodies

Anke Rattenholl; Hauke Lilie; Adelbert Grossmann; Anne Stern; Elisabeth Schwarz; Rainer Rudolph


Archive | 2002

Tricyclic lactam and sultam derivatives and their use as histone deacetylase inhibitors

Guy Georges; Adelbert Grossmann; Olaf Mundigl; Wolfgang von der Saal; Tim Sattelkau


Archive | 2006

Antibodies against IL-13 receptor alpha1 and uses thereof

Josef Endl; Maria E. Fuentes; Yvo Graus; Adelbert Grossmann; Sebastian Neumann; Paul Parren; Frank Rebers; Joerg Thomas Regula; Ralf Schumacher; Stefan Seeber; Jan Olaf Stracke; Kay-Gunnar Stubenrauch; Jan van de Winkel; Martine van Vugt; Sandra Vereecken-Verploegen


Archive | 1997

Method of activating denatured protein

Adelbert Grossmann


Archive | 2003

Thiophene hydroxamic acid derivatives

Adelbert Grossmann; Frank Herting; Matthias Koerner; Klaus-Peter Kuenkele; Anja Limberg; Olaf Mundigl; Ulrich Tibes


Archive | 2002

Tricyclic alkylhydroxamate derivatives

Adelbert Grossmann; Wolfgang von der Saal; Tim Sattelkau; Ulrich Tibes


Archive | 2001

Tetralone derivatives as antitumor agents

Guy Georges; Adelbert Grossmann; Tim Sattelkau; Wolfgang Schaefer; Ulrich Tibes

Collaboration


Dive into the Adelbert Grossmann's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge