Network


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

Hotspot


Dive into the research topics where Judith Baumeister is active.

Publication


Featured researches published by Judith Baumeister.


Blood | 2011

Antithrombotic drug candidate ALX-0081 shows superior preclinical efficacy and safety compared with currently marketed antiplatelet drugs

Hans Ulrichts; Karen Silence; Anne Schoolmeester; Peter de Jaegere; Stefaan Rossenu; Jan Roodt; Sofie Priem; Marc Lauwereys; Peter Casteels; Femke van Bockstaele; Katrien Verschueren; Patrick Stanssens; Judith Baumeister; Josefin Beate Holz

Neutralizing the interaction of the platelet receptor gpIb with VWF is an attractive strategy to treat and prevent thrombotic complications. ALX-0081 is a bivalent Nanobody which specifically targets the gpIb-binding site of VWF and interacts avidly with VWF. Nanobodies are therapeutic proteins derived from naturally occurring heavy-chain-only Abs and combine a small molecular size with a high inherent stability. ALX-0081 exerts potent activity in vitro and in vivo. Perfusion experiments with blood from patients with acute coronary syndrome on standard antithrombotics demonstrated complete inhibition of platelet adhesion after addition of ALX-0081, while in the absence of ALX-0081 residual adhesion was observed. In a baboon efficacy and safety model measuring acute thrombosis and surgical bleeding, ALX-0081 showed a superior therapeutic window compared with marketed antithrombotics. Pharmacokinetic and biodistribution experiments demonstrated target-mediated clearance of ALX-0081, which leads to a self-regulating disposition behavior. In conclusion, these preclinical data demonstrate that ALX-0081 combines a high efficacy with an improved safety profile compared with currently marketed antithrombotics. ALX-0081 has entered clinical development.


Arthritis Research & Therapy | 2015

The preclinical pharmacology of the high affinity anti-IL-6R Nanobody® ALX-0061 supports its clinical development in rheumatoid arthritis

Maarten Van Roy; Cedric Jozef Neotere Ververken; Els Beirnaert; Sven Hoefman; Joost Kolkman; Michel Vierboom; Elia Breedveld; Bert A. 't Hart; Sofie Poelmans; Lieselot Bontinck; Alex Hemeryck; Sandy Jacobs; Judith Baumeister; Hans Ulrichts

IntroductionThe pleiotropic cytokine interleukin-6 (IL-6) plays an important role in the pathogenesis of different diseases, including rheumatoid arthritis (RA). ALX-0061 is a bispecific Nanobody® with a high affinity and potency for IL-6 receptor (IL-6R), combined with an extended half-life by targeting human serum albumin. We describe here the relevant aspects of its in vitro and in vivo pharmacology.MethodsALX-0061 is composed of an affinity-matured IL-6R-targeting domain fused to an albumin-binding domain representing a minimized two-domain structure. A panel of different in vitro assays was used to characterize the biological activities of ALX-0061. The pharmacological properties of ALX-0061 were examined in cynomolgus monkeys, using plasma levels of total soluble (s)IL-6R as pharmacodynamic marker. Therapeutic effect was evaluated in a human IL-6-induced acute phase response model in the same species, and in a collagen-induced arthritis (CIA) model in rhesus monkeys, using tocilizumab as positive control.ResultsALX-0061 was designed to confer the desired pharmacological properties. A 200-fold increase of target affinity was obtained through affinity maturation of the parental domain. The high affinity for sIL-6R (0.19 pM) translated to a concentration-dependent and complete neutralization of sIL-6R in vitro. In cynomolgus monkeys, ALX-0061 showed a dose-dependent and complete inhibition of hIL-6-induced inflammatory parameters, including plasma levels of C-reactive protein (CRP), fibrinogen and platelets. An apparent plasma half-life of 6.6 days was observed after a single intravenous administration of 10 mg/kg ALX-0061 in cynomolgus monkeys, similar to the estimated expected half-life of serum albumin. ALX-0061 and tocilizumab demonstrated a marked decrease in serum CRP levels in a non-human primate CIA model. Clinical effect was confirmed in animals with active drug exposure throughout the study duration.ConclusionsALX-0061 represents a minimized bispecific biotherapeutic of 26 kDa, nearly six times smaller than monoclonal antibodies. High in vitro affinity and potency was demonstrated. Albumin binding as a half-life extension technology resulted in describable and expected pharmacokinetics. Strong IL-6R engagement was shown to translate to in vivo effect in non-human primates, demonstrated via biomarker deregulation as well as clinical effect. Presented results on preclinical pharmacological properties of ALX-0061 are supportive of clinical development in RA.


Annals of the Rheumatic Diseases | 2013

FRI0021 Alx-0061, an anti-IL-6r nanobody® for therapeutic use in rheumatoid arthritis, demonstrates in vitro a differential biological activity profile as compared to tocilizumab

M. Van Roy; A. Van de Sompel; K. De Smet; J. Jacobs; Tinneke Denayer; Hans Ulrichts; Judith Baumeister; Josefin-Beate Holz

Background Interleukin-6 (IL-6) is a pleiotropic cytokine inducing a wide range of biological activities via its receptor, which can either be soluble (sIL-6R) or membrane-bound (mIL-6R). Blocking of IL-6R results in clinical benefit in rheumatoid arthritis as demonstrated by the marketed IL-6R inhibitor tocilizumab (TCZ). Signalling via the mIL-6R (“classical pathway”) is confined to selected cell types due to the restricted expression of mIL-6R. However, IL-6 can also activate cells through sIL-6R in a process known as trans-signalling. Unwanted pharmacology associated with IL-6 pathway inhibition has been linked to inhibition of mIL-6R. Preferential inhibition of sIL-6R could therefore provide higher therapeutic efficacy with a better side effect profile compared to equivalent inhibition of both IL-6R forms (1). Nanobodies are therapeutic proteins based on the smallest functional fragments of heavy chain-only antibodies, naturally occurring in the Camelidae family. ALX-0061 is a bispecific anti-IL-6R Nanobody engineered to have an extended half-life in vivo by targeting human serum albumin (HSA), in combination with a high target affinity and potency using a single anti-IL-6R building block. Objectives ALX-0061 was extensively characterised using in vitro systems: biological activity and affinity for both sIL-6R and mIL-6R were assessed and compared to TCZ. Methods Biological activity of ALX-0061 and TCZ was analysed in a cell-based assay for mIL-6R, ELISA-based neutralisation assays for sIL-6R, and cell-binding and cell-signalling (mIL-6R) experiments in whole blood from human donors using flow cytometry. Due to very tight target binding, the affinity of ALX-0061 for sIL-6R could not be accurately determined via surface plasmon resonance. Consequently, the more sensitive GyrolabTM platform was used to assess affinity for both receptors. For the KD determination on mIL-6R, free compound concentrations were measured in the supernatant, after pre-incubation of mIL-6R-transfected cells with a constant compound concentration. Results Flow cytometry experiments demonstrated that ALX-0061 binds to mIL-6R expressed on peripheral blood leukocyte populations with expected pharmacology. ALX-0061 specifically neutralised sIL-6R with a 10-fold higher in vitro potency compared to TCZ, while the (apparent) affinity of ALX-0061 for sIL-6R (0.20 pM) was 2000-fold superior compared to TCZ (462 pM). In the mIL-6R-driven cell-based assay, however, in vitro potencies were similar for ALX-0061 and TCZ, with the latter one showing avid binding due to its bivalency. In addition, TCZ showed a 3-fold higher affinity for mIL-6R (160 pM) compared to sIL-6R, while the affinity of ALX-0061 was 45-fold lower for mIL-6R (9 pM) compared to sIL-6R. Conclusions ALX-0061 demonstrates in vitro a preferential biological activity profile for sIL-6R with a lesser activity for mIL-6R, while TCZ has a higher preference for mIL-6R. Preferential inhibition of sIL-6R trans-signalling by ALX-0061 could provide superior therapeutic efficacy with a better side effect profile than TCZ. ALX-0061 is currently in clinical development. Analysis of a phase I/II study demonstrated a strong efficacy and an attractive safety profile. References Waetzig G.H. & Rose-John S., Expert Opin Ther Targets (2012) 16(2) Disclosure of Interest None Declared


Blood | 2009

Microarray-based classifiers and prognosis models identify subgroups with distinct clinical outcomes and high risk of AML transformation of myelodysplastic syndrome

Khalid Abd-Elaziz; Pieter Willem Kamphuisen; Christophe Lyssens; Mariska Reuvers; Izaak den Daas; Femke van Bockstaele; Kristof Vercruysse; Hans Ulrichts; Judith Baumeister; Patricia Crabbe; Veerle Compernolle; Josefin-Beate Holz


Archive | 2012

Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains

Judith Baumeister; Marie-Paule Lucienne Armanda Bouche; Carlo Boutton; Marie-Ange Buyse; Veerle Snoeck; Stephanie Staelens


Antibodies | 2015

Pre-Clinical Intravenous Serum Pharmacokinetics of Albumin Binding and Non-Half-Life Extended Nanobodies® †

Sven Hoefman; Ingrid Ottevaere; Judith Baumeister; Maria Laura Sargentini-Maier


European Respiratory Journal | 2013

ALX-0962, an anti-IgE Nanobody® with a dual mode of action

Manuela Rinaldi; Tinneke Denayer; Sophie Thiolloy; Luis C. Perez Tosar; Marie-Ange Buyse; Philip De Decker; Emilie De Witte; Peter Meerts; Judith Baumeister; Josefin-Beate Holz


Bone | 2012

ALX-0141, an anti-rank-L targeting nanobody®, increases bone mass in cynomolgus monkeys

Rana Samadfam; Sandy Jacobs; M. Van Roy; A. McGuire; P. Schoen; Judith Baumeister; Susan Y. Smith; Josefin-Beate Holz


Archive | 2015

Methods for detecting and/or measuring anti-drug antibodies, in particular treatment-emergent anti-drug antibodies

Veerle Snoeck; Marie-Ange Buyse; Judith Baumeister


Archive | 2013

Biparatopische bindepolypeptide für cxcr2 und verwendungen davon

Zarin Brown; Michelle N. Bradley; Steven J. Charlton; Heeke Gino Anselmus Van; Karen Cromie; Bruno Dombrecht; Soren Steffensen; Judith Baumeister; Marie-Paule Lucienne Armanda Bouche; Carlo Boutton; Marie-Ange Buyse; Veerle Snoeck; Stephanie Staelens

Researchain Logo
Decentralizing Knowledge