Network


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

Hotspot


Dive into the research topics where Thomas Stohr is active.

Publication


Featured researches published by Thomas Stohr.


Blood | 2012

Evaluation of efficacy and safety of the anti-vWF Nanobody ALX-0681 in a preclinical baboon model of acquired thrombotic thrombocytopenic purpura

Filip Callewaert; Jan Roodt; Hans Ulrichts; Thomas Stohr; Walter J. Janse van Rensburg; Seb Lamprecht; Stefaan Rossenu; Sofie Priem; Wouter Willems; Josefin-Beate Holz

ALX-0681 is a therapeutic Nanobody targeting the A1-domain of VWF. It inhibits the interaction between ultra-large VWF and platelet GpIb-IX-V, which plays a crucial role in the pathogenesis of thrombotic thrombocytopenic purpura (TTP). In the present study, we report the efficacy and safety profile of ALX-0681 in a baboon model of acquired TTP. In this model, acute episodes of TTP are induced by administration of an ADAMTS13-inhibiting mAb. ALX-0681 completely prevented the rapid onset of severe thrombocytopenia and schistocytic hemolytic anemia. After induction of TTP, platelet counts also rapidly recovered on administration of ALX-0681. This effect was corroborated by the full neutralization of VWF activity. The schistocytic hemolytic anemia was also halted and partially reversed by ALX-0681 treatment. Brain CT scans and post mortem analysis did not reveal any sign of bleeding, suggesting that complete neutralization of VWF by ALX-0681 under conditions of thrombocytopenia was not linked with an excessive bleeding risk. The results obtained in this study demonstrate that ALX-0681 can successfully treat and prevent the most important hallmarks of acquired TTP without evidence of a severe bleeding risk. Therefore, ALX-0681 offers an attractive new therapeutic option for acquired TTP in the clinical setting.


Antimicrobial Agents and Chemotherapy | 2016

Generation and Characterization of ALX-0171, a Potent Novel Therapeutic Nanobody for the Treatment of Respiratory Syncytial Virus Infection

Laurent Detalle; Thomas Stohr; Concepción Palomo; Pedro A. Piedra; Brian E. Gilbert; Vicente Mas; Andrena Millar; Ultan F. Power; Catelijne Stortelers; Koen Allosery; José A. Melero; Erik Depla

ABSTRACT Respiratory syncytial virus (RSV) is an important causative agent of lower respiratory tract infections in infants and elderly individuals. Its fusion (F) protein is critical for virus infection. It is targeted by several investigational antivirals and by palivizumab, a humanized monoclonal antibody used prophylactically in infants considered at high risk of severe RSV disease. ALX-0171 is a trimeric Nanobody that binds the antigenic site II of RSV F protein with subnanomolar affinity. ALX-0171 demonstrated in vitro neutralization superior to that of palivizumab against prototypic RSV subtype A and B strains. Moreover, ALX-0171 completely blocked replication to below the limit of detection for 87% of the viruses tested, whereas palivizumab did so for 18% of the viruses tested at a fixed concentration. Importantly, ALX-0171 was highly effective in reducing both nasal and lung RSV titers when delivered prophylactically or therapeutically directly to the lungs of cotton rats. ALX-0171 represents a potent novel antiviral compound with significant potential to treat RSV-mediated disease.


PLOS ONE | 2014

Protective Effect of Different Anti-Rabies Virus VHH Constructs against Rabies Disease in Mice

Sanne Terryn; Aurélie Francart; Sophie Lamoral; Anna Hultberg; Heidi Rommelaere; Angela Wittelsberger; Filip Callewaert; Thomas Stohr; Kris Meerschaert; Ingrid Ottevaere; Catelijne Stortelers; Peter Vanlandschoot; Michael Kalai; Steven Van Gucht

Rabies virus causes lethal brain infection in about 61000 people per year. Each year, tens of thousands of people receive anti-rabies prophylaxis with plasma-derived immunoglobulins and vaccine soon after exposure. Anti-rabies immunoglobulins are however expensive and have limited availability. VHH are the smallest antigen-binding functional fragments of camelid heavy chain antibodies, also called Nanobodies. The therapeutic potential of anti-rabies VHH was examined in a mouse model using intranasal challenge with a lethal dose of rabies virus. Anti-rabies VHH were administered directly into the brain or systemically, by intraperitoneal injection, 24 hours after virus challenge. Anti-rabies VHH were able to significantly prolong survival or even completely rescue mice from disease. The therapeutic effect depended on the dose, affinity and brain and plasma half-life of the VHH construct. Increasing the affinity by combining two VHH with a glycine-serine linker into bivalent or biparatopic constructs, increased the neutralizing potency to the picomolar range. Upon direct intracerebral administration, a dose as low as 33 µg of the biparatopic Rab-E8/H7 was still able to establish an anti-rabies effect. The effect of systemic treatment was significantly improved by increasing the half-life of Rab-E8/H7 through linkage with a third VHH targeted against albumin. Intraperitoneal treatment with 1.5 mg (2505 IU, 1 ml) of anti-albumin Rab-E8/H7 prolonged the median survival time from 9 to 15 days and completely rescued 43% of mice. For comparison, intraperitoneal treatment with the highest available dose of human anti-rabies immunoglobulins (65 mg, 111 IU, 1 ml) only prolonged survival by 2 days, without rescue. Overall, the therapeutic benefit seemed well correlated with the time of brain exposure and the plasma half-life of the used VHH construct. These results, together with the ease-of-production and superior thermal stability, render anti-rabies VHH into valuable candidates for development of alternative post exposure treatment drugs against rabies.


PLOS ONE | 2015

Kinetics of Respiratory Syncytial Virus (RSV) Memphis Strain 37 (M37) Infection in the Respiratory Tract of Newborn Lambs as an RSV Infection Model for Human Infants

Alejandro Larios Mora; Laurent Detalle; Albert G. van Geelen; Michael S. Davis; Thomas Stohr; Jack M. Gallup; Mark R. Ackermann

Rationale Respiratory syncytial virus (RSV) infection in preterm and newborn infants can result in severe bronchiolitis and hospitalization. The lamb lung has several key features conducive to modeling RSV infection in human infants, including susceptibility to human strains of RSV such as the A2, Long, and Memphis Strain 37 (M37). In this study, the kinetics of M37 infection was investigated in newborn lambs in order to better define clinical, viral, physiological, and immunological parameters as well as the pathology and lesions. Methods Newborn lambs were nebulized with M37 hRSV (6 mL of 1.27 x 107 FFU/mL), monitored daily for clinical responses, and respiratory tissues were collected from groups of lambs at days 1, 3, 4, 6, and 8 post-inoculation for the assessment of viral replication parameters, lesions and also cellular, immunologic and inflammatory responses. Results Lambs had increased expiratory effort (forced expiration) at days 4, 6, and 8 post-inoculation. Nasal wash lacked RSV titers at day 1, but titers were present at low levels at days 3 (peak), 4, and 8. Viral titers in bronchoalveolar lavage fluid (BALF) reached a plateau at day 3 (4.6 Log10 FFU/mL), which was maintained until day 6 (4.83 Log10 FFU/mL), and were markedly reduced or absent at day 8. Viral RNA levels (detected by RT-qPCR) in BALF were indistinguishable at days 3 (6.22 ± 0.08 Log10 M37 RNA copies/mL; mean ± se) and 4 (6.20 ± 0.16 Log10 M37 RNA copies/mL; mean ± se) and increased slightly on day 6 (7.15 ± 0.2 Log10 M37 RNA copies/mL; mean ± se). Viral antigen in lung tissue as detected by immunohistochemistry was not seen at day 1, was present at days 3 and 4 before reaching a peak by day 6, and was markedly reduced by day 8. Viral antigen was mainly present in airways (bronchi, bronchioles) at day 3 and was increasingly present in alveolar cells at days 4 and 6, with reduction at day 8. Histopathologic lesions such as bronchitis/bronchiolitis, epithelial necrosis and hyperplasia, peribronchial lymphocyte infiltration, and syncytial cells, were consistent with those described previously for lambs and infants. Conclusion This work demonstrates that M37 hRSV replication in the lower airways of newborn lambs is robust with peak replication on day 3 and sustained until day 6. These findings, along with the similarities of lamb lung to those of infants in terms of alveolar development, airway branching and epithelium, susceptibility to human RSV strains, lesion characteristics (bronchiolitis), lung size, clinical parameters, and immunity, further establish the neonatal lamb as a model with key features that mimic RSV infection in infants.


Journal of Biological Chemistry | 2016

High Throughput Combinatorial Formatting of PcrV Nanobodies for Efficient Potency Improvement.

Evelyn De Tavernier; Laurent Detalle; Erika Morizzo; Annelies Roobrouck; Severine De Taeye; Melanie Rieger; Tom Verhaeghe; Andreia Correia; Rob van Hegelsom; Rita Figueiredo; Jeroen Noens; Soren Steffensen; Thomas Stohr; Willem Van de Velde; Erik Depla; Bruno Dombrecht

Improving potencies through concomitant blockage of multiple epitopes and avid binding by fusing multiple (different) monovalent Nanobody building blocks via linker sequences into one multivalent polypeptide chain is an elegant alternative to affinity maturation. We explored a large and random formatting library of bivalent (combinations of two identical) and biparatopic (combinations of two different) Nanobodies for functional blockade of Pseudomonas aeruginosa PcrV. PcrV is an essential part of the P. aeruginosa type III secretion system (T3SS), and its oligomeric nature allows for multiple complex binding and blocking options. The library screening yielded a large number of promising biparatopic lead candidates, revealing significant (and non-trivial) preferences in terms of Nanobody building block and epitope bin combinations and orientations. Excellent potencies were confirmed upon further characterization in two different P. aeruginosa T3SS-mediated cytotoxicity assays. Three biparatopic Nanobodies were evaluated in a lethal mouse P. aeruginosa challenge pneumonia model, conferring 100% survival upon prophylactic administration and reducing lung P. aeruginosa burden by up to 2 logs. At very low doses, they protected the mice from P. aeruginosa infection-related changes in lung histology, myeloperoxidase production, and lung weight. Importantly, the most potent Nanobody still conferred protection after therapeutic administration up to 24 h post-infection. The concept of screening such formatting libraries for potency improvement is applicable to other targets and biological therapeutic platforms.


mAbs | 2018

Delivery of ALX-0171 by inhalation greatly reduces respiratory syncytial virus disease in newborn lambs

Alejandro Larios Mora; Laurent Detalle; Jack M. Gallup; Albert G. van Geelen; Thomas Stohr; Linde Duprez; Mark R. Ackermann

ABSTRACT Respiratory syncytial virus (RSV) is a common cause of acute lower respiratory disease in infants and young children worldwide. Currently, treatment is supportive and no vaccines are available. The use of newborn lambs to model hRSV infection in human infants may provide a valuable tool to assess safety and efficacy of new antiviral drugs and vaccines. ALX-0171 is a trivalent Nanobody targeting the hRSV fusion (F) protein and its therapeutic potential was evaluated in newborn lambs infected with a human strain of RSV followed by daily ALX-0171 nebulization for 3 or 5 consecutive days. Colostrum-deprived newborn lambs were infected with hRSV-M37 before being treated by daily nebulization with either ALX-0171 or placebo. Two different treatment regimens were examined: day 1–5 or day 3–5 post-infection. Lambs were monitored daily for general well-being and clinical parameters. Respiratory tissues and bronchoalveolar lavage fluid were collected at day 6 post-inoculation for the quantification of viral lesions, lung viral titers, viral antigen and lung histopathology. Administration by inhalation of ALX-0171 was well-tolerated in these hRSV-infected newborn lambs. Robust antiviral effects and positive effects on hRSV-induced lung lesions and reduction in symptoms of illness were noted. These effects were still apparent when treatment start was delayed and coincided with peak viral loads (day 3 post-infection) and at a time point when signs of RSV disease were apparent. The latter design is expected to have high translational value for planned clinical trials. These results are indicative of the therapeutic potential of ALX-0171 in infants.


Translational Medicine#R##N#Tools and Techniques | 2016

New Developments in the Use of Biomarkers in Translational Medicine

Benedikte Serruys; Thomas Stohr; Hans Ulrichts; Katrien Vanheusden; Maarten Van Roy

In recent years there has been an explosion of published research on biomarkers yet with limited translation into clinical practice and back to bench. Classically, biomarker experiments involved the assessment of proteins in blood. Many developments in biomarker research have been performed in the past years that make biomarkers an extremely valuable tool in translational medicine. The goal of this chapter is to highlight a number of recent developments that advanced the field and may help to value the sophisticated use of biomarkers in biomedical research.


Cancer Research | 2013

Abstract LB-316: The anti-c-Met Nanobody®, a novel and promising anti-cancer therapeutic.

Tinneke Denayer; Thomas Stohr; Gerald Beste; Ann Brige; Cedric Jozef Neotere Ververken; Josefin-Beate Holz

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC The hepatocyte growth factor (HGF) and its receptor, c-Met, are implicated in many human cancers to promote tumor proliferation, migration, invasion and angiogenesis. Dysregulation of the HGF/c-Met pathway is known to correlate with poor prognosis. In recent years, multiple anti-cancer agents have been developed interfering at different levels in the HGF/c-Met pathway. Here, we investigated the in vitro and in vivo efficacy of a novel Nanobody effectively antagonizing c-Met. Nanobodies are therapeutic proteins based on the smallest functional fragments of heavy chain antibodies, naturally occurring in Camelidae. The Nanobody platform allows designing modular drugs, including multivalent, biparatopic and bifunctional molecules. The anti-c-Met Nanobody is a bispecific Nanobody that comprises two building blocks, one targeting c-Met and one binding human serum albumin for half-life extension purposes. In vitro characterization revealed that the Nanobody is able to specifically and completely inhibit HGF/c-Met interaction with high potency in an ELISA-based assay. In addition, the anti-c-Met Nanobody was able to completely block the HGF-dependent c-Met-phosphorylation in the A549 non-small cell lung cancer cell line. Importantly, no agonistic activity was observed in absence of HGF, as the Nanobody was designed to have a monovalent interaction with c-Met. Next, the in vivo effect was translated towards a potent in vivo effect. The anti-tumor efficacy of the anti-c-Met Nanobody was assessed in xenograft mouse models in which autocrine U87MG (HGF+, c-Met+) glioblastoma cells, or autocrine KP4 (HGF+, c-Met+) pancreatic carcinoma cells were subcutaneously injected. In the U87MG xenograft model, Nanobody treatment (10 mg/kg i.p 3x/week) resulted in significant tumor growth inhibition compared to vehicle and sustained tumor growth suppression after cessation of the treatment. Treatment with the anti-c-Met Nanobody in the KP4 xenograft model (10 mg/kg i.p 3x/week) led to a significant tumor growth inhibition and even tumor regression. In the latter model, serum levels of soluble c-Met, IL-8 and HGF were shown to be significantly reduced upon Nanobody treatment, which could make these markers attractive as potential translational pharmacodynamics biomarkers. Furthermore, the ratio of tyrosine phosphorylated c-Met over total c-Met levels was significantly lower in KP4 tumors treated with Nanobody versus vehicle. It could be postulated based on literature data that the anti-c-Met Nanobody, by possessing an anti-albumin Nanobody building block, has the potential to show superior tumor penetration than c-Met targeting antibodies. Hence, the anti-c-Met Nanobody might be a valuable novel biological for the treatment of cancer driven by HGF-c-Met signaling. Citation Format: Tinneke Denayer, Thomas Stohr, Gerald Beste, Ann Brige, Cedric Ververken, Josefin-Beate Holz. The anti-c-Met Nanobody®, a novel and promising anti-cancer therapeutic. [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 LB-316. doi:10.1158/1538-7445.AM2013-LB-316


New Horizons in Translational Medicine | 2014

Animal models in translational medicine: Validation and prediction

Tinneke Denayer; Thomas Stohr; Maarten Van Roy


Archive | 2017

baboon model of acquired thrombotic thrombocytopenic purpura Short title: ALX-0681 treatment of acquired TTP in baboons Authors

Filip Callewaert; Jan Roodt; Hans Ulrichts; Thomas Stohr; Walter J. Janse van Rensburg; Seb Lamprecht; Stefaan Rossenu; Sofie Priem; Wouter Willems; Josefin-Beate Holz

Collaboration


Dive into the Thomas Stohr's collaboration.

Researchain Logo
Decentralizing Knowledge