Network


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

Hotspot


Dive into the research topics where Matthias Van Woensel is active.

Publication


Featured researches published by Matthias Van Woensel.


Cancers | 2013

Formulations for Intranasal Delivery of Pharmacological Agents to Combat Brain Disease: A New Opportunity to Tackle GBM?

Matthias Van Woensel; Nathalie Wauthoz; Rémi Rosiere; Karim Amighi; Véronique Mathieu; Florence Lefranc; Stefaan Van Gool; Steven De Vleeschouwer

Despite recent advances in tumor imaging and chemoradiotherapy, the median overall survival of patients diagnosed with glioblastoma multiforme does not exceed 15 months. Infiltration of glioma cells into the brain parenchyma, and the blood-brain barrier are important hurdles to further increase the efficacy of classic therapeutic tools. Local administration methods of therapeutic agents, such as convection enhanced delivery and intracerebral injections, are often associated with adverse events. The intranasal pathway has been proposed as a non-invasive alternative route to deliver therapeutics to the brain. This route will bypass the blood-brain barrier and limit systemic side effects. Upon presentation at the nasal cavity, pharmacological agents reach the brain via the olfactory and trigeminal nerves. Recently, formulations have been developed to further enhance this nose-to-brain transport, mainly with the use of nanoparticles. In this review, the focus will be on formulations of pharmacological agents, which increase the nasal permeation of hydrophilic agents to the brain, improve delivery at a constant and slow release rate, protect therapeutics from degradation along the pathway, increase mucoadhesion, and facilitate overall nasal transport. A mounting body of evidence is accumulating that the underexplored intranasal delivery route might represent a major breakthrough to combat glioblastoma.


Frontiers in Immunology | 2016

Exploiting the Immunogenic Potential of Cancer Cells for Improved Dendritic Cell Vaccines.

Lien Vandenberk; Jochen Belmans; Matthias Van Woensel; Matteo Riva; Stefaan Van Gool

Cancer immunotherapy is currently the hottest topic in the oncology field, owing predominantly to the discovery of immune checkpoint blockers. These promising antibodies and their attractive combinatorial features have initiated the revival of other effective immunotherapies, such as dendritic cell (DC) vaccinations. Although DC-based immunotherapy can induce objective clinical and immunological responses in several tumor types, the immunogenic potential of this monotherapy is still considered suboptimal. Hence, focus should be directed on potentiating its immunogenicity by making step-by-step protocol innovations to obtain next-generation Th1-driving DC vaccines. We review some of the latest developments in the DC vaccination field, with a special emphasis on strategies that are applied to obtain a highly immunogenic tumor cell cargo to load and to activate the DCs. To this end, we discuss the effects of three immunogenic treatment modalities (ultraviolet light, oxidizing treatments, and heat shock) and five potent inducers of immunogenic cell death [radiotherapy, shikonin, high-hydrostatic pressure, oncolytic viruses, and (hypericin-based) photodynamic therapy] on DC biology and their application in DC-based immunotherapy in preclinical as well as clinical settings.


Journal of Controlled Release | 2016

Development of siRNA-loaded chitosan nanoparticles targeting Galectin-1 for the treatment of glioblastoma multiforme via intranasal administration

Matthias Van Woensel; Nathalie Wauthoz; Rémi Rosiere; Véronique Mathieu; Robert Kiss; Florence Lefranc; Brecht Steelant; Ellen Dilissen; Stefaan Van Gool; Thomas Mathivet; Holger Gerhardt; Karim Amighi; Steven De Vleeschouwer

Galectin-1 (Gal-1) is a naturally occurring galactose-binding lectin, which is overexpressed in glioblastoma multiforme (GBM). Gal-1 is associated with tumor progression, and is a potent immune suppressor in the tumor micro-environment. To inhibit Gal-1 in GBM, an effective therapy is required that reaches the central nervous system tumor, with limited systemic effects. In this study, we report for the first time that concentrated chitosan nanoparticle suspensions can deliver small interfering RNA (siRNA) into the central nervous system tumor within hours after intranasal administration. These nanoparticles are able to complex siRNA targeting Gal-1 to a high percentage, and protect them from RNAse degradation. Moreover, a successful intracellular delivery of anti-Gal-1 siRNA resulted in a decreased expression of Gal-1 in both murine and human GBM cells. Sequence specific RNAinterference, resulted in more than 50% Gal-1 reduction in tumor bearing mice. This study indicates that the intranasal pathway is an underexplored transport route for delivering siRNA-based therapies targeting Gal-1 in the treatment of GBM.


Scientific Reports | 2017

Sensitization of glioblastoma tumor micro-environment to chemo- and immunotherapy by Galectin-1 intranasal knock-down strategy

Matthias Van Woensel; Thomas Mathivet; Nathalie Wauthoz; Rémi Rosiere; Abhishek D. Garg; Patrizia Agostinis; Véronique Mathieu; Robert Kiss; Florence Lefranc; Louis Boon; Jochen Belmans; Stefaan Van Gool; Holger Gerhardt; Karim Amighi; Steven De Vleeschouwer

In this study, we evaluated the consequences of reducing Galectin-1 (Gal-1) in the tumor micro-environment (TME) of glioblastoma multiforme (GBM), via nose-to-brain transport. Gal-1 is overexpressed in GBM and drives chemo- and immunotherapy resistance. To promote nose-to-brain transport, we designed siRNA targeting Gal-1 (siGal-1) loaded chitosan nanoparticles that silence Gal-1 in the TME. Intranasal siGal-1 delivery induces a remarkable switch in the TME composition, with reduced myeloid suppressor cells and regulatory T cells, and increased CD4+ and CD8+ T cells. Gal-1 knock-down reduces macrophages’ polarization switch from M1 (pro-inflammatory) to M2 (anti-inflammatory) during GBM progression. These changes are accompanied by normalization of the tumor vasculature and increased survival for tumor bearing mice. The combination of siGal-1 treatment with temozolomide or immunotherapy (dendritic cell vaccination and PD-1 blocking) displays synergistic effects, increasing the survival of tumor bearing mice. Moreover, we could confirm the role of Gal-1 on lymphocytes in GBM patients by matching the Gal-1 expression and their T cell signatures. These findings indicate that intranasal siGal-1 nanoparticle delivery could be a valuable adjuvant treatment to increase the efficiency of immune-checkpoint blockade and chemotherapy.


OncoImmunology | 2017

Preclinical efficacy of immune-checkpoint monotherapy does not recapitulate corresponding biomarkers-based clinical predictions in glioblastoma

Abhishek D. Garg; Lien Vandenberk; Matthias Van Woensel; Jochen Belmans; Marco B.E. Schaaf; Louis Boon; Steven De Vleeschouwer; Patrizia Agostinis

ABSTRACT Glioblastoma (GBM) is resistant to most multimodal therapies. Clinical success of immune-checkpoint inhibitors (ICIs) has spurred interest in applying ICIs targeting CTLA4, PD1 or IDO1 against GBM. This amplifies the need to ascertain GBMs intrinsic susceptibility (or resistance) toward these ICIs, through clinical biomarkers that may also “guide and prioritize” preclinical testing. Here, we interrogated the TCGA and/or REMBRANDT human patient-cohorts to predict GBMs predisposition toward ICIs. We exploited various broad clinical biomarkers, including mutational or predicted-neoantigen burden, pre-existing or basal levels of tumor-infiltrating T lymphocytes (TILs), differential expression of immune-checkpoints within the tumor and their correlation with particular TILs/Treg-associated functional signature and prognostic impact of differential immune-checkpoint expression. Based on these analyses, we found that predictive biomarkers of ICI responsiveness exhibited inconsistent patterns in GBM patients, i.e., they either predicted ICI resistance (as compared with typical ICI-responsive cancer-types like melanoma, lung cancer or bladder cancer) or susceptibility to therapeutic targeting of CTLA4 or IDO1. On the other hand, our comprehensive literature meta-analysis and preclinical testing of ICIs using an orthotopic GL261-glioma mice model, indicated significant antitumor properties of anti-PD1 antibody, whereas blockade of IDO1 or CTLA4 either failed or provided very marginal advantage. These trends raise the need to better assess the applicability of ICIs and associated biomarkers for GBM.


International Journal of Pharmaceutics | 2016

Development and evaluation of well-tolerated and tumor-penetrating polymeric micelle-based dry powders for inhaled anti-cancer chemotherapy.

Rémi Rosiere; Matthias Van Woensel; Véronique Mathieu; Ingrid Langer; Thomas Mathivet; Marjorie Vermeersch; Karim Amighi; Nathalie Wauthoz

Despite the direct access to the lung offered by the inhalation route, drug penetration into lung tumors could remain an important issue. In this study, folate-polyethylene glycol-hydrophobically-modified dextran (F-PEG-HMD) micelles were developed as an effective pulmonary drug delivery system to reach and penetrate lung tumors and cancer cells. The F-PEG-HMD micelles were able to enter HeLa and M109-HiFR, two folate receptor-expressing cancer cell lines, in vitro, and in vivo after administration by inhalation to orthotopic M109-HiFR lung tumor grafted mice. Paclitaxel-loaded F-PEG-HMD micelles characterized in PBS by a Z-average diameter of ∼50 nm and a zeta potential of ∼-4 mV were prepared with an encapsulation efficiency of ∼100%. The loaded micelles reduced HeLa and M109-HiFR cell growth, with half maximal inhibitory concentrations of 37 and 150 nM, respectively. Dry powders embedding the paclitaxel-loaded F-PEG-HMD micelles were developed by spray-drying. In vitro, good deposition profiles were obtained, with a fine particle fraction of up to 50% and good ability to re-disperse the micelles in physiological buffer. A polymeric micelle-based dry powder without paclitaxel was well-tolerated in vivo, as assessed in healthy mice by determination of total protein content, cell count, and cytokine IL-1β, IL-6, and TNF-α concentrations in bronchoalveolar lavage fluids.


The Journal of Allergy and Clinical Immunology | 2017

Histamine and T helper cytokine–driven epithelial barrier dysfunction in allergic rhinitis

Brecht Steelant; Sven Seys; Laura Van Gerven; Matthias Van Woensel; Ricard Farré; Paulina Wawrzyniak; Inge Kortekaas Krohn; Dominique Bullens; Karel Talavera; Ulrike Raap; Louis Boon; Cezmi A. Akdis; Guy E. Boeckxstaens; Jan Ceuppens; Peter Hellings

Background: Allergic rhinitis (AR) is characterized by mucosal inflammation, driven by activated immune cells. Mast cells and TH2 cells might decrease epithelial barrier integrity in AR, maintaining a leaky epithelial barrier. Objective: We sought to investigate the role of histamine and TH2 cells in driving epithelial barrier dysfunction in AR. Methods: Air‐liquid interface cultures of primary nasal epithelial cells were used to measure transepithelial electrical resistance, paracellular flux of fluorescein isothiocyanate‐dextran 4 kDa, and mRNA expression of tight junctions. Nasal secretions were collected from healthy control subjects, AR patients, and idiopathic rhinitis patients and were tested in vitro. In addition, the effect of activated TH1 and TH2 cells, mast cells, and neurons was tested in vitro. The effect of IL‐4, IL‐13, IFN‐&ggr;, and TNF‐&agr; on mucosal permeability was tested in vivo. Results: Histamine as well as nasal secretions of AR but not idiopathic rhinitis patients rapidly decreased epithelial barrier integrity in vitro. Pretreatment with histamine receptor‐1 antagonist, azelastine prevented the early effect of nasal secretions of AR patients on epithelial integrity. Supernatant of activated TH1 and TH2 cells impaired epithelial integrity, while treatment with anti‐TNF‐&agr; or anti‐IL‐4R&agr; monoclonal antibodies restored the TH1‐ and TH2‐induced epithelial barrier dysfunction, respectively. IL‐4, IFN‐&ggr;, and TNF‐&agr; enhanced mucosal permeability in mice. Antagonizing IL‐4 prevented mucosal barrier disruption and tight junction downregulation in a mouse model of house dust mite allergic airway inflammation. Conclusions: Our data indicate a key role for allergic inflammatory mediators in modulating nasal epithelial barrier integrity in the pathophysiology in AR.


Embo Molecular Medicine | 2017

Dynamic stroma reorganization drives blood vessel dysmorphia during glioma growth

Thomas Mathivet; Claire Bouleti; Matthias Van Woensel; Fabio Stanchi; Tina Verschuere; Li-Kun Phng; Joost Dejaegher; Marly Balcer; Ken Matsumoto; Petya B Georgieva; Jochen Belmans; Raf Sciot; Christian Stockmann; Massimiliano Mazzone; Steven De Vleeschouwer; Holger Gerhardt

Glioma growth and progression are characterized by abundant development of blood vessels that are highly aberrant and poorly functional, with detrimental consequences for drug delivery efficacy. The mechanisms driving this vessel dysmorphia during tumor progression are poorly understood. Using longitudinal intravital imaging in a mouse glioma model, we identify that dynamic sprouting and functional morphogenesis of a highly branched vessel network characterize the initial tumor growth, dramatically changing to vessel expansion, leakage, and loss of branching complexity in the later stages. This vascular phenotype transition was accompanied by recruitment of predominantly pro‐inflammatory M1‐like macrophages in the early stages, followed by in situ repolarization to M2‐like macrophages, which produced VEGF‐A and relocate to perivascular areas. A similar enrichment and perivascular accumulation of M2 versus M1 macrophages correlated with vessel dilation and malignancy in human glioma samples of different WHO malignancy grade. Targeting macrophages using anti‐CSF1 treatment restored normal blood vessel patterning and function. Combination treatment with chemotherapy showed survival benefit, suggesting that targeting macrophages as the key driver of blood vessel dysmorphia in glioma progression presents opportunities to improve efficacy of chemotherapeutic agents. We propose that vessel dysfunction is not simply a general feature of tumor vessel formation, but rather an emergent property resulting from a dynamic and functional reorganization of the tumor stroma and its angiogenic influences.


Stem Cells Translational Medicine | 2016

Clinical‐Grade Human Multipotent Adult Progenitor Cells Block CD8+ Cytotoxic T Lymphocytes

Jeroen Plessers; Emily Dekimpe; Matthias Van Woensel; Valerie D. Roobrouck; Dominique Bullens; Jef Pinxteren; Catherine M. Verfaillie; Stefaan Van Gool

MultiStem cells are clinical‐grade multipotent adult bone marrow‐derived progenitor cells (MAPCs), with extensive replication potential and broader differentiation capacity compared with mesenchymal stem cells. Human MAPCs suppress T‐cell proliferation induced by alloantigens and mutually interact with allogeneic natural killer cells. In this study, the interaction between MultiStem and CD8+ cytotoxic T lymphocytes (CTLs) was addressed for the first time. In an in vitro setting, the immunogenicity of MultiStem, the susceptibility of MultiStem toward CTL‐mediated lysis, and its effects on CTL function were investigated. MultiStem was nonimmunogenic for alloreactive CTL induction and was—even after major histocompatibility complex class I upregulation—insensitive to alloantigen‐specific CTL‐mediated lysis. Furthermore, MultiStem reduced CTL proliferation and significantly decreased perforin expression during the T‐cell activation phase. As a consequence, MultiStem dose‐dependently impaired the induction of CTL function. These effects of MultiStem were mediated predominantly through contact‐dependent mechanisms. Moreover, MultiStem cells considerably influenced the expression of T‐cell activation markers CD25, CD69, and human leukocyte antigen‐DR. The MultiStem‐induced CD8−CD69+ T‐cell population displayed a suppressive effect on the induction of CTL function during a subsequent mixed‐lymphocyte culture. Finally, the killer activity of activated antigen‐specific CTLs during their cytolytic effector phase was also diminished in the presence of MultiStem. This study confirms that these clinical‐grade MAPCs are an immune‐modulating population that inhibits CTL activation and effector responses and are, consequently, a highly valuable cell population for adoptive immunosuppressive therapy in diseases where damage is induced by CTLs.


Molecular Pharmaceutics | 2018

New Folate-Grafted Chitosan Derivative To Improve Delivery of Paclitaxel-Loaded Solid Lipid Nanoparticles for Lung Tumor Therapy by Inhalation

Rémi Rosiere; Matthias Van Woensel; Michel Gelbcke; Véronique Mathieu; Julien Hecq; Thomas Mathivet; Marjorie Vermeersch; Pierre Van Antwerpen; Karim Amighi; Nathalie Wauthoz

Inhaled chemotherapy for the treatment of lung tumors requires that drug delivery systems improve selectivity for cancer cells and tumor penetration and allow sufficient lung residence. To this end, we developed solid lipid nanoparticles (SLN) with modified surface properties. We successfully synthesized a new folate-grafted copolymer of polyethylene glycol (PEG) and chitosan, F-PEG-HTCC, with a PEG-graft ratio of 7% and a molecular weight range of 211-250 kDa. F-PEG-HTCC-coated, paclitaxel-loaded SLN were prepared with an encapsulation efficiency, mean diameter, and zeta potential of about 100%, 250 nm, and +32 mV, respectively. The coated SLN entered folate receptor (FR)-expressing HeLa and M109-HiFR cells in vitro and M109 tumors in vivo after pulmonary delivery. The coated SLN significantly decreased the in vitro half-maximum inhibitory concentrations of paclitaxel in M109-HiFR cells (60 vs 340 nM, respectively). We demonstrated that FR was involved in these improvements, especially in M109-HiFR cells. After pulmonary delivery in vivo, the coated SLN had a favorable pharmacokinetic profile, with pulmonary exposure to paclitaxel prolonged to up to 6 h and limited systemic distribution. Our preclinical findings therefore demonstrated the positive impact of the coated SLN on the delivery of paclitaxel by inhalation.

Collaboration


Dive into the Matthias Van Woensel's collaboration.

Top Co-Authors

Avatar

Stefaan Van Gool

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Véronique Mathieu

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Karim Amighi

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Steven De Vleeschouwer

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Nathalie Wauthoz

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Rémi Rosiere

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Thomas Mathivet

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Florence Lefranc

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jochen Belmans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Holger Gerhardt

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge