Network


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

Hotspot


Dive into the research topics where Didier Colau is active.

Publication


Featured researches published by Didier Colau.


Immunity | 2008

Restoring the Association of the T Cell Receptor with CD8 Reverses Anergy in Human Tumor-Infiltrating Lymphocytes

Nathalie Demotte; Vincent Stroobant; Pierre J. Courtoy; Patrick Van Der Smissen; Didier Colau; Immanuel F. Luescher; Claire Hivroz; Julie Nicaise; Jean-Luc Squifflet; Michel Mourad; Danièle Godelaine; Thierry Boon; Pierre van der Bruggen

For several days after antigenic stimulation, human cytolytic T lymphocyte (CTL) clones exhibit a decrease in their effector activity and in their binding to human leukocyte antigen (HLA)-peptide tetramers. We observed that, when in this state, CTLs lose the colocalization of the T cell receptor (TCR) and CD8. Effector function and TCR-CD8 colocalization were restored with galectin disaccharide ligands, suggesting that the binding of TCR to galectin plays a role in the distancing of TCR from CD8. These findings appear to be applicable in vivo, as TCR was observed to be distant from CD8 on human tumor-infiltrating lymphocytes, which were anergic. These lymphocytes recovered effector functions and TCR-CD8 colocalization after ex vivo treatment with galectin disaccharide ligands. The separation of TCR and CD8 molecules could be one major mechanism of anergy in tumors and other chronic stimulation conditions.


Journal of Immunology | 2007

Comparative Prime-Boost Vaccinations Using Semliki Forest Virus, Adenovirus, and ALVAC Vectors Demonstrate Differences in the Generation of a Protective Central Memory CTL Response against the P815 Tumor

Tanja I. Näslund; Catherine Uyttenhove; Eva Nordström; Didier Colau; Guy Warnier; Mikael Jondal; Benoı̂t J. Van den Eynde; Peter Liljeström

Tumor-specific Ags are potential target molecules in the therapeutic treatment of cancer. One way to elicit potent immune responses against these Ags is to use recombinant viruses, which activate both the innate and the adaptive arms of the immune system. In this study, we have compared Semliki Forest virus (SFV), adenovirus, and ALVAC (poxvirus) vectors for their capacity to induce CD8+ T cell responses against the P1A tumor Ag and to elicit protection against subsequent challenge injection of P1A-expressing P815 tumor cells in DBA/2 mice. Both homologous and heterologous prime-boost regimens were studied. In most cases, both higher CD8+ T cell responses and better tumor protections were observed in mice immunized with heterologous prime-boost regimens, suggesting that the combination of different viral vectors is beneficial for the induction of an effective immune response. However, homologous immunization with SFV provided potent tumor protection despite a rather moderate primary CD8+ T cell response as compared with mice immunized with recombinant adenovirus. SFV-immunized mice showed a rapid and more extensive expansion of P1A-specific CD8+ T cells in the tumor-draining lymph node after tumor challenge and had a higher frequency of CD62L+ P1A-specific T cells in the blood, spleen, and lymph nodes as compared with adenoimmunized mice. Our results indicate that not only the magnitude but in particular the quality of the CD8+ T cell response correlates with tumor protection.


Blood | 2009

Peripheral blood lymphocytes genetically modified to express the self/tumor antigen MAGE-A3 induce antitumor immune responses in cancer patients

Raffaella Fontana; Marco Bregni; Arcadi Cipponi; Laura Raccosta; Cristina Rainelli; Daniela Maggioni; Francesca Lunghi; Fabio Ciceri; Sylvain Mukenge; Claudio Doglioni; Didier Colau; Pierre G. Coulie; Claudio Bordignon; Catia Traversari; Vincenzo Russo

Dendritic cell (DC) targeting in vivo has recently been shown to confer strong and protective cytotoxic T lymphocyte (CTL)-based immunity in tumor murine models. Our group has recently demonstrated in preclinical models that the infusion of genetically modified lymphocytes (GMLs) expressing the self/tumor antigen TRP-2 is able to elicit functional TRP-2-specific effectors with antitumor activity by targeting DCs in vivo. Here we have analyzed vaccine- and tumor-specific immune responses of 10 melanoma patients treated with autologous GMLs expressing the cancer germline gene MAGE-A3. Three of 10 patients treated with MAGE-A3-GML showed an increase of circulating anti-MAGE-A3 T cells, and developed skin delayed-type hypersensitivity to MAGE-A3. Interestingly, in 2 of these patients, with progressive and measurable tumors at study entry, anti-MAGE-A3 T cells were detected not only in the blood but also within tumors resected after vaccination. These results demonstrate that the infusion of MAGE-A3-GML elicits antitumor T cells, which are capable of trafficking to inflamed tissues and of infiltrating tumors. Clinical studies on a larger group of patients are needed to evaluate the clinical efficacy of such a strategy.


Journal of Immunology | 2012

Analysis of the Processing of Seven Human Tumor Antigens by Intermediate Proteasomes

Benoît Guillaume; Vincent Stroobant; Marie-Pierre Bousquet-Dubouch; Didier Colau; Jacques Chapiro; Nicolas Parmentier; Alexandre Dalet; Benoît Van den Eynde

We recently described two proteasome subtypes that are intermediate between the standard proteasome and the immunoproteasome. They contain only one (β5i) or two (β1i and β5i) of the three inducible catalytic subunits of the immunoproteasome. They are present in tumor cells and abundant in normal human tissues. We described two tumor antigenic peptides that are uniquely produced by these intermediate proteasomes. In this work, we studied the production by intermediate proteasomes of tumor antigenic peptides known to be produced exclusively by the immunoproteasome (MAGE-A3114–122, MAGE-C242–50, MAGE-C2336–344) or the standard proteasome (Melan-A26–35, tyrosinase369–377, gp100209–217). We observed that intermediate proteasomes efficiently produced the former peptides, but not the latter. Two peptides from the first group were equally produced by both intermediate proteasomes, whereas MAGE-C2336–344 was only produced by intermediate proteasome β1i-β5i. Those results explain the recognition of tumor cells devoid of immunoproteasome by CTL recognizing peptides not produced by the standard proteasome. We also describe a third antigenic peptide that is produced exclusively by an intermediate proteasome: peptide MAGE-C2191–200 is produced only by intermediate proteasome β1i-β5i. Analyzing in vitro digests, we observed that the lack of production by a given proteasome usually results from destruction of the antigenic peptide by internal cleavage. Interestingly, we observed that the immunoproteasome and the intermediate proteasomes fail to cleave between hydrophobic residues, despite a higher chymotrypsin-like activity measured on fluorogenic substrates. Altogether, our results indicate that the repertoire of peptides produced by intermediate proteasomes largely matches the repertoire produced by the immunoproteasome, but also contains additional peptides.


European Journal of Medicinal Chemistry | 2014

Detailed analysis and follow-up studies of a high-throughput screening for indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors

Ute F. Röhrig; Somi Reddy Majjigapu; Marc Chambon; Sylvian Bron; Luc Pilotte; Didier Colau; Benoît Van den Eynde; Gerardo Turcatti; Pierre Vogel; Vincent Zoete; Olivier Michielin

Indoleamine 2,3-dioxygenase 1 (IDO1) is a key regulator of immune responses and therefore an important therapeutic target for the treatment of diseases that involve pathological immune escape, such as cancer. Here, we describe a robust and sensitive high-throughput screen (HTS) for IDO1 inhibitors using the Prestwick Chemical Library of 1200 FDA-approved drugs and the Maybridge HitFinder Collection of 14,000 small molecules. Of the 60 hits selected for follow-up studies, 14 displayed IC50 values below 20 μM under the secondary assay conditions, and 4 showed an activity in cellular tests. In view of the high attrition rate we used both experimental and computational techniques to identify and to characterize compounds inhibiting IDO1 through unspecific inhibition mechanisms such as chemical reactivity, redox cycling, or aggregation. One specific IDO1 inhibitor scaffold, the imidazole antifungal agents, was chosen for rational structure-based lead optimization, which led to more soluble and smaller compounds with micromolar activity.


Bioorganic & Medicinal Chemistry | 2011

Indol-2-yl ethanones as novel indoleamine 2,3-dioxygenase (IDO) inhibitors

Eduard Dolusic; Pierre Larrieu; Sébastien Blanc; Frédéric Sapunaric; Bernadette Norberg; Laurence Moineaux; Delphine Colette; Vincent Stroobant; Luc Pilotte; Didier Colau; Thierry Ferain; Graeme Fraser; Moreno Galleni; Jean-Marie Frère; Bernard Masereel; Benoît Van den Eynde; Johan Wouters; Raphaël Frédérick

Indoleamine 2,3-dioxygenase (IDO) is a heme dioxygenase which has been shown to be involved in the pathological immune escape of diseases such as cancer. The synthesis and structure-activity relationships (SAR) of a novel series of IDO inhibitors based on the indol-2-yl ethanone scaffold is described. In vitro and in vivo biological activities have been evaluated, leading to compounds with IC(50) values in the micromolar range in both tests. Introduction of small substituents in the 5- and 6-positions of the indole ring, indole N-methylation and variations of the aromatic side chain are all well tolerated. An iron coordinating group on the linker is a prerequisite for biological activity, thus corroborating the virtual screening results.


Journal of Immunology | 2012

Minimal Tolerance to a Tumor Antigen Encoded by a Cancer-Germline Gene

Ivo Huijbers; Saïdi M. Soudja; Catherine Uyttenhove; Michel Buferne; Else-Marit Inderberg-Suso; Didier Colau; Luc Pilotte; Céline Powis de Tenbossche; Patrick Chomez; Francis Brasseur; Anne-Marie Schmitt-Verhulst; Benoît Van den Eynde

Central tolerance toward tissue-restricted Ags is considered to rely on ectopic expression in the thymus, which was also observed for tumor Ags encoded by cancer-germline genes. It is unknown whether endogenous expression shapes the T cell repertoire against the latter Ags and explains their weak immunogenicity. We addressed this question using mouse cancer-germline gene P1A, which encodes antigenic peptide P1A35–43 presented by H-2Ld. We made P1A-knockout (P1A-KO) mice and asked whether their anti-P1A35–43 immune responses were stronger than those of wild-type mice and whether P1A-KO mice responded to other P1A epitopes, against which wild-type mice were tolerized. We observed that both types of mice mounted similar P1A35–43-specific CD8 T cell responses, although the frequency of P1A35–43-specific CD8 T cells generated in response to P1A-expressing tumors was slightly higher in P1A-KO mice. This higher reactivity allowed naive P1A-KO mice to reject spontaneously P1A-expressing tumors, which progressed in wild-type mice. TCR-Vβ usage of P1A35–43-specific CD8 cells was slightly modified in P1A-KO mice. Peptide P1A35–43 remained the only P1A epitope recognized by CD8 T cells in both types of mice, which also displayed similar thymic selection of a transgenic TCR recognizing P1A35–43. These results indicate the existence of a minimal tolerance to an Ag encoded by a cancer-germline gene and suggest that its endogenous expression only slightly affects diversification of the T cell repertoire against this Ag.


Cancer Immunology, Immunotherapy | 2006

Lack of tumor recognition by cytolytic T lymphocyte clones recognizing peptide 195-203 encoded by gene MAGE-A3 and presented by HLA-A24 molecules.

Tomoko So; Takeshi Hanagiri; Jacques Chapiro; Didier Colau; Francis Brasseur; Kosei Yasumoto; Thierry Boon; Pierre Coulie

Gene MAGE-A3 encodes tumor-specific antigenic peptides recognized by T cells on many tumors. MAGE-A3 peptides presented by HLA class I molecules have been identified using CD8 lymphocytes stimulated with cells that either expressed gene MAGE-A3 or were pulsed with candidate peptides. One antigen identified with the latter method is peptide MAGE-A3195–203 IMPKAGLLI, presented by HLA-A24 molecules. It has been used to vaccinate advanced cancer patients. Here, we have used HLA/peptide tetramers to detect T cells recognizing this peptide. Their frequency was estimated to be 2xa0×xa010−8 of the blood CD8 cells in non-cancerous HLA-A24+ individuals, which is tenfold lower than the reported frequencies of T cells against other MAGE peptides. In the blood of a patient vaccinated with MAGE-A3, the estimated frequency was 5xa0×xa010−7. Anti-MAGE-3.A24 cytolytic T cell clones were derived, that lysed peptide-pulsed cells with half-maximal effect at the low concentration of 500xa0pM. However, these CTL did not recognize a panel of HLA-A24+ tumor cells that expressed MAGE-A3 at levels similar to those found in HLA-A1+ tumor cells recognized by anti-MAGE-3.A1 CTLs. Furthermore, 293-EBNA cells transfected with MAGE-A3 and HLA-A24 constructs were hardly recognized by the anti-MAGE-3.A24 CTL clones. These results suggest that peptide MAGE-A3195–203 is poorly processed and is not an appropriate target for cancer immunotherapy.


Nature Communications | 2017

Resistance to cancer immunotherapy mediated by apoptosis of tumor-infiltrating lymphocytes.

Jingjing Zhu; Céline Powis de Tenbossche; Stefania Cané; Didier Colau; Nicolas van Baren; Christophe Lurquin; Anne-Marie Schmitt-Verhulst; Peter Liljeström; Catherine Uyttenhove; Benoît Van den Eynde

Despite impressive clinical success, cancer immunotherapy based on immune checkpoint blockade remains ineffective in many patients due to tumoral resistance. Here we use the autochthonous TiRP melanoma model, which recapitulates the tumoral resistance signature observed in human melanomas. TiRP tumors resist immunotherapy based on checkpoint blockade, cancer vaccines or adoptive T-cell therapy. TiRP tumors recruit and activate tumor-specific CD8+ T cells, but these cells then undergo apoptosis. This does not occur with isogenic transplanted tumors, which are rejected after adoptive T-cell therapy. Apoptosis of tumor-infiltrating lymphocytes can be prevented by interrupting the Fas/Fas-ligand axis, and is triggered by polymorphonuclear-myeloid-derived suppressor cells, which express high levels of Fas-ligand and are enriched in TiRP tumors. Blocking Fas-ligand increases the anti-tumor efficacy of adoptive T-cell therapy in TiRP tumors, and increases the efficacy of checkpoint blockade in transplanted tumors. Therefore, tumor-infiltrating lymphocytes apoptosis is a relevant mechanism of immunotherapy resistance, which could be blocked by interfering with the Fas/Fas-ligand pathway.Cancer immunotherapy is ineffective in a subset of patients. Here the authors show that, in a mouse model of melanoma, resistance to immune checkpoint inhibitors relies on loss of tumor-specific T cells through FasL-mediated apoptosis triggered by polymorphonuclear-myeloid-derived suppressor cells.


Cancer Immunology, Immunotherapy | 2006

A new LAGE-1 peptide recognized by cytolytic T lymphocytes on HLA-A68 tumors

Zhaojun Sun; Bernard Lethe; Yi Zhang; Vincenzo Russo; Didier Colau; Vincent Stroobant; Thierry Boon; Pierre van der Bruggen

Antigens encoded by genes of the LAGE family, including LAGE-1 and NY-ESO-1, are of interest for cancer immunotherapy because they are tumor-specific and shared by tumors of different histological types. Several clinical trials are in progress with NY-ESO-1 peptides, protein, recombinant poxviruses, and dendritic cells pulsed with peptides. In this study, CD8 T lymphocytes from an individual without cancer were stimulated with dendritic cells infected with a recombinant avian poxvirus encoding a complete LAGE-1 protein. A CTL clone was isolated that recognized a new LAGE-1 peptide, ELVRRILSR, which corresponds to position 103–111 of the protein sequence. It is presented by HLA-A6801 molecules. When tumor cells expressing LAGE-1 were transfected with HLA-A68, they were lysed by the CTL clone, indicating that the peptide is processed in tumor cells. These results indicate that the LAGE-1.A68 peptide can be used for antitumoral vaccination. We observed also that specific T cells could be detected in a blood sample with a high sensitivity by using an A68/LAGE-1 fluorescent multimer.

Collaboration


Dive into the Didier Colau's collaboration.

Top Co-Authors

Avatar

Benoît Van den Eynde

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Pierre Coulie

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Catherine Uyttenhove

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Vincent Stroobant

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Luc Pilotte

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Pierre van der Bruggen

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Sophie Lucas

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Thierry Boon

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Céline Powis de Tenbossche

Ludwig Institute for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge