Network


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

Hotspot


Dive into the research topics where Jacques Chapiro is active.

Publication


Featured researches published by Jacques Chapiro.


Immunological Reviews | 2002

Tumor‐specific shared antigenic peptides recognized by human T cells

Pierre van der Bruggen; Yi Zhang; Pascal Chaux; Vincent Stroobant; Christophe Panichelli; Erwin S. Schultz; Jacques Chapiro; Benoît Van den Eynde; Francis Brasseur; Thierry Boon

Summary: The first tumor‐specific shared antigens and the cancer‐germline genes that code for these antigens were identified with antitumor cytolytic T lymphocytes obtained from cancer patients. A few HLA class I‐restricted antigenic peptides were identified by this ‘direct approach’. A large set of additional cancer‐germline genes have now been identified by purely genetic approaches or by screening tumor cDNA expression libraries with the serum of cancer patients. As a result, a vast number of sequences are known that can code for tumor‐specific shared antigens, but most of the encoded antigenic peptides have not yet been identified. We review here recent ‘reverse immunology’ approaches for the identification of new antigenic peptides. They are based on in vitro stimulation of naive T cells with dendritic cells that have either been loaded with a cancer‐germline protein or that have been transduced with viruses carrying cancer‐germline coding sequences. These approaches have led to the identification of many new antigenic peptides presented by class I or class II molecules. We also describe some aspects of the processing and presentation of these antigenic peptides.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Two abundant proteasome subtypes that uniquely process some antigens presented by HLA class I molecules

Benoît Guillaume; Jacques Chapiro; Vincent Stroobant; Didier Colau; Benoît Van Holle; Grégory Parvizi; Marie-Pierre Bousquet-Dubouch; Ivan Théate; Nicolas Parmentier; Benoît Van den Eynde

Most antigenic peptides presented by MHC class I molecules result from the degradation of intracellular proteins by the proteasome. In lymphoid tissues and cells exposed to IFNγ, the standard proteasome is replaced by the immunoproteasome, in which all of the standard catalytic subunits β1, β2, and β5 are replaced by their inducible counterparts β1i, β2i, and β5i, which have different cleavage specificities. The immunoproteasome thereby shapes the repertoire of antigenic peptides. The existence of additional forms of proteasomes bearing a mixed assortment of standard and inducible catalytic subunits has been suggested. Using a new set of unique subunit-specific antibodies, we have now isolated, quantified, and characterized human proteasomes 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. These intermediate proteasomes represent between one-third and one-half of the proteasome content of human liver, colon, small intestine, and kidney. They are also present in human tumor cells and dendritic cells. We identified two tumor antigens of clinical interest that are processed exclusively either by intermediate proteasomes β5i (MAGE-A3271–279) or by intermediate proteasomes β1i-β5i (MAGE-A10254–262). The existence of these intermediate proteasomes broadens the repertoire of antigens presented to CD8 T cells and implies that the antigens presented by a given cell depend on their proteasome content.


Cancer Research | 2011

Antigen Spreading Contributes to MAGE Vaccination- Induced Regression of Melanoma Metastases

Véronique Corbière; Jacques Chapiro; Vincent Stroobant; Wenbin Ma; Christophe Lurquin; Bernard Lethe; Nicolas van Baren; Benoît Van den Eynde; Thierry Boon; Pierre G. Coulie

A core challenge in cancer immunotherapy is to understand the basis for efficacious vaccine responses in human patients. In previous work we identified a melanoma patient who displayed a low-level antivaccine cytolytic T-cell (CTL) response in blood with tumor regression after vaccination with melanoma antigens (MAGE). Using a genetic approach including T-cell receptor β (TCRβ) cDNA libraries, we found very few antivaccine CTLs in regressing metastases. However, a far greater number of TCRβ sequences were found with several of these corresponding to CTL clones specific for nonvaccine tumor antigens, suggesting that antigen spreading was occurring in regressing metastases. In this study, we found another TCR belonging to tumor-specific CTL enriched in regressing metastases and detectable in blood only after vaccination. We used the TCRβ sequence to detect and clone the desired T cells from tumor-infiltrating lymphocytes isolated from the patient. This CD8 clone specifically lysed autologous melanoma cells and displayed HLA-A2 restriction. Its target antigen was identified as the mitochondrial enzyme caseinolytic protease. The target antigen gene was mutated in the tumor, resulting in production of a neoantigen. Melanoma cell lysis by the CTL was increased by IFN-γ treatment due to preferential processing of the antigenic peptide by the immunoproteasome. These results argue that tumor rejection effectors in the patient were indeed CTL responding to nonvaccine tumor-specific antigens, further supporting our hypothesis. Among such antigens, the mutated antigen we found is the only antigen against which no T cells could be detected before vaccination. We propose that antigen spreading of an antitumor T-cell response to truly tumor-specific antigens contributes decisively to tumor regression.


Journal of Experimental Medicine | 2002

The Production of a New MAGE-3 Peptide Presented to Cytolytic T Lymphocytes by HLA-B40 Requires the Immunoproteasome

Erwin S. Schultz; Jacques Chapiro; Christophe Lurquin; Stéphane Claverol; Odile Burlet-Schiltz; Guy Warnier; Vincenzo Russo; Sandra Morel; Frédéric Lévy; Thierry Boon; Benoît Van den Eynde; Pierre van der Bruggen

By stimulating human CD8+ T lymphocytes with autologous dendritic cells infected with an adenovirus encoding MAGE-3, we obtained a cytotoxic T lymphocyte (CTL) clone that recognized a new MAGE-3 antigenic peptide, AELVHFLLL, which is presented by HLA-B40. This peptide is also encoded by MAGE-12. The CTL clone recognized MAGE-3–expressing tumor cells only when they were first treated with IFN-γ. Since this treatment is known to induce the exchange of the three catalytic subunits of the proteasome to form the immunoproteasome, this result suggested that the processing of this MAGE-3 peptide required the immunoproteasome. Transfection experiments showed that the substitution of β5i (LMP7) for β5 is necessary and sufficient for producing the peptide, whereas a mutated form of β5i (LMP7) lacking the catalytically active site was ineffective. Mass spectrometric analyses of in vitro digestions of a long precursor peptide with either proteasome type showed that the immunoproteasome produced the antigenic peptide more efficiently, whereas the standard proteasome more efficiently introduced cleavages destroying the antigenic peptide. This is the first example of a tumor-specific antigen exclusively presented by tumor cells expressing the immunoproteasome.


Journal of Immunology | 2006

Destructive cleavage of antigenic peptides either by the immunoproteasome or by the standard proteasome results in differential antigen presentation.

Jacques Chapiro; Stéphane Claverol; Fanny Piette; Wenbin Ma; Vincent Stroobant; Benoît Guillaume; Jean-Edouard Gairin; Sandra Morel; Odile Burlet-Schiltz; Bernard Monsarrat; Thierry Boon; Benoît Van den Eynde

The immunoproteasome (IP) is usually viewed as favoring the production of antigenic peptides presented by MHC class I molecules, mainly because of its higher cleavage activity after hydrophobic residues, referred to as the chymotrypsin-like activity. However, some peptides have been found to be better produced by the standard proteasome. The mechanism of this differential processing has not been described. By studying the processing of three tumor antigenic peptides of clinical interest, we demonstrate that their differential processing mainly results from differences in the efficiency of internal cleavages by the two proteasome types. Peptide gp100209–217 (ITDQVPSFV) and peptide tyrosinase369–377 (YMDGTMSQV) are destroyed by the IP, which cleaves after an internal hydrophobic residue. Conversely, peptide MAGE-C2336–344 (ALKDVEERV) is destroyed by the standard proteasome by internal cleavage after an acidic residue, in line with its higher postacidic activity. These results indicate that the IP may destroy some antigenic peptides due to its higher chymotrypsin-like activity, rather than favor their production. They also suggest that the sets of peptides produced by the two proteasome types differ more than expected. Considering that mature dendritic cells mainly contain IPs, our results have implications for the design of immunotherapy strategies.


Nature Immunology | 2010

Production of an antigenic peptide by insulin-degrading enzyme

Nicolas Parmentier; Vincent Stroobant; Didier Colau; Philippe de Diesbach; Sandra Morel; Jacques Chapiro; Peter van Endert; Benoît Van den Eynde

Most antigenic peptides presented by major histocompatibility complex (MHC) class I molecules are produced by the proteasome. Here we show that a proteasome-independent peptide derived from the human tumor protein MAGE-A3 is produced directly by insulin-degrading enzyme (IDE), a cytosolic metallopeptidase. Cytotoxic T lymphocyte recognition of tumor cells was reduced after metallopeptidase inhibition or IDE silencing. Separate inhibition of the metallopeptidase and the proteasome impaired degradation of MAGE-A3 proteins, and simultaneous inhibition of both further stabilized MAGE-A3 proteins. These results suggest that MAGE-A3 proteins are degraded along two parallel pathways that involve either the proteasome or IDE and produce different sets of antigenic peptides presented by MHC class I molecules.


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.


International Journal of Cancer | 2011

A MAGE-C2 antigenic peptide processed by the immunoproteasome is recognized by cytolytic T cells isolated from a melanoma patient after successful immunotherapy.

Wenbin Ma; Nathalie Vigneron; Jacques Chapiro; Vincent Stroobant; Catherine Germeau; Thierry Boon; Pierre Coulie; Benoît Van den Eynde

We have pursued our analysis of a melanoma patient who showed almost complete tumor regression following vaccination with MAGE‐A1 and MAGE‐A3 antigens. We previously described high frequencies of tumor‐specific CTL precursors in blood samples collected after but also before vaccination. A set of CTL clones were derived that recognized antigens different from those of the vaccine. Two of these antigens were peptides encoded by another MAGE gene, MAGE‐C2. Here we describe the antigen recognized by another tumor‐specific CTL clone. It proved to be a third antigenic peptide encoded by gene MAGE‐C2, ASSTLYLVF. It is presented by HLA‐B57 molecules and proteasome‐dependent. Tumor cells exposed to interferon‐gamma (IFN‐γ) were better recognized by the anti‐MAGE‐C242‐50 CTL clone. This mainly resulted from a better processing of the peptide by the immunoproteasome as compared to the standard proteasome. Mass spectrometric analyses showed that the latter destroyed the antigenic peptide by cleaving between two internal hydrophobic residues. Despite its higher “chymotryptic‐like” (posthydrophobic) activity, the immunoproteasome did not cleave at this position, in line with the suggestion that hydrophobic residues immediately downstream from a cleavage site impair cleavage by the immunoproteasome. We previously reported that one of the other MAGE‐C2 peptides recognized by CTL from this patient was also better processed by the immunoproteasome. Together, these results support the notion that the tumor regression of this patient was mediated by an antitumor response shaped by IFN‐γ and dominated by CTL directed against peptides that are better produced by the immunoproteasome, such as the MAGE‐C2 peptides.


Science | 2004

An antigenic peptide produced by peptide splicing in the proteasome

Nathalie Vigneron; Vincent Stroobant; Jacques Chapiro; Annie Ooms; Gérard Degiovanni; Sandra Morel; Pierre van der Bruggen; Thierry Boon; Benoît Van den Eynde


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

Collaboration


Dive into the Jacques Chapiro's collaboration.

Top Co-Authors

Avatar

Benoît Van den Eynde

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Vincent Stroobant

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Thierry Boon

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Sandra Morel

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Didier Colau

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Benoît Guillaume

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Nathalie Vigneron

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Nicolas Parmentier

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

Wenbin Ma

Ludwig Institute for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge