Valérie Dutoit
Ludwig Institute for Cancer Research
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Valérie Dutoit.
Immunological Reviews | 2002
Pedro Romero; Danila Valmori; Mikael J. Pittet; Alfred Zippelius; Donata Rimoldi; Frédéric Lévy; Valérie Dutoit; Maha Ayyoub; Verena Rubio-Godoy; Olivier Michielin; Philippe Guillaume; Pascal Batard; Immanuel F. Luescher; Ferdy Lejeune; Danielle Liénard; Nathalie Rufer; Pierre-Yves Dietrich; Daniel E. Speiser; Jean-Charles Cerottini
Summary: Some cancer patients mount spontaneous T‐ and B‐cell responses against their tumor cells. Autologous tumor reactive CD8 cytolytic T lymphocyte (CTL) and CD4 T‐cell clones as well as antibodies from these patients have been used for the identification of genes encoding the target antigens. This knowledge opened the way for new approaches to the immunotherapy of cancer. In this review, we describe the characterization of the structure‐function properties of the melanocyte/melanoma tumor antigen Melan‐A/MART‐1, the assessment of the T‐cell repertoire available against this antigen in healthy individuals, and the analysis of naturally acquired and/or vaccine‐induced CTL responses to this antigen in patients with metastatic melanoma.
Journal of Immunology | 2002
Danila Valmori; Valérie Dutoit; Valérie Schnuriger; Anne-Lise Quiquerez; Mikael J. Pittet; Philippe Guillaume; Verena Rubio-Godoy; Paul R. Walker; Donata Rimoldi; Danielle Liénard; Jean-Charles Cerottini; Pedro Romero; Pierre-Yves Dietrich
Both the underlying molecular mechanisms and the kinetics of TCR repertoire selection following vaccination against tumor Ags in humans have remained largely unexplored. To gain insight into these questions, we performed a functional and structural longitudinal analysis of the TCR of circulating CD8+ T cells specific for the HLA-A2-restricted immunodominant epitope from the melanocyte differentiation Ag Melan-A in a melanoma patient who developed a vigorous and sustained Ag-specific T cell response following vaccination with the corresponding synthetic peptide. We observed an increase in functional avidity of Ag recognition and in tumor reactivity in the postimmune Melan-A-specific populations as compared with the preimmune blood sample. Improved Ag recognition correlated with an increase in the t1/2 of peptide/MHC interaction with the TCR as assessed by kinetic analysis of A2/Melan-A peptide multimer staining decay. Ex vivo analysis of the clonal composition of Melan-A-specific CD8+ T cells at different time points during vaccination revealed that the response was the result of asynchronous expansion of several distinct T cell clones. Some of these T cell clones were also identified at a metastatic tumor site. Collectively, these data show that tumor peptide-driven immune stimulation leads to the selection of high-avidity T cell clones of increased tumor reactivity that independently evolve within oligoclonal populations.
Journal of Immunology | 2000
Danila Valmori; Valérie Dutoit; Danielle Liénard; Ferdy Lejeune; Daniel E. Speiser; Donata Rimoldi; Vincenzo Cerundolo; Pierre-Yves Dietrich; Jean-Charles Cerottini; Pedro Romero
The assessment of the TCR repertoire expressed by tumor-specific CD8+ T lymphocytes has been hampered to date by the difficulty of targeting the analysis to lymphocytes directed against a single epitope. In the present study we have used fluorescent A2/Melan-A tetramers in conjunction with anti-CD8 and anti-TCR β-chain variable (BV) mAbs to analyze by flow cytometry the BV segment usage by Melan-A-specific CD8+ T cells in tumor-infiltrated lymph nodes (TILN) and tumor-infiltrating lymphocytes (TIL) from A2 melanoma patients. Analysis of TILN populations revealed small proportions of A2/Melan-A tetramer+ cells expressing many different BV together with over-representation of A2/Melan-A tetramer+ cells expressing certain BVs. The BV usage by A2/Melan-A tetramer+ lymphocytes in TIL was more restricted than that in TILN. Moreover, the predominant BV segments were quite distinct in populations derived from different patients. A2/Melan-A tetramer+ cells expressing the dominant BVs found in TILN could also be found in the corresponding peptide-stimulated autologous PBMC, although A2/Melan-A tetramer+ lymphocytes expressing additional BVs were also identified. Together, these results suggest that a large and diverse repertoire of Melan-A-specific T cells using different BV TCR segments is available in A2 melanoma patients.
European Journal of Immunology | 2008
Donatella Ciuffreda; Denis Comte; Matthias Cavassini; Emiliano Giostra; Leo H. Buhler; Monika Perruchoud; Markus H. Heim; Manuel Battegay; Daniel Genné; Beat Mulhaupt; Raffaele Malinverni; Carl Oneta; Enos Bernasconi; Martine Monnat; Andreas Cerny; Christian Chuard; Jan Borovicka; Gilles Mentha; Manuel Pascual; Jean-Jacques Gonvers; Giuseppe Pantaleo; Valérie Dutoit
HCV infection has a severe course of disease in HIV/HCV co‐infection and in liver transplant recipients. However, the mechanisms involved remain unclear. Here, we evaluated functional profiles of HCV‐specific T‐cell responses in 86 HCV mono‐infected patients, 48 HIV/HCV co‐infected patients and 42 liver transplant recipients. IFN‐γ and IL‐2 production and ability of CD4 and CD8 T cells to proliferate were assessed after stimulation with HCV‐derived peptides. We observed that HCV‐specific T‐cell responses were polyfunctional in HCV mono‐infected patients, with presence of proliferating single IL‐2‐, dual IL‐2/IFN‐γ and single IFN‐γ‐producing CD4+ and dual IL‐2/IFN‐γ and single IFN‐γ‐producing CD8+ cells. In contrast, HCV‐specific T‐cell responses had an effector profile in HIV/HCV co‐infected individuals and liver transplant recipients with absence of single IL‐2‐producing HCV‐specific CD4+ and dual IL‐2/IFN‐γ‐producing CD8+ T cells. In addition, HCV‐specific proliferation of CD4+ and CD8+ T cells was severely impaired in HIV/HCV co‐infected patients and liver transplant recipients. Importantly, “only effector” T‐cell responses were associated with significantly higher HCV viral load and more severe liver fibrosis scores. Therefore, the present results suggest that immune‐based mechanisms may contribute to explain the accelerated course of HCV infection in conditions of HIV‐1 co‐infection and liver transplantation.
Journal of Immunology | 2000
Donata Rimoldi; Verena Rubio-Godoy; Valérie Dutoit; Danielle Liénard; Suzanne Salvi; Philippe Guillaume; Daniel E. Speiser; Elisabeth Stockert; Giulio C. Spagnoli; Catherine Servis; Jean-Charles Cerottini; Ferdy Lejeune; Pedro Romero; Danila Valmori
Recent studies have shown that CTL epitopes derived from tumor-associated Ags can be encoded by both primary and nonprimary open reading frames (ORF). In this study we have analyzed the HLA-A2-restricted CD8+ T cell response to a recently identified CTL epitope derived from an alternative ORF product of gene LAGE-1 (named CAMEL), and the highly homologous gene NY-ESO-1 in melanoma patients. Using MHC/peptide tetramers we detected CAMEL1–11-specific CD8+ T cells in peptide-stimulated PBMC as well as among tumor-infiltrated lymph node cells from several patients. Sorting and expansion of tetramer+ CD8+ T cells allowed the isolation of tetramerbright and tetramerdull populations that specifically recognized the peptide Ag with high and low avidity, respectively. Remarkably, only high avidity CAMEL-specific CTL were able to recognize Ag-expressing tumor cells. A large series of HLA-A2-positive melanoma cell lines was characterized for the expression of LAGE-1 and NY-ESO-1 mRNA and protein and tested for recognition by CAMEL-specific CTL as well as CTL that recognize a peptide (NY-ESO-1157–165) encoded by the primary ORF products of the LAGE-1 and NY-ESO-1 genes. This analysis revealed that tumor-associated CD8+ T cell epitopes are simultaneously and efficiently generated from both primary and nonprimary ORF products of LAGE-1 and NY-ESO-1 genes and, importantly, that this occurs in the majority of melanoma tumors. These findings underscore the in vivo immunological relevance of CTL epitopes derived from nonprimary ORF products and support their use as candidate vaccines for inducing tumor specific cell-mediated immunity against cancer.
International Journal of Cancer | 2009
Virginie Clément; Valérie Dutoit; Denis Marino; Pierre-Yves Dietrich; Ivan Radovanovic
In human gliomas, self‐renewing and tumor‐initiating cells are characterized by the expression marker CD133. Although, widely used, the validity of CD133 is debated as recent data show that CD133+ and CD133− cells share similar stemness and tumorigenic properties. To clarify this “CD133 controversy”, we reexamined the methods of purification and the stem behavior of both CD133 compartments in fresh gliomas and gliomasphere cultures. Using human anti‐CD133‐coupled microbeads and magnetic activated cell sorting, we observed a nonspecific sorting of glioma cells irrespective of their CD133 expression. In contrast, when purified by fluorescence activating cell sorting, a specific expression and enrichment of CD133 was successfully observed in fresh human gliomas and gliomasphere cultures. However, neither the expression of stemness genes nor the long‐term self‐renewal capacities of CD133+ and CD133− cells were significantly different, even after fresh isolation. Altogether, our data show that purification of CD133+ glioma cells using hCD133‐microbeads presents a lack of specificity and demonstrate that the use of CD133 as a unique glioma stem cell marker is likely not sufficient to tag the whole self‐renewing tumor cell reservoir.
Proceedings of the National Academy of Sciences of the United States of America | 2001
Verena Rubio-Godoy; Valérie Dutoit; Donata Rimoldi; Danielle Liénard; Ferdy Lejeune; Daniel E. Speiser; Philippe Guillaume; Jean-Charles Cerottini; Pedro Romero; Danila Valmori
Activation of CD8+ cytolytic T lymphocytes (CTLs) by antigen is triggered by the interaction of clonotypic αβ T cell receptors (TCRs) with antigenic peptides bound to MHC class I molecules (pMHC complexes). Fluorescent multimeric pMHC complexes have been shown to specifically stain antigen-specific CTLs by directly binding the TCR. In tumor-infiltrating lymphocytes from a melanoma patient we found a high frequency of tyrosinase368–376 peptide-specific cells as detected by IFN-γ ELISPOT, without detectable staining with the corresponding A2/peptide multimers. Surprisingly, these T cells were able to lyse tyrosinase368–376 peptide-pulsed target cells as efficiently as other specific T cells that were stained by multimers. Analysis of the staining patterns under different conditions of incubation time and temperature revealed that these results were explained by major differences in TCR-multimeric ligand interaction kinetics among the clones. Whereas no direct quantitative correlation between antigenic peptide concentration required for CTL effector functions and equilibrium multimer binding was observed interclonally, the latter was profoundly affected by the kinetics of TCR-ligand interaction. More importantly, our data indicate that similar levels of T cell activation can be achieved by independent CD8+ T cell clonotypes displaying different TCR/pMHC complex dissociation rates.
Journal of Clinical Investigation | 2002
Valérie Dutoit; Robert N. Taub; Kyriakos P. Papadopoulos; Susan Talbot; Mary-Louise Keohan; Michelle Brehm; Sacha Gnjatic; Paul E. Harris; Brygida Bisikirska; Philippe Guillaume; Jean-Charles Cerottini; Charles S. Hesdorffer; Lloyd J. Old; Danila Valmori
The cancer-testis antigen NY-ESO-1 is one of the most promising candidates for generic vaccination of cancer patients. Here we analyzed the CD8(+) T cell response to a NY-ESO-1 peptide vaccine composed of the two previously defined peptides 157-165 and 157-167, administered with GM-CSF as a systemic adjuvant. The NY-ESO-1 peptide vaccine elicited a CD8(+) T cell response directed against multiple distinct epitopes in the 157-167 region, as revealed by using A2/peptide multimers incorporating overlapping A2 binding peptides in this region. However, only a minor fraction of the elicited CD8(+) T cells, namely those recognizing the peptide 157-165 with sufficiently high functional avidity, recognized the naturally processed target on NY-ESO-1(+) tumor cells. In contrast, the majority of peptide 157-165-specific CD8(+) T cells exhibited lower functional avidity and no tumor reactivity. In addition, vaccine-elicited CD8(+) T cells specific for other overlapping epitopes in the 157-167 region failed to significantly recognize NY-ESO-1-expressing tumor targets. Thus, because of the complexity of the CD8(+) T cell repertoire that can be elicited by vaccination with synthetic peptides, a precise definition of the targeted epitope, and hence, of the corresponding peptide to be used as immunogen, is required to ensure a precise tumor targeting.
Journal of Immunology | 2002
Valérie Dutoit; Verena Rubio-Godoy; Marie-Agnès Doucey; Pascal Batard; Danielle Liénard; Donata Rimoldi; Daniel E. Speiser; Philippe Guillaume; Jean-Charles Cerottini; Pedro Romero; Danila Valmori
Avidity of Ag recognition by tumor-specific T cells is one of the main parameters that determines the potency of a tumor rejection Ag. In this study we show that the relative efficiency of staining of tumor Ag-specific T lymphocytes with the corresponding fluorescent MHC class I/peptide multimeric complexes can considerably vary with staining conditions and does not necessarily correlate with avidity of Ag recognition. Instead, we found a clear correlation between avidity of Ag recognition and the stability of MHC class I/peptide multimeric complexes interaction with TCR as measured in dissociation kinetic experiments. These findings are relevant for both identification and isolation of tumor-reactive CTL.
European Urology | 2013
Arnoud J. Templeton; Valérie Dutoit; Richard Cathomas; Christian Rothermundt; Daniela Bärtschi; Cornelia Dröge; O. Gautschi; Markus Borner; Eva Fechter; Frank Stenner; Ralph Winterhalder; Beat Müller; Ralph Schiess; Peter Wild; Jan H. Rüschoff; George N. Thalmann; Pierre-Yves Dietrich; Ruedi Aebersold; Dirk Klingbiel; Silke Gillessen
BACKGROUND The phosphatase and tensin homolog (PTEN) tumor suppressor gene is deregulated in many advanced prostate cancers, leading to activation of the phosphatidylinositol 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) pathway and thus increased cell survival. OBJECTIVE To evaluate everolimus, an inhibitor of mTOR, in patients with metastatic castration-resistant prostate cancer (mCRPC), and to explore potentially predictive serum biomarkers by proteomics, the significance of PTEN status in tumor tissue, and the impact of everolimus on immune cell subpopulations and function. DESIGN, SETTING, AND PARTICIPANTS A total of 37 chemotherapy-naive patients with mCRPC and progressive disease were recruited to this single-arm phase 2 trial (ClinicalTrials.gov identifier NCT00976755). INTERVENTION Everolimus was administered continuously at a dose of 10mg daily. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary end point was progression-free survival (PFS) at 12 wk defined as the absence of prostate-specific antigen (PSA), radiographic progression, or clinical progression. Groups were compared using Wilcoxon rank-sum tests or Fisher exact tests for continuous and discrete variables, respectively. Time-to-event end points were analyzed using the Kaplan-Meier method and univariate Cox regression. RESULTS AND LIMITATIONS A total of 13 patients (35%; 95% confidence interval, 20-53) met the primary end point. Confirmed PSA response ≥50% was seen in two (5%), and four further patients (11%) had a PSA decline ≥30%. Higher serum levels of carboxypeptidase M and apolipoprotein B were predictive for reaching the primary end point. Deletion of PTEN was associated with longer PFS and response. Treatment was associated with a dose-dependent decrease of CD3, CD4, and CD8 T lymphocytes and CD8 proliferation and an increase in regulatory T cells. Small sample size was the major limitation of the study. CONCLUSIONS Everolimus activity in unselected patients with mCRPC is moderate, but PTEN deletion could be predictive for response. Several serum glycoproteins were able to predict PFS at 12 wk. Prospective validation of these potential biomarkers is warranted. TRIAL REGISTRATION This study is registered with ClinicalTrials.gov with the identifier NCT00976755. Results of this study were presented in part at the 47th Annual Meeting of the American Society of Clinical Oncology (June 3-7, 2011; Chicago, IL, USA) and the annual meeting of the German, Austrian, and Swiss Societies for Oncology and Hematology (September 30-October 4, 2011; Basel, Switzerland).