Network


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

Hotspot


Dive into the research topics where Pierre-Yves Dietrich is active.

Publication


Featured researches published by Pierre-Yves Dietrich.


International Journal of Cancer | 1999

Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1.

Marie Marchand; Nicolas van Baren; P. Weynants; Vincent Brichard; B. Dréno; Marie Hélène Tessier; Elaine M. Rankin; Giorgio Parmiani; Yves Humblet; A. Bourlond; Romain Vanwijck; Danielle Liénard; M. Beauduin; Pierre-Yves Dietrich; Vincenzo Russo; Joseph Kerger; Giuseppe Masucci; Elke Jäger; Jacques De Grève; Jens Atzpodien; Francis Brasseur; Pierre Coulie; Pierre van der Bruggen; Thierry Boon

Thirty‐nine tumor‐bearing patients with metastatic melanoma were treated with 3 subcutaneous injections of the MAGE‐3.A1 peptide at monthly intervals. No significant toxicity was observed. Of the 25 patients who received the complete treatment, 7 displayed significant tumor regressions. All but one of these regressions involved cutaneous metastases. Three regressions were complete and 2 of these led to a disease‐free state, which persisted for more than 2 years after the beginning of treatment. No evidence for a cytolytic T lymphocyte (CTL) response was found in the blood of the 4 patients who were analyzed, including 2 who displayed complete tumor regression. Our results suggest that injection of the MAGE‐3.A1 peptide induced tumor regression in a significant number of the patients, even though no massive CTL response was produced. Int. J. Cancer 80:219–230, 1999.


Clinical Cancer Research | 2004

Clinical Trial Substantiates the Predictive Value of O-6-Methylguanine-DNA Methyltransferase Promoter Methylation in Glioblastoma Patients Treated with Temozolomide

Monika E. Hegi; Annie Claire Diserens; Sophie Godard; Pierre-Yves Dietrich; Luca Regli; Sandrine Ostermann; Philippe Otten; Guy van Melle; Nicolas de Tribolet; Roger Stupp

Purpose: In the setting of a prospective clinical trial, we determined the predictive value of the methylation status of the O-6-methylguanine-DNA methyltransferase (MGMT) promoter for outcome in glioblastoma patients treated with the alkylating agent temozolomide. Expression of this excision repair enzyme has been associated with resistance to alkylating chemotherapy. Experimental Design: The methylation status of MGMT in the tumor biopsies was evaluated in 38 patients undergoing resection for newly diagnosed glioblastoma and enrolled in a Phase II trial testing concomitant and adjuvant temozolomide and radiation. The epigenetic silencing of the MGMT gene was determined using methylation-specific PCR. Results: Inactivation of the MGMT gene by promoter methylation was associated with longer survival (P = 0.0051; Log-rank test). At 18 months, survival was 62% (16 of 26) for patients testing positive for a methylated MGMT promoter but reached only 8% (1 of 12) in absence of methylation (P = 0.002; Fisher’s exact test). In the presence of other clinically relevant factors, methylation of the MGMT promoter remains the only significant predictor (P = 0.017; Cox regression). Conclusions: This prospective clinical trial identifies MGMT-methylation status as an independent predictor for glioblastoma patients treated with a methylating agent. The association of the epigenetic inactivation of the DNA repair gene MGMT with better outcome in this homogenous cohort may have important implications for the design of future trials and supports efforts to deplete MGMT by O-6-benzylguanine, a noncytotoxic substrate of this enzyme.


Nature Medicine | 2012

Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival

Steffen Walter; Toni Weinschenk; A. Stenzl; Romuald Zdrojowy; Anna Pluzanska; Cezary Szczylik; Michael Staehler; Wolfram Brugger; Pierre-Yves Dietrich; Regina Mendrzyk; Norbert Hilf; Oliver Schoor; Jens Fritsche; Andrea Mahr; Dominik Maurer; Verona Vass; Claudia Trautwein; Peter Lewandrowski; Christian Flohr; Heike Pohla; Janusz J. Stanczak; Vincenzo Bronte; Susanna Mandruzzato; Tilo Biedermann; Graham Pawelec; Evelyna Derhovanessian; Hisakazu Yamagishi; Tsuneharu Miki; Fumiya Hongo; N. Takaha

IMA901 is the first therapeutic vaccine for renal cell cancer (RCC) consisting of multiple tumor-associated peptides (TUMAPs) confirmed to be naturally presented in human cancer tissue. We treated a total of 96 human leukocyte antigen A (HLA-A)*02+ subjects with advanced RCC with IMA901 in two consecutive studies. In the phase 1 study, the T cell responses of the patients to multiple TUMAPs were associated with better disease control and lower numbers of prevaccine forkhead box P3 (FOXP3)+ regulatory T (Treg) cells. The randomized phase 2 trial showed that a single dose of cyclophosphamide reduced the number of Treg cells and confirmed that immune responses to multiple TUMAPs were associated with longer overall survival. Furthermore, among six predefined populations of myeloid-derived suppressor cells, two were prognostic for overall survival, and among over 300 serum biomarkers, we identified apolipoprotein A-I (APOA1) and chemokine (C-C motif) ligand 17 (CCL17) as being predictive for both immune response to IMA901 and overall survival. A randomized phase 3 study to determine the clinical benefit of treatment with IMA901 is ongoing.


Journal of Clinical Oncology | 2010

Phase I/IIa Study of Cilengitide and Temozolomide With Concomitant Radiotherapy Followed by Cilengitide and Temozolomide Maintenance Therapy in Patients With Newly Diagnosed Glioblastoma

Roger Stupp; Monika E. Hegi; Bart Neyns; Roland Goldbrunner; Uwe Schlegel; Paul Clement; Gerhard G. Grabenbauer; Adrian F. Ochsenbein; Matthias Simon; Pierre-Yves Dietrich; Torsten Pietsch; Christine Hicking; Joerg Tonn; Annie Claire Diserens; Alessia Pica; Mirjam Hermisson; Stefan Krueger; Martin Picard; Michael Weller

PURPOSE Invasion and migration are key processes of glioblastoma and are tightly linked to tumor recurrence. Integrin inhibition using cilengitide has shown synergy with chemotherapy and radiotherapy in vitro and promising activity in recurrent glioblastoma. This multicenter, phase I/IIa study investigated the efficacy and safety of cilengitide in combination with standard chemoradiotherapy in newly diagnosed glioblastoma. PATIENTS AND METHODS Patients (age > or = 18 to < or = 70 years) were treated with cilengitide (500 mg) administered twice weekly intravenously in addition to standard radiotherapy with concomitant and adjuvant temozolomide. Treatment was continued until disease progression or for up to 35 weeks. The primary end point was progression-free survival (PFS) at 6 months. RESULTS Fifty-two patients (median age, 57 years; 62% male) were included. Six- and 12-month PFS rates were 69% (95% CI, 54% to 80%) and 33% (95% CI, 21% to 46%). Median PFS was 8 months (95% CI, 6.0 to 10.7 months). Twelve- and 24-month overall survival (OS) rates were 68% (95% CI, 53% to 79%) and 35% (95% CI, 22% to 48%). Median OS was 16.1 months (95% CI, 13.1 to 23.2 months). PFS and OS were longer in patients with tumors with O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation (13.4 and 23.2 months) versus those without MGMT promoter methylation (3.4 and 13.1 months). The combination of cilengitide with temozolomide and radiotherapy was well tolerated, with no additional toxicity. No pharmacokinetic interactions between temozolomide and cilengitide were identified. CONCLUSION Compared with historical controls, the addition of concomitant and adjuvant cilengitide to standard chemoradiotherapy demonstrated promising activity in patients with glioblastoma with MGMT promoter methylation.


Annals of Oncology | 2001

Multicenter phase II trial of temozolomide in patients with glioblastoma multiforme at first relapse

M. Brada; Khê Hoang-Xuan; Roy Rampling; Pierre-Yves Dietrich; L. Y. Dirix; D. Macdonald; J. J. Heimans; Bernard A. Zonnenberg; J. M. Bravo-Marques; Roger Henriksson; Roger Stupp; N. Yue; J. Bruner; M. Dugan; S. Rao; S. Zaknoen

BACKGROUND Recurrent glioblastoma multiforme (GBM) is resistant to most therapeutic endeavors, with low response rates and survival rarely exceeding six months. There are no clearly established chemotherapeutic regimens and the aim of treatment is palliation with improvement in the quality of life. PATIENTS AND METHODS We report an open-label, uncontrolled, multicenter phase II trial of temozolomide in 138 patients (intent-to-treat [ITT] population) with glioblastoma multiforme at first relapse and a Karnofsky performance status (KPS) > or = 70. One hundred twenty-eight patients were histologically confirmed with GBM or gliosarcoma (GS) by independent central review. Chemotherapy-naïve patients were treated with temozolomide 200 mg/m2/day orally for the first five days of a 28-day cycle. Patients previously treated with nitrosourea-containing adjuvant chemotherapy received 150 mg/m2/day for the first five days of a 28-day cycle. In the absence of grade 3 or 4 toxicity, patients on the 150 mg/m2 dose schedule were eligible for a 200 mg/m2 dose on the next cycle. RESULTS The primary endpoint was six-month progression-free survival assessed with strict radiological and clinical criteria. Secondary endpoints included radiological response and Health-related Quality of Life (HQL). Progression-free survival at six months was 18% (95% confidence interval (CI): 11%-26%) for the eligible-histology population. Median progression-free survival and median overall survival were 2.1 months and 5.4 months, respectively. The six-month survival rate was 46%. The objective response rate (complete response and partial response) determined by independent central review of gadolinium-enhanced magnetic resonance imaging (MRI) scans was 8% for both the ITT and eligible-histology populations, with an additional 43% and 45% of patients, respectively, having stable disease (SD). Objectively assessed response and maintenance of a progression-free status were both associated with HQL benefits (characterized by improvements over baseline in HQL domains). Temozolomide had an acceptable safety profile, with only 9% of therapy cycles requiring a dose reduction due to thrombocytopenia. There was no evidence of cumulative hematologic toxicity. CONCLUSIONS Temozolomide demonstrated modest clinical efficacy, with an acceptable safety profile and measurable improvement in quality of life in patients with recurrent GBM. The use of this drug should be explored further in an adjuvant setting and in combination with other agents.


Journal of Clinical Oncology | 2010

Long-Term Follow-Up of Patients With Follicular Lymphoma Receiving Single-Agent Rituximab at Two Different Schedules in Trial SAKK 35/98

Giovanni Martinelli; Shu Fang Hsu Schmitz; Urs Utiger; Thomas Cerny; Urs Hess; Simona Bassi; Emmie Okkinga; Roger Stupp; Rolf A. Stahel; Marc Heizmann; Daniel A. Vorobiof; Andreas Lohri; Pierre-Yves Dietrich; Emanuele Zucca; Michele Ghielmini

PURPOSE We report the long-term results of a randomized clinical trial comparing induction therapy with once per week for 4 weeks single-agent rituximab alone versus induction followed by 4 cycles of maintenance therapy every 2 months in patients with follicular lymphoma. PATIENTS AND METHODS Patients (prior chemotherapy 138; chemotherapy-naive 64) received single-agent rituximab and if nonprogressive, were randomly assigned to no further treatment (observation) or four additional doses of rituximab given at 2-month intervals (prolonged exposure). RESULTS At a median follow-up of 9.5 years and with all living patients having been observed for at least 5 years, the median event-free survival (EFS) was 13 months for the observation and 24 months for the prolonged exposure arm (P < .001). In the observation arm, patients without events at 8 years were 5%, while in the prolonged exposure arm they were 27%. Of previously untreated patients receiving prolonged treatment after responding to rituximab induction, at 8 years 45% were still without event. The only favorable prognostic factor for EFS in a multivariate Cox regression was the prolonged rituximab schedule (hazard ratio, 0.59; 95% CI, 0.39 to 0.88; P = .009), whereas being chemotherapy naive, presenting with stage lower than IV, and showing a VV phenotype at position 158 of the Fc-gamma RIIIA receptor were not of independent prognostic value. No long-term toxicity potentially due to rituximab was observed. CONCLUSION An important proportion of patients experienced long-term remission after prolonged exposure to rituximab, particularly if they had no prior treatment and responded to rituximab induction.


Journal of Clinical Oncology | 2005

Tumoral and Immunologic Response After Vaccination of Melanoma Patients With an ALVAC Virus Encoding MAGE Antigens Recognized by T Cells

Nicolas van Baren; Marie-Claude Bonnet; Brigitte Dreno; Amir Khammari; T. Dorval; Sophie Piperno-Neumann; Danielle Liénard; Daniel E. Speiser; Marie Marchand; Vincent Brichard; Bernard Escudier; Sylvie Négrier; Pierre-Yves Dietrich; Dominique Maraninchi; Susanne Osanto; Ralf G. Meyer; Gerd Ritter; Philippe Moingeon; Jim Tartaglia; Pierre van der Bruggen; Pierre G. Coulie; Thierry Boon

PURPOSE To evaluate the toxicity, antitumoral effectiveness, and immunogenicity of repeated vaccinations with ALVAC miniMAGE-1/3, a recombinant canarypox virus containing a minigene encoding antigenic peptides MAGE-3(168-176) and MAGE-1(161-169), which are presented by HLA-A1 and B35 on tumor cells and can be recognized by cytolytic T lymphocytes (CTLs). MATERIALS AND METHODS The vaccination schedule comprised four sequential injections of the recombinant virus, followed by three booster vaccinations with the MAGE-3(168-176) and MAGE-1(161-169) peptides. The vaccines were administered, both intradermally and subcutaneously, at 3-week intervals. RESULTS Forty patients with advanced cancer were treated, including 37 melanoma patients. The vaccines were generally well tolerated with moderate adverse events, consisting mainly of transient inflammatory reactions at the virus injection sites. Among the 30 melanoma patients assessable for tumor response, a partial response was observed in one patient, and disease stabilization in two others. The remaining patients had progressive disease. Among the patients with stable or progressive disease, five showed evidence of tumor regression. A CTL response against the MAGE-3 vaccine antigen was detected in three of four patients with tumor regression, and in only one of 11 patients without regression. CONCLUSION Repeated vaccination with ALVAC miniMAGE-1/3 is associated with tumor regression and with a detectable CTL response in a minority of melanoma patients. There is a significant correlation between tumor regression and CTL response. The contribution of vaccine-induced CTL in the tumor regression process is discussed in view of the immunologic events that could be analyzed in detail in one patient.


Immunological Reviews | 2002

Antigenicity and immunogenicity of Melan-A/MART-1 derived peptides as targets for tumor reactive CTL in human melanoma.

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.


Current Opinion in Immunology | 1998

Tumor expression of Fas ligand (CD95L) and the consequences.

Paul R. Walker; Philippe Saas; Pierre-Yves Dietrich

Tumors of diverse origin have recently been shown to express CD95 ligand and to induce apoptosis in CD95-expressing targets in vitro; however, in vivo, enhanced tumor growth and rejection have both been observed as a consequence of either immunosuppressive or proinflammatory functions of CD95 ligand. The final in vivo outcome of CD95 ligand expression will depend upon a complex balance of interactions relevant for each tumor in its particular microenvironment.


Glia | 2000

TWEAK stimulation of astrocytes and the proinflammatory consequences

Philippe Saas; José Boucraut; Paul R. Walker; Anne-Lise Quiquerez; Maryse Billot; Sophie Desplat-Jégo; Yves Chicheportiche; Pierre-Yves Dietrich

Astrocytes exert many active roles in brain homeostasis, potentially including the regulation of immune reactions. They possess a substantial aptitude for plasticity and, indeed, functional and phenotypic changes are frequently encountered in reactive gliosis observed in brain injuries. The significance of reactive astrocytes is still poorly defined, but it is clear that these cells are an important source of cytokines in inflamed brain. How tumor necrosis factor (TNF) and TNF‐receptor family members contribute to this reaction is an interesting issue that is currently being explored. It was previously shown that reactive astrocytes express high levels of Fas (CD95) and respond to Fas ligand (CD95L) by apoptosis or IL‐8 production. TWEAK (Apo‐3 ligand) is a recently identified member of the TNF family that is produced mainly by leukocytes that can infiltrate the inflamed brain and thus influence astrocyte behavior. Here we show that human astrocytes derived from different regions of the brain specifically bind TWEAK and are totally resistant to TWEAK mediated apoptosis. In addition, high amounts of IL‐8 and IL‐6 were secreted by astrocytes after TWEAK exposure. Finally, expression of cell surface molecules involved in the propagation and/or maintenance of brain inflammation was determined. TWEAK significantly increased ICAM‐1 expression on astrocytes, whereas no modification was detected in the expression of Fas, TNFRI, B7‐1, or MHC molecules. In conclusion, the proinflammatory effects induced by TWEAK on astrocytes in culture recapitulate many characteristics of reactive astrocytes observed in vivo, suggesting that TWEAK could play a significant role in brain inflammation. GLIA 32:102–107, 2000.

Collaboration


Dive into the Pierre-Yves Dietrich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Valérie Dutoit

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Pedro Romero

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Danielle Liénard

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jean-Charles Cerottini

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roger Stupp

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge