Network


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

Hotspot


Dive into the research topics where Sebastien Rinaldetti is active.

Publication


Featured researches published by Sebastien Rinaldetti.


Leukemia | 2015

Safety and efficacy of imatinib in CML over a period of 10 years: data from the randomized CML-study IV

Lida Kalmanti; Susanne Saussele; Michael Lauseker; Markus Müller; Christian Dietz; L Heinrich; Benjamin Hanfstein; Ulrike Proetel; A. Fabarius; S. W. Krause; Sebastien Rinaldetti; Jolanta Dengler; C. Falge; E Oppliger-Leibundgut; Andreas Burchert; Andreas Neubauer; Lothar Kanz; Frank Stegelmann; Michael Pfreundschuh; Karsten Spiekermann; Christof Scheid; Markus Pfirrmann; Andreas Hochhaus; Jörg Hasford; Rüdiger Hehlmann

Tyrosine kinase inhibitors (TKI) have changed the natural course of chronic myeloid leukemia (CML). With the advent of second-generation TKI safety and efficacy issues have gained interest. The randomized CML - Study IV was used for a long-term evaluation of imatinib (IM). 1503 patients have received IM, 1379 IM monotherapy. After a median observation of 7.1 years, 965 patients (64%) still received IM. At 10 years, progression-free survival was 82%, overall survival 84%, 59% achieved MR5, 72% MR4.5, 81% MR4, 89% major molecular remission and 92% MR2 (molecular equivalent to complete cytogenetic remission). All response levels were reached faster with IM800 mg except MR5. Eight-year probabilities of adverse drug reactions (ADR) were 76%, of grades 3–4 22%, of non-hematologic 73%, and of hematologic 28%. More ADR were observed with IM800 mg and IM400 mg plus interferon α (IFN). Most patients had their first ADR early with decreasing frequency later on. No new late toxicity was observed. ADR to IM are frequent, but mostly mild and manageable, also with IM 800 mg and IM 400 mg+IFN. The deep molecular response rates indicate that most patients are candidates for IM discontinuation. After 10 years, IM continues to be an excellent initial choice for most patients with CML.


Leukemia | 2017

Assessment of imatinib as first-line treatment of chronic myeloid leukemia: 10-year survival results of the randomized CML study IV and impact of non-CML determinants

Rüdiger Hehlmann; Michael Lauseker; Susanne Saußele; Markus Pfirrmann; S. W. Krause; H. J. Kolb; Andreas Neubauer; D. K. Hossfeld; Christoph Nerl; Alois Gratwohl; Dominik Heim; Tim H. Brümmendorf; A. Fabarius; Claudia Haferlach; Brigitte Schlegelberger; Markus Müller; S. Jeromin; Ulrike Proetel; K. Kohlbrenner; A. Voskanyan; Sebastien Rinaldetti; Wolfgang Seifarth; B. Spieß; Leopold Balleisen; Maria-Elisabeth Goebeler; Matthias Hänel; Anthony D. Ho; Jolanta Dengler; C. Falge; Lothar Kanz

Chronic myeloid leukemia (CML)-study IV was designed to explore whether treatment with imatinib (IM) at 400 mg/day (n=400) could be optimized by doubling the dose (n=420), adding interferon (IFN) (n=430) or cytarabine (n=158) or using IM after IFN-failure (n=128). From July 2002 to March 2012, 1551 newly diagnosed patients in chronic phase were randomized into a 5-arm study. The study was powered to detect a survival difference of 5% at 5 years. After a median observation time of 9.5 years, 10-year overall survival was 82%, 10-year progression-free survival was 80% and 10-year relative survival was 92%. Survival between IM400 mg and any experimental arm was not different. In a multivariate analysis, risk group, major-route chromosomal aberrations, comorbidities, smoking and treatment center (academic vs other) influenced survival significantly, but not any form of treatment optimization. Patients reaching the molecular response milestones at 3, 6 and 12 months had a significant survival advantage. For responders, monotherapy with IM400 mg provides a close to normal life expectancy independent of the time to response. Survival is more determined by patients’ and disease factors than by initial treatment selection. Although improvements are also needed for refractory disease, more life-time can currently be gained by carefully addressing non-CML determinants of survival.


Oncotarget | 2017

FOXM1 predicts overall and disease specific survival in muscle-invasive urothelial carcinoma and presents a differential expression between bladder cancer subtypes

Sebastien Rinaldetti; Ralph M. Wirtz; Thomas Stefan Worst; Markus Eckstein; Cleo Aaron Weiss; Johannes Breyer; Wolfgang Otto; Christian Bolenz; Arndt Hartmann; Philipp Erben

Forkhead box M1 (FOXM1) is a late cell cycle gene that plays a crucial role in carcinogenesis and chemotherapeutic drug resistance. In this study, the impact of FOXM1 expression on patient outcome was investigated for the first time in formalin fixed and paraffin embedded (FFPE) samples of chemotherapy naïve muscle-invasive bladder cancer (MIBC) patients. Expression analyses were performed on the Mannheim cohort (n=84) and validated on the independent Chungbuk cohort (n=61). In a Cox’ proportional hazards model, a distinct FOXM1 expression cut-off dividing both cohorts in a ‘high-risk’ and ‘low-risk’ group has been determined. Multivariate analyses showed that FOXM1 is an independent risk factor for outcome prediction superior to the TNM system. The FOXM1 ‘high-risk’ group had a 4- to 7-fold increased risk of death (p<0.03) and presented further an overexpression of MKI67. Recent studies showed that MIBCs can be subclassified in breast cancer-like subtypes: basal, luminal and p53-like. Here we demonstrated that FOXM1 was differentially expressed between MIBC subtypes concordant to its subtype specific expression in breast cancer. Since the proto-oncogene FOXM1 is known to play an important role in cisplatin resistance and to be a promising drug target, this study supports FOXM1 as a crucial biomarker in the personalization of MIBC therapy and urges prospective translational studies.


PLOS ONE | 2018

The benefit of quality control charts (QCC) for routine quantitative BCR-ABL1 monitoring in chronic myeloid leukemia

Birgit Spiess; Nicole Naumann; Norbert Galuschek; Sebastien Rinaldetti; Ute Kossak-Roth; Irina Tarnopolscaia; Elena Felde; Alice Fabarius; Wolf-Karsten Hofmann; Susanne Saußele; Wolfgang Seifarth

Quantitative real-time polymerase chain reaction (qRT-PCR) is state of the art in molecular monitoring of minimal residual disease in chronic myeloid leukemia (CML). In this context, maintenance of assay fidelity and detection of technical inaccuracy are crucial. Beside multiple common negative controls for the clinical sample preparations, quality control charts (QCC) are a common validation tool to sustain high process quality by continuously recording of qRT-PCR control parameters. Here, we report on establishment and benefit of QCC in qRT-PCR-based CML diagnostics. The absolute quantification of BCR-ABL1 fusion transcripts in patient samples is based on coamplification of a serially diluted reference plasmid (pME-2). For QCC establishment the measured Ct values of each pME-2 standard dilution (4–400,000) of a test set resembling 21 sequential qRT-PCR experiments were recorded and statistically evaluated. Test set data were used for determination of warning limits (mean +/- 2-fold standard deviation) and control (intervention) limits (mean +/- 3-fold standard deviation) to allow rapid detection of defined out-of-control situations which may require intervention. We have retrospectively analyzed QCC data of 282 sequential qRT-PCR experiments (564 reactions). Data evaluation using QCCs revealed three out-of-control situations that required intervention like experiment repeats, renewal of pME-2 standards, replacement of reagents or personnel re-training. In conclusion, with minimal more effort and hands-on time QCC rank among the best tools to grant high quality and reproducibility in CML routine molecular diagnosis.


Oncotarget | 2018

Subclassification, survival prediction and drug target analyses of chemotherapy-naïve muscle-invasive bladder cancer with a molecular screening

Sebastien Rinaldetti; Eugen Rempel; Thomas Stefan Worst; Markus Eckstein; Annette Steidler; Cleo Aaron Weiss; Christian Bolenz; Arndt Hartmann; Philipp Erben

Background Transcriptome expression studies identified distinct muscle invasive bladder cancer (MIBC) subtypes closely related with breast cancer subclasses. Here we developed a sensitive quantification method for MIBC subclassification (luminal, basal, p53-like). In addition, the subtype specific expression of drug targets has been investigated. Methods Absolute quantification (nCounter) of a 64-gene panel was performed on MIBC patients (n=47) treated exclusively with radical cystectomy (RC). In conjunction of 170 MIBCs from 3 independent cohorts, a minimal set of consensus genes has been established. Survival of the consensus subtypes has been assessed by multivariate analysis. Relevant drug targets were tested for their subtype specificity in a clustering independent assessment. Results A reduced 36-gene panel stably clustered into 3 subtypes throughout the cohorts (luminal, basal, infiltrated). Patients treated by RC only, showed worst 8-year disease specific survival (DSS) for the luminal subtype in contrast to the infiltrated subtype (17% vs. 73%, p=0.011). In multivariate analyses, the risk stratification based on luminal versus not-luminal MIBC proved to be an independent predictor for DSS superior to the TNM system in patients with RC. Drug targets (e.g. ERBB2, FGFR, AR, PDGFRB) showed a distinct subtype attribution. The subtypes based on this nCounter screening could further be validated by the TCGA cohort. Conclusion This MIBC subtype screening predicted survival and allowed an analysis of subtype specific drug targets, thus being a powerful tool for the translation of personalized MIBC treatment concepts.


Leukemia | 2018

Defining therapy goals for major molecular remission in chronic myeloid leukemia: Results of the randomized CML Study IV

Susanne Saussele; Rüdiger Hehlmann; Alice Fabarius; Sabine Jeromin; Ulrike Proetel; Sebastien Rinaldetti; Katharina Kohlbrenner; Hermann Einsele; Christiane Falge; Lothar Kanz; Andreas Neubauer; Michael Kneba; Frank Stegelmann; Michael Pfreundschuh; Cornelius F. Waller; Elisabeth Oppliger Leibundgut; Dominik Heim; Stefan W. Krause; Wolf-Karsten Hofmann; Joerg Hasford; Markus Pfirrmann; Martin C. Müller; Andreas Hochhaus; Michael Lauseker

Major molecular remission (MMR) is an important therapy goal in chronic myeloid leukemia (CML). So far, MMR is not a failure criterion according to ELN management recommendation leading to uncertainties when to change therapy in CML patients not reaching MMR after 12 months. At monthly landmarks, for different molecular remission status Hazard ratios (HR) were estimated for patients registered to CML study IV who were divided in a learning and a validation sample. The minimum HR for MMR was found at 2.5 years with 0.28 (compared to patients without remission). In the validation sample, a significant advantage for progression-free survival (PFS) for patients in MMR could be detected (p-value 0.007). The optimal time to predict PFS in patients with MMR could be validated in an independent sample at 2.5 years. With our model we provide a suggestion when to define lack of MMR as therapy failure and thus treatment change should be considered. The optimal response time for 1% BCR-ABL at about 12–15 months was confirmed and for deep molecular remission no specific time point was detected. Nevertheless, it was demonstrated that the earlier the MMR is achieved the higher is the chance to attain deep molecular response later.


Journal of Cancer Research and Clinical Oncology | 2018

FOXM1 predicts disease progression in non-muscle invasive bladder cancer

Sebastien Rinaldetti; Ralph M. Wirtz; Thomas Stefan Worst; Arndt Hartmann; Johannes Breyer; Lars Dyrskjøt; Philipp Erben

PurposeThe proto-oncogene forkhead box M1 (FOXM1) is associated with poor survival in many cancers. The impact of FOXM1 expression on progression-free survival (PFS) of non-muscle invasive bladder cancer (NMIBC) has not yet been investigated. The differential expression of FOXM1 between the different molecular NMIBC subtypes has further been assessed.MethodsTranscript levels of FOXM1 and MKI67 were determined in 460 NMIBC patients (UROMOL cohort) by RNA-Seq and validated in silico by the Chungbuk and Lund cohort (n = 277). FOXM1 and MKI67 cutoffs were identified by the minimal p value method. Variables were evaluated by multivariable Cox regression analyses in order to identify independent predictors.ResultsFOXM1 is an independent predictor for PFS superior to current histological, clinical and molecular staging methods. Patients with high FOXM1 expression have a 6- to 8-fold higher risk of progression in multivariable analysis (p < 0.03). Highest transcript levels were found in the Class 2 and genomically unstable molecular NMIBC subtype (p < 0.03). The proto-oncogene further positively correlated with tumor grade and stage. NMIBCs with high FOXM1 expression showed a PFS advantage when treated with intravesical BCG instillation.ConclusionFOXM1 is a highly prognostic marker for disease progression of NMIBC superior to current histological, clinical and molecular staging methods and MKI67. It is mainly expressed in the Class 2 and genomically unstable molecular bladder cancer subtypes. Its role in drug resistance development makes FOXM1 valuable biomarker for NMIBC risk stratification.


Clinical Lymphoma, Myeloma & Leukemia | 2018

Effect of ABCG2, OCT1, and ABCB1 (MDR1) Gene Expression on Treatment-Free Remission in a EURO-SKI Subtrial

Sebastien Rinaldetti; Markus Pfirrmann; Kirsi Marjaana Manz; Joelle Guilhot; Christian Dietz; Panayiotidis Panagiotidis; Birgit Spiess; Wolfgang Seifarth; Alice Fabarius; Martin Müller; Maria Pagoni; Maria Dimou; Jolanta Dengler; Cornelius F. Waller; Tim H. Brümmendorf; Regina Herbst; Andreas Burchert; Carsten Janβen; Maria. Elisabeth. Goebeler; Philipp J. Jost; S. Hanzel; Philippe Schafhausen; Gabriele Prange-Krex; T. Illmer; Viktor Janzen; Martine Klausmann; Robert Eckert; Gerd Büschel; Alexander Kiani; Wolf-Karsten Hofmann

Introduction Tyrosine kinase inhibitors (TKIs) can safely be discontinued in chronic myeloid leukemia (CML) patients with sustained deep molecular response. ABCG2 (breast cancer resistance protein), OCT1 (organic cation transporter 1), and ABCB1 (multidrug resistance protein 1) gene products are known to play a crucial role in acquired pharmacogenetic TKI resistance. Their influence on treatment‐free remission (TFR) has not yet been investigated. Materials and Methods RNA was isolated on the last day of TKI intake from peripheral blood leukocytes of 132 chronic phase CML patients who discontinued TKI treatment within the European Stop Tyrosine Kinase Inhibitor Study trial. Plasmid standards were designed including subgenic inserts of OCT1, ABCG2, and ABCB1 together with GUSB as reference gene. For expression analyses, quantitative real‐time polymerase chain reaction was used. Multiple Cox regression analysis was performed. In addition, gene expression cutoffs for patient risk stratification were investigated. Results The TFR rate of 132 patients, 12 months after TKI discontinuation, was 54% (95% confidence interval [CI], 46%‐62%). ABCG2 expression (‰) was retained as the only significant variable (P = .02; hazard ratio, 1.04; 95% CI, 1.01‐1.07) in multiple Cox regression analysis. Only for the ABCG2 efflux transporter, a significant cutoff was found (P = .04). Patients with an ABCG2/GUSB transcript level >4.5‰ (n = 93) showed a 12‐month TFR rate of 47% (95% CI, 37%‐57%), whereas patients with low ABCG2 expression (≤4.5‰; n = 39) had a 12‐month TFR rate of 72% (95% CI, 55%‐82%). Conclusion In this study, we investigated the effect of pharmacogenetics in the context of a CML treatment discontinuation trial. The transcript levels of the efflux transporter ABCG2 predicted TFR after TKI discontinuation. Micro‐Abstract Within the EURO‐SKI trial, 132 chronic phase CML patients discontinued imatinib treatment. RNA was isolated from peripheral blood in order to analyze the expression of MDR1, ABCG2 and OCT1. ABCG2 was predictive for treatment‐free remission in Cox regression analysis. High transcript levels of the ABCG2 efflux transporter (>4.5‰) were associated with a twofold higher risk of relapse.


BJUI | 2018

FOXM1 overexpression is associated with adverse outcome and predicts response to intravesical instillation therapy in stage pT1 non-muscle-invasive bladder cancer

Johannes Breyer; Ralph M. Wirtz; Philipp Erben; Sebastien Rinaldetti; Thomas Stefan Worst; Robert Stoehr; Markus Eckstein; Danijel Sikic; Stefan Denzinger; Maximilian Burger; Arndt Hartmann; Wolfgang Otto

To investigate the role of forkhead box protein M1 (FOXM1) mRNA expression and its prognostic value in stage pT1 non‐muscle‐invasive bladder cancer (NMIBC).


Annals of Hematology | 2015

Impact of unbalanced minor route versus major route karyotypes at diagnosis on prognosis of CML

Alice Fabarius; Lida Kalmanti; Christian Dietz; Michael Lauseker; Sebastien Rinaldetti; Claudia Haferlach; Gudrun Göhring; Brigitte Schlegelberger; Martine Jotterand; Benjamin Hanfstein; Wolfgang Seifarth; Mathias Hänel; Claus-Henning Köhne; Hans Walter Lindemann; Wolfgang E. Berdel; Peter Staib; Martin C. Müller; Ulrike Proetel; Leopold Balleisen; Maria-Elisabeth Goebeler; Jolanta Dengler; Christiane Falge; Lothar Kanz; Andreas Burchert; Michael Kneba; Frank Stegelmann; Michael Pfreundschuh; Cornelius F. Waller; Karsten Spiekermann; Tim H. Brümmendorf

Collaboration


Dive into the Sebastien Rinaldetti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arndt Hartmann

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge