Network


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

Hotspot


Dive into the research topics where Oliver Bechter is active.

Publication


Featured researches published by Oliver Bechter.


The New England Journal of Medicine | 2017

Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma

Jedd D. Wolchok; Vanna Chiarion-Sileni; Rene Gonzalez; Piotr Rutkowski; Jean-Jacques Grob; C. Lance Cowey; Christopher D. Lao; John Wagstaff; Dirk Schadendorf; Pier Francesco Ferrucci; Michael Smylie; Reinhard Dummer; Andrew F. Hill; David Hogg; John B. A. G. Haanen; Matteo S. Carlino; Oliver Bechter; Michele Maio; Iván Márquez-Rodas; Massimo Guidoboni; Grant A. McArthur; Celeste Lebbe; Paolo Antonio Ascierto; Jonathan Cebon; Jeffrey A. Sosman; Michael A. Postow; Margaret K. Callahan; Dana Walker; Linda Rollin; Rafia Bhore

BACKGROUND Nivolumab combined with ipilimumab resulted in longer progression‐free survival and a higher objective response rate than ipilimumab alone in a phase 3 trial involving patients with advanced melanoma. We now report 3‐year overall survival outcomes in this trial. METHODS We randomly assigned, in a 1:1:1 ratio, patients with previously untreated advanced melanoma to receive nivolumab at a dose of 1 mg per kilogram of body weight plus ipilimumab at a dose of 3 mg per kilogram every 3 weeks for four doses, followed by nivolumab at a dose of 3 mg per kilogram every 2 weeks; nivolumab at a dose of 3 mg per kilogram every 2 weeks plus placebo; or ipilimumab at a dose of 3 mg per kilogram every 3 weeks for four doses plus placebo, until progression, the occurrence of unacceptable toxic effects, or withdrawal of consent. Randomization was stratified according to programmed death ligand 1 (PD‐L1) status, BRAF mutation status, and metastasis stage. The two primary end points were progression‐free survival and overall survival in the nivolumab‐plus‐ipilimumab group and in the nivolumab group versus the ipilimumab group. RESULTS At a minimum follow‐up of 36 months, the median overall survival had not been reached in the nivolumab‐plus‐ipilimumab group and was 37.6 months in the nivolumab group, as compared with 19.9 months in the ipilimumab group (hazard ratio for death with nivolumab plus ipilimumab vs. ipilimumab, 0.55 [P<0.001]; hazard ratio for death with nivolumab vs. ipilimumab, 0.65 [P<0.001]). The overall survival rate at 3 years was 58% in the nivolumab‐plus‐ipilimumab group and 52% in the nivolumab group, as compared with 34% in the ipilimumab group. The safety profile was unchanged from the initial report. Treatment‐related adverse events of grade 3 or 4 occurred in 59% of the patients in the nivolumab‐plus‐ipilimumab group, in 21% of those in the nivolumab group, and in 28% of those in the ipilimumab group. CONCLUSIONS Among patients with advanced melanoma, significantly longer overall survival occurred with combination therapy with nivolumab plus ipilimumab or with nivolumab alone than with ipilimumab alone. (Funded by Bristol‐Myers Squibb and others; CheckMate 067 ClinicalTrials.gov number, NCT01844505.)


The Lancet Haematology | 2015

Non-invasive detection of genomic imbalances in Hodgkin/Reed-Sternberg cells in early and advanced stage Hodgkin's lymphoma by sequencing of circulating cell-free DNA: a technical proof-of-principle study

Peter Vandenberghe; Iwona Wlodarska; Thomas Tousseyn; Luc Dehaspe; Daan Dierickx; Magali Verheecke; Anne Uyttebroeck; Oliver Bechter; Michel Delforge; Vincent Vandecaveye; Nathalie Brison; Gregor Verhoef; Eric Legius; Frédéric Amant; Joris Vermeesch

BACKGROUND Hodgkins lymphoma is one of the most common lymphoid neoplasms in young adults, but the low abundance of neoplastic Hodgkin/Reed-Sternberg cells in the tumour hampers the elucidation of its pathogenesis, biology, and diversity. After an incidental observation that genomic aberrations known to occur in Hodgkins lymphoma were detectable in circulating cell-free DNA, this study was undertaken to investigate whether circulating cell-free DNA can be informative about genomic imbalances in Hodgkins lymphoma. METHODS We applied massive parallel sequencing to circulating cell-free DNA in a prospective study of patients with biopsy proven nodular sclerosis Hodgkins lymphoma. Genomic imbalances in Hodgkin/Reed-Sternberg cells were investigated by fluorescence in-situ hybridisation (FISH) on tumour specimens. FINDINGS By non-invasive prenatal testing, we observed several genomic imbalances in circulating cell-free DNA of a pregnant woman, who was subsequently diagnosed with early-stage nodular sclerosis Hodgkins lymphoma stage IIA during gestation. FISH on tumour tissue confirmed corresponding genomic imbalances in Hodgkin/Reed-Sternberg cells. We prospectively studied circulating cell-free DNA of nine nodular sclerosis Hodgkins lymphoma cases: eight at first diagnosis and one at first relapse. Seven patients had stage IIA disease and two had stage IVB disease. In eight, genomic imbalances were detected, including, among others, gain of chromosomes 2p and 9p, known to occur in Hodgkins lymphoma. These gains and losses in circulating cell-free DNA were extensively validated by FISH on Hodgkin/Reed-Sternberg cells in biopsy samples. Initiation of chemotherapy induced normalisation of circulating cell-free DNA profiles within 2-6 weeks. The cell cycle indicator Ki67 and cleaved caspase-3 were detected in Hodgkin/Reed-Sternberg cells by immunohistochemistry, suggesting high turnover of Hodgkin/Reed-Sternberg cells. INTERPRETATION In early and advanced stage nodular sclerosis Hodgkins lymphoma, genomic imbalances in Hodgkin/Reed-Sternberg cells can be identified by massive parallel sequencing of circulating cell-free DNA at diagnosis. The rapid normalisation of circulating cell-free DNA profiles on therapy initiation suggests a potential role for circulating cell-free DNA profiling in early response monitoring. This finding creates several new possibilities for exploring the diversity of Hodgkins lymphoma, and has potential implications for the future clinical development of biomarkers and precision therapy for this malignancy. FUNDING KU Leuven-University of Leuven and University Hospitals Leuven.


The Lancet | 2017

Brentuximab vedotin or physician's choice in CD30-positive cutaneous T-cell lymphoma (ALCANZA): an international, open-label, randomised, phase 3, multicentre trial

H. Miles Prince; Youn H. Kim; Steven M. Horwitz; Reinhard Dummer; Julia Scarisbrick; Pietro Quaglino; Pier Luigi Zinzani; Pascal Wolter; Jose A Sanches; Pablo L. Ortiz-Romero; Oleg E. Akilov; Larisa J. Geskin; Judith Trotman; Kerry Taylor; Stéphane Dalle; Michael Weichenthal; Jan Walewski; David E. Fisher; B. Dréno; Rudolf Stadler; Tatyana Feldman; Timothy M. Kuzel; Yinghui Wang; Maria Corinna Palanca-Wessels; Erin Zagadailov; William L. Trepicchio; Wenwen Zhang; Hui-Min Lin; Yi Liu; Dirk Huebner

BACKGROUND Cutaneous T-cell lymphomas are rare, generally incurable, and associated with reduced quality of life. Present systemic therapies rarely provide reliable and durable responses. We aimed to assess efficacy and safety of brentuximab vedotin versus conventional therapy for previously treated patients with CD30-positive cutaneous T-cell lymphomas. METHODS In this international, open-label, randomised, phase 3, multicentre trial, we enrolled adult patients with CD30-positive mycosis fungoides or primary cutaneous anaplastic large-cell lymphoma who had been previously treated. Patients were enrolled across 52 centres in 13 countries. Patients were randomly assigned (1:1) centrally by an interactive voice and web response system to receive intravenous brentuximab vedotin 1·8 mg/kg once every 3 weeks, for up to 16 3-week cycles, or physicians choice (oral methotrexate 5-50 mg once per week or oral bexarotene 300 mg/m2 once per day) for up to 48 weeks. The primary endpoint was the proportion of patients in the intention-to-treat population achieving an objective global response lasting at least 4 months per independent review facility. Safety analyses were done in all patients who received at least one dose of study drug. This trial was registered with ClinicalTrials.gov, number NCT01578499. FINDINGS Between Aug 13, 2012, and July 31, 2015, 131 patients were enrolled and randomly assigned to a group (66 to brentuximab vedotin and 65 to physicians choice), with 128 analysed in the intention-to-treat population (64 in each group). At a median follow-up of 22·9 months (95% CI 18·4-26·1), the proportion of patients achieving an objective global response lasting at least 4 months was 56·3% (36 of 64 patients) with brentuximab vedotin versus 12·5% (eight of 64) with physicians choice, resulting in a between-group difference of 43·8% (95% CI 29·1-58·4; p<0·0001). Grade 3-4 adverse events were reported in 27 (41%) of 66 patients in the brentuximab vedotin group and 29 (47%) of 62 patients in the physicians choice group. Peripheral neuropathy was seen in 44 (67%) of 66 patients in the brentuximab vedotin group (n=21 grade 2, n=6 grade 3) and four (6%) of 62 patients in the physicians choice group. One of the four on-treatment deaths was deemed by the investigator to be treatment-related in the brentuximab vedotin group; no on-treatment deaths were reported in the physicians choice group. INTERPRETATION Significant improvement in objective response lasting at least 4 months was seen with brentuximab vedotin versus physicians choice of methotrexate or bexarotene. FUNDING Millennium Pharmaceuticals Inc (a wholly owned subsidiary of Takeda Pharmaceutical Company Ltd), Seattle Genetics Inc.


Oncotarget | 2016

The impact of adjuvant chemotherapy in older breast cancer patients on clinical and biological aging parameters

Barbara Brouwers; Sigrid Hatse; Lissandra Dal Lago; Patrick Neven; Peter Vuylsteke; Bruna Dalmasso; Guy Debrock; Heidi Van Den Bulck; Ann Smeets; Oliver Bechter; Jithendra Kini Bailur; Cindy Kenis; Annouschka Laenen; Patrick Schöffski; Graham Pawelec; Fabrice Journé; Ghanem Elias Ghanem; Hans Wildiers

Purpose This prospective observational study aimed to evaluate the impact of adjuvant chemotherapy on biological and clinical markers of aging and frailty. Methods Women ≥ 70 years old with early breast cancer were enrolled after surgery and assigned to a chemotherapy (Docetaxel and Cyclophosphamide) group (CTG, n=57) or control group (CG, n=52) depending on their planned adjuvant treatment. Full geriatric assessment (GA) and Quality of Life (QoL) were evaluated at inclusion (T0), after 3 months (T1) and at 1 year (T2). Blood samples were collected to measure leukocyte telomere length (LTL), levels of interleukin-6 (IL-6) and other circulating markers potentially informative for aging and frailty: Interleukin-10 (IL-10), Tumor Necrosis Factor Alpha (TNF-α), Insulin-like Growth Factor 1 (IGF-1), Monocyte Chemotactic Protein 1 (MCP-1) and Regulated on Activation, Normal T cell Expressed and Secreted (RANTES). Results LTL decreased significantly but comparably in both groups, whereas IL-6 was unchanged at T2. However, IL-10, TNF-α, IGF-1 and MCP-1 suggested a minor biological aging effect of chemotherapy. Clinical frailty and QoL decreased at T1 in the CTG, but recovered at T2, while remaining stable in the CG. Conclusion Chemotherapy (TC) is unlikely to amplify clinical aging or induce frailty at 1 year. Accordingly, there is no impact on the most established aging biomarkers (LTL, IL-6).


JCI insight | 2016

Deep sequencing reveals microRNAs predictive of antiangiogenic drug response

Jesús García-Donas; Benoit Beuselinck; Lucía Inglada-Pérez; Osvaldo Graña; Patrick Schöffski; Agnieszka Wozniak; Oliver Bechter; María Apellániz-Ruiz; Luis J. Leandro-García; Emilio Esteban; Daniel Castellano; Aranzazu Gonzalez del Alba; Miguel Angel Climent; Susana Hernando; José Ángel Arranz; Manuel Morente; David G. Pisano; Mercedes Robledo; Cristina Rodríguez-Antona

The majority of metastatic renal cell carcinoma (RCC) patients are treated with tyrosine kinase inhibitors (TKI) in first-line treatment; however, a fraction are refractory to these antiangiogenic drugs. MicroRNAs (miRNAs) are regulatory molecules proven to be accurate biomarkers in cancer. Here, we identified miRNAs predictive of progressive disease under TKI treatment through deep sequencing of 74 metastatic clear cell RCC cases uniformly treated with these drugs. Twenty-nine miRNAs were differentially expressed in the tumors of patients who progressed under TKI therapy (P values from 6 × 10-9 to 3 × 10-3). Among 6 miRNAs selected for validation in an independent series, the most relevant associations corresponded to miR-1307-3p, miR-155-5p, and miR-221-3p (P = 4.6 × 10-3, 6.5 × 10-3, and 3.4 × 10-2, respectively). Furthermore, a 2 miRNA-based classifier discriminated individuals with progressive disease upon TKI treatment (AUC = 0.75, 95% CI, 0.64-0.85; P = 1.3 × 10-4) with better predictive value than clinicopathological risk factors commonly used. We also identified miRNAs significantly associated with progression-free survival and overall survival (P = 6.8 × 10-8 and 7.8 × 10-7 for top hits, respectively), and 7 overlapped with early progressive disease. In conclusion, this is the first miRNome comprehensive study, to our knowledge, that demonstrates a predictive value of miRNAs for TKI response and provides a new set of relevant markers that can help rationalize metastatic RCC treatment.


Melanoma Research | 2017

Dabrafenib plus trametinib rechallenge in four melanoma patients who previously progressed on this combination

Aljosja Rogiers; Pascal Wolter; Oliver Bechter

In unresectable or metastatic melanoma with a BRAF V600 mutation, combined BRAF/MEK targeted therapy improves clinical outcomes. Yet, disease progression because of acquired resistance occurs in the majority of patients. There is emerging evidence that resistance to BRAF-inhibitor-based targeted therapy can be reversible in some cases. We retrospectively analyzed four patients with BRAF-mutant stage IV cutaneous melanoma who were treated with dabrafenib plus trametinib and rechallenged with the same combination after previously experiencing progression. At initial treatment with dabrafenib plus trametinib, three patients achieved a partial response and one patient achieved a complete response. Progression-free survival varied from 9.9 to 24.3 (median 19.8) months. The targeted therapy-free interval ranged from 2.3 to 11.7 (median 8.8) months. At rechallenge, all four patients had a partial response, with progression-free survival ranging from 3.6 to 6.8 (median 5.2) months. Clinical benefit and a second radiological response can be obtained upon readministration of dabrafenib plus trametinib after previously acquiring resistance to this combination. A better understanding of the biological underpinnings of genomic and nongenomic mechanisms of resistance to BRAF-inhibitor-based targeted therapy is needed to identify patients who may benefit from this rechallenge approach.


Acta Clinica Belgica | 2018

Incidence of osteonecrosis of the jaw in patients with bone metastases treated sequentially with bisphosphonates and denosumab

Tine Loyson; Tom van Cann; Patrick Schöffski; Paul Clement; Oliver Bechter; Isabel Spriet; Ruxandra-Gabriela Coropciuc; Constantinus Politis; Raf Vandeweyer; Joseph Schoenaers; Herlinde Dumez; Patrick Berteloot; Patrick Neven; Kristiaan Nackaerts; Feng Jung Woei-A-Jin; Kevin Punie; Hans Wildiers; Benoit Beuselinck

Abstract Objectives: Osteonecrosis of the jaw (ONJ) is a serious adverse event of bone resorption inhibitors (BRIs), such as bisphosphonates and denosumab. Bisphosphonates and denosumab inhibit osteoclast function through different pharmacological effects and bisphosphonates are retained in bone for several months to years. Sequential treatment with bisphosphonates and denosumab might lead to an overlapping treatment effect, due to the addition of the effect of denosumab on the residual bisphosphonate effect. Therefore, the aim of our study was to investigate if switching from denosumab to bisphosphonates is associated with a higher incidence of ONJ. Methods: We retrospectively reviewed records of patients with solid tumors and bone metastases treated with denosumab after prior treatment with bisphosphonates at the University Hospitals Leuven (sequential group). Patients treated with denosumab or bisphosphonates alone were used as control groups. Results: We identified 110 patients sequentially treated with bisphosphonates and denosumab with a median total BRI exposure of 36 months (sequential group). Median bisphosphonates exposure was 16 months and median denosumab exposure was 13 months. About 299 patients were included in the bisphosphonates control group with a median bisphosphonate exposure 19 months. About 6.7% (20/299) of patients developed ONJ. About 240 patients were included in the denosumab control group with a median denosumab exposure 17.5 months. About 10.0% of patients (24/240) developed ONJ. In the sequential group, 15.5% of patients (17/110) developed ONJ. The incidence of ONJ was 1.8% (2/110), 6.3% (6/99), 4.9% (4/82), 5.6% (3/54), and 3.4% (1/29), respectively in the first, second, third, fourth, and fifth year of BRI exposure, an ONJ-incidence similar to ONJ-incidence in the denosumab control group. In a time-to-ONJ-analysis, the curves of the sequential group and the denosumab control group were overlapping. In the sequential group, most of the ONJs occurred in the first year of denosumab exposure and in a matched control group analysis, with correction for median BRI-exposure, ONJ cases tend to occur earlier in the sequential group compared to ONJ cases in the bisphosphonates group. Conclusion: Cancer patients with bone metastases treated with BRIs seem to have a slightly higher risk of ONJ early after switching from bisphosphonates to denosumab compared to patients remaining on bisphosphonates. Nevertheless, based on the global ONJ-incidence, the switch from bisphosphonates to denosumab can be considered as safe as an equivalent exposure to denosumab from the start on.


Virchows Archiv | 2017

Molecular genetic and immunotherapeutic targets in metastatic melanoma

C. Melis; Aljosja Rogiers; Oliver Bechter; Joost van den Oord

In recent years, melanoma treatment has radically changed with the emergence of targeted therapies and immunotherapies. Both have led to improved survival for patients with advanced or unresectable melanoma. Targeted therapies with BRAF inhibitors in the lead use the presence of activating driver mutations to inhibit tumour growth. Forty to 60% of melanomas harbour BRAF mutations, which makes them susceptible to treatment with BRAF and/or MEK inhibitors. In parallel, the development of immunotherapeutic agents has also expanded. These agents stimulate the endogenous immune system of the patient to eradicate cancer cells. Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and programmed death 1 (PD-1) resulted in durable responses in a subset of patients. An important issue with immunotherapy lies in the identification of patients who will benefit from treatment. In this review, we will discuss these recent developments in melanoma therapy and highlight the role of the pathologist in both types of treatment.


Cell | 2018

Toward Minimal Residual Disease-Directed Therapy in Melanoma.

Florian Rambow; Aljosja Rogiers; Oskar Marin-Bejar; Sara Aibar; Julia Femel; Michael Dewaele; Panagiotis Karras; Daniel Brown; Young Hwan Chang; Maria Debiec-Rychter; Carmen Adriaens; Enrico Radaelli; Pascal Wolter; Oliver Bechter; Reinhard Dummer; Mitchell P. Levesque; Adriano Piris; Dennie T. Frederick; Genevieve M. Boland; Keith T. Flaherty; Joost van den Oord; Thierry Voet; Stein Aerts; Amanda W. Lund; Jean-Christophe Marine

Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.


Cancer Imaging | 2014

Whole-body diffusion-weighted imaging for staging lymphoma: are apparent diffusion coefficient derived histogram parameters useful for lesion characterisation?

K. De Paepe; F De Keyzer; Pascal Wolter; Oliver Bechter; Ann Janssens; Daan Dierickx; Raymond Oyen; Gregor Verhoef; Vincent Vandecaveye

Methods Fifteen patients with histopathology proven lymphoma (11 Non-Hodgkin; 4 Hodgkin lymphomas) underwent WB-DWI using 2 b-values (0-1000 s/mm). On coronal reformatted b1000 WB-DWI images, regions of interest (ROI) were drawn semi-automatically on lymph nodes in all nodal stations (n=267) and in axial and appendicular bone regions (n=53). For each ROI, a histogram was constructed from which volume, mean(ADC), median (ADC), skewness(ADC), and kurtosis(ADC) were calculated. Mann-Whitney-U tests were performed to detect significant differences between malignant and benign ROIs per tissue type. Receiver-operating-characteristic curves (ROC) were constructed from which an optimal threshold was determined as well as sensitivity, specificity and accuracy. PET/CT plus bone marrow biopsy (BMB) served as reference standard.

Collaboration


Dive into the Oliver Bechter's collaboration.

Top Co-Authors

Avatar

Pascal Wolter

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Patrick Schöffski

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Aljosja Rogiers

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Benoit Beuselinck

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Herlinde Dumez

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Vincent Vandecaveye

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daan Dierickx

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Gregor Verhoef

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Hans Wildiers

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge