Network


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

Hotspot


Dive into the research topics where Barbara De Moerloose is active.

Publication


Featured researches published by Barbara De Moerloose.


Nature Genetics | 2010

PHF6 mutations in T-cell acute lymphoblastic leukemia

Pieter Van Vlierberghe; Teresa Palomero; Hossein Khiabanian; Joni Van der Meulen; Mireia Castillo; Nadine Van Roy; Barbara De Moerloose; Jan Philippé; Sara González-García; María L. Toribio; Tom Taghon; Linda Zuurbier; Barbara Cauwelier; Christine J. Harrison; Claire Schwab; Markus Pisecker; Sabine Strehl; Anton W. Langerak; Jozef Gecz; Edwin Sonneveld; Rob Pieters; Elisabeth Paietta; Jacob M. Rowe; Peter H. Wiernik; Yves Benoit; Jean Soulier; Bruce Poppe; Xiaopan Yao; Carlos Cordon-Cardo; Jules P.P. Meijerink

Tumor suppressor genes on the X chromosome may skew the gender distribution of specific types of cancer. T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy with an increased incidence in males. In this study, we report the identification of inactivating mutations and deletions in the X-linked plant homeodomain finger 6 (PHF6) gene in 16% of pediatric and 38% of adult primary T-ALL samples. Notably, PHF6 mutations are almost exclusively found in T-ALL samples from male subjects. Mutational loss of PHF6 is importantly associated with leukemias driven by aberrant expression of the homeobox transcription factor oncogenes TLX1 and TLX3. Overall, these results identify PHF6 as a new X-linked tumor suppressor in T-ALL and point to a strong genetic interaction between PHF6 loss and aberrant expression of TLX transcription factors in the pathogenesis of this disease.


The New England Journal of Medicine | 2018

Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia

Shannon L. Maude; Theodore W. Laetsch; Jochen Buechner; Susana Rives; Michael Boyer; Henrique Bittencourt; Peter Bader; Michael R. Verneris; Heather E. Stefanski; Gary Douglas Myers; Muna Qayed; Barbara De Moerloose; Hidefumi Hiramatsu; Krysta Schlis; Kara L. Davis; Paul L. Martin; Eneida R. Nemecek; Gregory A. Yanik; Christina Peters; André Baruchel; Nicolas Boissel; Francoise Mechinaud; Adriana Balduzzi; Joerg Krueger; Carl H. June; Bruce L. Levine; Patricia A. Wood; Tetiana Taran; Mimi Leung; Karen Thudium Mueller

Background In a single‐center phase 1–2a study, the anti‐CD19 chimeric antigen receptor (CAR) T‐cell therapy tisagenlecleucel produced high rates of complete remission and was associated with serious but mainly reversible toxic effects in children and young adults with relapsed or refractory B‐cell acute lymphoblastic leukemia (ALL). Methods We conducted a phase 2, single‐cohort, 25‐center, global study of tisagenlecleucel in pediatric and young adult patients with CD19+ relapsed or refractory B‐cell ALL. The primary end point was the overall remission rate (the rate of complete remission or complete remission with incomplete hematologic recovery) within 3 months. Results For this planned analysis, 75 patients received an infusion of tisagenlecleucel and could be evaluated for efficacy. The overall remission rate within 3 months was 81%, with all patients who had a response to treatment found to be negative for minimal residual disease, as assessed by means of flow cytometry. The rates of event‐free survival and overall survival were 73% (95% confidence interval [CI], 60 to 82) and 90% (95% CI, 81 to 95), respectively, at 6 months and 50% (95% CI, 35 to 64) and 76% (95% CI, 63 to 86) at 12 months. The median duration of remission was not reached. Persistence of tisagenlecleucel in the blood was observed for as long as 20 months. Grade 3 or 4 adverse events that were suspected to be related to tisagenlecleucel occurred in 73% of patients. The cytokine release syndrome occurred in 77% of patients, 48% of whom received tocilizumab. Neurologic events occurred in 40% of patients and were managed with supportive care, and no cerebral edema was reported. Conclusions In this global study of CAR T‐cell therapy, a single infusion of tisagenlecleucel provided durable remission with long‐term persistence in pediatric and young adult patients with relapsed or refractory B‐cell ALL, with transient high‐grade toxic effects. (Funded by Novartis Pharmaceuticals; ClinicalTrials.gov number, NCT02435849.)


Blood | 2016

Prevalence, clinical characteristics, and prognosis of GATA2-related myelodysplastic syndromes in children and adolescents

Marcin W. Wlodarski; Shinsuke Hirabayashi; Victor Pastor; Jan Starý; Henrik Hasle; Riccardo Masetti; Michael Dworzak; M Schmugge; Marry M. van den Heuvel-Eibrink; Marek Ussowicz; Barbara De Moerloose; Albert Catala; Owen P. Smith; Petr Sedlacek; Arjan C. Lankester; Marco Zecca; Victoria Bordon; Susanne Matthes-Martin; Jonas Abrahamsson; Jörn Sven Kühl; Karl Walter Sykora; Michael H. Albert; Bartlomiej Przychodzien; Jaroslaw P. Maciejewski; Stephan Schwarz; Gudrun Göhring; Brigitte Schlegelberger; Annamaria Cseh; Peter Noellke; Ayami Yoshimi

Germline GATA2 mutations cause cellular deficiencies with high propensity for myeloid disease. We investigated 426 children and adolescents with primary myelodysplastic syndrome (MDS) and 82 cases with secondary MDS enrolled in 2 consecutive prospective studies of the European Working Group of MDS in Childhood (EWOG-MDS) conducted in Germany over a period of 15 years. Germline GATA2 mutations accounted for 15% of advanced and 7% of all primary MDS cases, but were absent in children with MDS secondary to therapy or acquired aplastic anemia. Mutation carriers were older at diagnosis and more likely to present with monosomy 7 and advanced disease compared with wild-type cases. For stratified analysis according to karyotype, 108 additional primary MDS patients registered with EWOG-MDS were studied. Overall, we identified 57 MDS patients with germline GATA2 mutations. GATA2 mutations were highly prevalent among patients with monosomy 7 (37%, all ages) reaching its peak in adolescence (72% of adolescents with monosomy 7). Unexpectedly, monocytosis was more frequent in GATA2-mutated patients. However, when adjusted for the selection bias from monosomy 7, mutational status had no effect on the hematologic phenotype. Finally, overall survival and outcome of hematopoietic stem cell transplantation (HSCT) were not influenced by mutational status. This study identifies GATA2 mutations as the most common germline defect predisposing to pediatric MDS with a very high prevalence in adolescents with monosomy 7. GATA2 mutations do not confer poor prognosis in childhood MDS. However, the high risk for progression to advanced disease must guide decision-making toward timely HSCT.


Blood | 2015

The H3K27me3 demethylase UTX is a gender-specific tumor suppressor in T-cell acute lymphoblastic leukemia

Joni Van der Meulen; Viraj Sanghvi; Konstantinos Mavrakis; Kaat Durinck; Fang Fang; Filip Matthijssens; Pieter Rondou; Monica Rosen; Tim Pieters; Peter Vandenberghe; Eric Delabesse; Tim Lammens; Barbara De Moerloose; Björn Menten; Nadine Van Roy; Bruno Verhasselt; Bruce Poppe; Yves Benoit; Tom Taghon; Ari Melnick; Franki Speleman; Hans-Guido Wendel; Pieter Van Vlierberghe

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive form of leukemia that is mainly diagnosed in children and shows a skewed gender distribution toward males. In this study, we report somatic loss-of-function mutations in the X-linked histone H3K27me3 demethylase ubiquitously transcribed X (UTX) chromosome, in human T-ALL. Interestingly, UTX mutations were exclusively present in male T-ALL patients and allelic expression analysis revealed that UTX escapes X-inactivation in female T-ALL lymphoblasts and normal T cells. Notably, we demonstrate in vitro and in vivo that the H3K27me3 demethylase UTX functions as a bona fide tumor suppressor in T-ALL. Moreover, T-ALL driven by UTX inactivation exhibits collateral sensitivity to pharmacologic H3K27me3 inhibition. All together, our results show how a gender-specific and therapeutically relevant defect in balancing H3K27 methylation contributes to T-cell leukemogenesis.


Blood | 2014

ABT-199 mediated inhibition of BCL-2 as a novel therapeutic strategy in T-cell acute lymphoblastic leukemia

Sofie Peirs; Filip Matthijssens; Steven Goossens; Inge Vande Walle; Katia Ruggero; Charles E. de Bock; Sandrine Degryse; Kirsten Canté-Barrett; Delphine Briot; Emmanuelle Clappier; Tim Lammens; Barbara De Moerloose; Yves Benoit; Bruce Poppe; Jules P.P. Meijerink; Jan Cools; Jean Soulier; Terence H. Rabbitts; Tom Taghon; Franki Speleman; Pieter Van Vlierberghe

T-cell acute lymphoblastic leukemia (T-ALL) is a high-risk subtype of acute lymphoblastic leukemia (ALL) with gradually improved survival through introduction of intensified chemotherapy. However, therapy-resistant or refractory T-ALL remains a major clinical challenge. Here, we evaluated B-cell lymphoma (BCL)-2 inhibition by the BH3 mimetic ABT-199 as a new therapeutic strategy in human T-ALL. The T-ALL cell line LOUCY, which shows a transcriptional program related to immature T-ALL, exhibited high in vitro and in vivo sensitivity for ABT-199 in correspondence with high levels of BCL-2. In addition, ABT-199 showed synergistic therapeutic effects with different chemotherapeutic agents including doxorubicin, l-asparaginase, and dexamethasone. Furthermore, in vitro analysis of primary patient samples indicated that some immature, TLX3- or HOXA-positive primary T-ALLs are highly sensitive to BCL-2 inhibition, whereas TAL1 driven tumors mostly showed poor ABT-199 responses. Because BCL-2 shows high expression in early T-cell precursors and gradually decreases during normal T-cell differentiation, differences in ABT-199 sensitivity could partially be mediated by distinct stages of differentiation arrest between different molecular genetic subtypes of human T-ALL. In conclusion, our study highlights BCL-2 as an attractive molecular target in specific subtypes of human T-ALL that could be exploited by ABT-199.


Journal of Clinical Oncology | 2015

Collaborative Efforts Driving Progress in Pediatric Acute Myeloid Leukemia

C. Michel Zwaan; Edward A. Kolb; Dirk Reinhardt; Jonas Abrahamsson; Souichi Adachi; Richard Aplenc; Eveline S. J. M. de Bont; Barbara De Moerloose; Michael Dworzak; Brenda Gibson; Henrik Hasle; Guy Leverger; Franco Locatelli; Christine Ragu; Raul C. Ribeiro; Carmelo Rizzari; Jeffrey E. Rubnitz; Owen P. Smith; Lillian Sung; Daisuke Tomizawa; Marry M. van den Heuvel-Eibrink; Ursula Creutzig; Gertjan J. L. Kaspers

Diagnosis, treatment, response monitoring, and outcome of pediatric acute myeloid leukemia (AML) have made enormous progress during the past decades. Because AML is a rare type of childhood cancer, with an incidence of approximately seven occurrences per 1 million children annually, national and international collaborative efforts have evolved. This overview describes these efforts and includes a summary of the history and contributions of each of the main collaborative pediatric AML groups worldwide. The focus is on translational and clinical research, which includes past, current, and future clinical trials. Separate sections concern acute promyelocytic leukemia, myeloid leukemia of Down syndrome, and relapsed AML. A plethora of novel antileukemic agents that have emerged, including new classes of drugs, are summarized as well. Finally, an important aspect of the treatment of pediatric AML--supportive care--and late effects are discussed. The future is bright, with a wide range of emerging innovative therapies and with more and more international collaboration that ultimately aim to cure all children with AML, with fewer adverse effects and without late effects.


Blood | 2011

Aberrant DNA methylation characterizes juvenile myelomonocytic leukemia with poor outcome

Christiane Olk-Batz; Anna R. Poetsch; Peter Nöllke; Rainer Claus; Manuela Zucknick; Inga Sandrock; Tania Witte; Brigitte Strahm; Henrik Hasle; Marco Zecca; Jan Stary; Eva Bergstraesser; Barbara De Moerloose; Monika Trebo; Marry M. van den Heuvel-Eibrink; Dorota Wojcik; Franco Locatelli; Christoph Plass; Charlotte M. Niemeyer; Christian Flotho

Aberrant DNA methylation contributes to the malignant phenotype in virtually all types of cancer, including myeloid leukemia. We hypothesized that CpG island hypermethylation also occurs in juvenile myelomonocytic leukemia (JMML) and investigated whether it is associated with clinical, hematologic, or prognostic features. Based on quantitative measurements of DNA methylation in 127 JMML cases using mass spectrometry (MassARRAY), we identified 4 gene CpG islands with frequent hypermethylation: BMP4 (36% of patients), CALCA (54%), CDKN2B (22%), and RARB (13%). Hypermethylation was significantly associated with poor prognosis: when the methylation data were transformed into prognostic scores using a LASSO Cox regression model, the 5-year overall survival was 0.41 for patients in the top tertile of scores versus 0.72 in the lowest score tertile (P = .002). Among patients given allogeneic hematopoietic stem cell transplantation, the 5-year cumulative incidence of relapse was 0.52 in the highest versus 0.10 in the lowest score tertile (P = .007). In multivariate models, DNA methylation retained prognostic value independently of other clinical risk factors. Longitudinal analyses indicated that some cases acquired a more extensively methylated phenotype at relapse. In conclusion, our data suggest that a high-methylation phenotype characterizes an aggressive biologic variant of JMML and is an important molecular predictor of outcome.


Leukemia | 2011

Hematopoietic stem cell transplantation for advanced myelodysplastic syndrome in children: results of the EWOG-MDS 98 study

Brigitte Strahm; Peter Nöllke; Marco Zecca; Elisabeth T. Korthof; Marc Bierings; Ingrid Furlan; Petr Sedlacek; A Chybicka; M Schmugge; Victoria Bordon; Christina Peters; Aengus O'Marcaigh; C D de Heredia; Eva Bergstraesser; Barbara De Moerloose; M.M. van den Heuvel-Eibrink; Jan Starý; Monika Trebo; Dorota Wojcik; Charlotte M. Niemeyer; F. Locatelli

We report on the outcome of children with advanced primary myelodysplastic syndrome (MDS) transplanted from an HLA-matched sibling (MSD) or an unrelated donor (UD) following a preparative regimen with busulfan, cyclophosphamide and melphalan. Ninety-seven patients with refractory anemia with excess blasts (RAEB, n=53), RAEB in transformation (RAEB-T, n=29) and myelodysplasia-related acute myeloid leukemia (MDR-AML, n=15) enrolled in the European Working Group of MDS in Childhood (EWOG-MDS) 98 study and given hematopoietic stem cell transplantation (HSCT) were analyzed. Median age at HSCT was 11.1 years (range 1.4–19.0). Thirty-nine children were transplanted from an MSD, whereas 58 were given the allograft from a UD (n=57) or alternative family donor (n=1). Stem cell source was bone marrow (n=69) or peripheral blood (n=28). With a median follow-up of 3.9 years (range 0.1–10.9), the 5-year probability of overall survival is 63%, while the 5-year cumulative incidence of transplantation-related mortality (TRM) and relapse is 21% each. Age at HSCT greater than 12 years, interval between diagnosis and HSCT longer than 4 months, and occurrence of acute or extensive chronic graft-versus-host disease were associated with increased TRM. The risk of relapse increased with more advanced disease. This study indicates that HSCT following a myeloablative preparative regimen offers a high probability of survival for children with advanced MDS.


Blood | 2012

Spliceosomal gene aberrations are rare, coexist with oncogenic mutations, and are unlikely to exert a driver effect in childhood MDS and JMML

Shinsuke Hirabayashi; Christian Flotho; Jessica Moetter; Michael Heuser; Henrik Hasle; Bernd Gruhn; Thomas Klingebiel; Felicitas Thol; Brigitte Schlegelberger; Irith Baumann; Brigitte Strahm; Jan Stary; Franco Locatelli; Marco Zecca; Eva Bergstraesser; Michael Dworzak; Marry M. van den Heuvel-Eibrink; Barbara De Moerloose; Seishi Ogawa; Charlotte M. Niemeyer; Marcin W. Wlodarski

Somatic mutations of the spliceosomal machinery occur frequently in adult patients with myelodysplastic syndrome (MDS). We resequenced SF3B1, U2AF35, and SRSF2 in 371 children with MDS or juvenile myelomonocytic leukemia. We found missense mutations in 2 juvenile myelomonocytic leukemia cases and in 1 child with systemic mastocytosis with MDS. In 1 juvenile myelomonocytic leukemia patient, the SRSF2 mutation that initially coexisted with an oncogenic NRAS mutation was absent at relapse, whereas the NRAS mutation persisted and a second, concomitant NRAS mutation later emerged. The patient with systemic mastocytosis and MDS carried both mutated U2AF35 and KIT in a single clone as confirmed by clonal sequencing. In the adult MDS patients sequenced for control purposes, we detected previously reported mutations in 7/30 and a novel SRSF2 deletion (c.284_307del) in 3 of 30 patients. These findings implicate that spliceosome mutations are rare in pediatric MDS and juvenile myelomonocytic leukemia and are unlikely to operate as driver mutations.


Journal of Clinical Oncology | 2010

Gene Expression–Based Classification As an Independent Predictor of Clinical Outcome in Juvenile Myelomonocytic Leukemia

Silvia Bresolin; Marco Zecca; Christian Flotho; Luca Trentin; Andrea Zangrando; Laura Sainati; Jan Stary; Barbara De Moerloose; Henrik Hasle; Charlotte M. Niemeyer; Geertruy te Kronnie; Franco Locatelli; Giuseppe Basso

PURPOSE Juvenile myelomonocytic leukemia (JMML) is a rare early childhood myelodysplastic/myeloproliferative disorder characterized by an aggressive clinical course. Age and hemoglobin F percentage at diagnosis have been reported to predict both survival and outcome after hematopoietic stem cell transplantation (HSCT). However, no genetic markers with prognostic relevance have been identified so far. We applied gene expression-based classification to JMML samples in order to identify prognostic categories related to clinical outcome. PATIENTS AND METHODS Samples of 44 patients with JMML were available for microarray gene expression analysis. A diagnostic classification (DC) model developed for leukemia and myelodysplastic syndrome classification was used to classify the specimens and identify prognostically relevant categories. Statistical analysis was performed to determine the prognostic value of the classification and the genes identifying prognostic categories were further analyzed through R software. RESULTS The samples could be divided into two major groups: 20 specimens were classified as acute myeloid leukemia (AML) -like and 20 samples as nonAML-like. Four patients could not be assigned to a unique class. The 10-year probability of survival after diagnosis of AML-like and nonAML-like patients was significantly different (7% v 74%; P = .0005). Similarly, the 10-year event-free survival after HSCT was 6% for AML-like and 63% for nonAML-like patients (P = .0010). CONCLUSION Gene expression-based classification identifies two groups of patients with JMML with distinct prognosis outperforming all known clinical parameters in terms of prognostic relevance. Gene expression-based classification could thus be prospectively used to guide clinical/therapeutic decisions.

Collaboration


Dive into the Barbara De Moerloose's collaboration.

Top Co-Authors

Avatar

Yves Benoit

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar

Tim Lammens

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Dworzak

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Philippé

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge