Network


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

Hotspot


Dive into the research topics where Marry M. van den Heuvel-Eibrink is active.

Publication


Featured researches published by Marry M. van den Heuvel-Eibrink.


Blood | 2009

Novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia: results of an international retrospective study

Brian V. Balgobind; Susana C. Raimondi; Jochen Harbott; Martin Zimmermann; Todd A. Alonzo; Anne Auvrignon; H. Berna Beverloo; Myron Chang; Ursula Creutzig; Michael Dworzak; Erik Forestier; Brenda Gibson; Henrik Hasle; Christine J. Harrison; Nyla A. Heerema; Gertjan J. L. Kaspers; Anna Leszl; Nathalia Litvinko; Luca Lo Nigro; Akira Morimoto; Christine Perot; Rob Pieters; Dirk Reinhardt; Jeffrey E. Rubnitz; Franklin O. Smith; Jan Stary; Irina Stasevich; Sabine Strehl; Takashi Taga; Daisuke Tomizawa

Translocations involving chromosome 11q23 frequently occur in pediatric acute myeloid leukemia (AML) and are associated with poor prognosis. In most cases, the MLL gene is involved, and more than 50 translocation partners have been described. Clinical outcome data of the 11q23-rearranged subgroups are scarce because most 11q23 series are too small for meaningful analysis of subgroups, although some studies suggest that patients with t(9;11)(p22;q23) have a more favorable prognosis. We retrospectively collected outcome data of 756 children with 11q23- or MLL-rearranged AML from 11 collaborative groups to identify differences in outcome based on translocation partners. All karyotypes were centrally reviewed before assigning patients to subgroups. The event-free survival of 11q23/MLL-rearranged pediatric AML at 5 years from diagnosis was 44% (+/- 5%), with large differences across subgroups (11% +/- 5% to 92% +/- 5%). Multivariate analysis identified the following subgroups as independent prognostic predictors: t(1;11)(q21;q23) (hazard ratio [HR] = 0.1, P = .004); t(6;11)(q27;q23) (HR = 2.2, P < .001); t(10;11)(p12;q23) (HR = 1.5, P = .005); and t(10;11)(p11.2;q23) (HR = 2.5, P = .005). We could not confirm the favorable prognosis of the t(9;11)(p22;q23) subgroup. We identified large differences in outcome within 11q23/MLL-rearranged pediatric AML and novel subgroups based on translocation partners that independently predict clinical outcome. Screening for these translocation partners is needed for accurate treatment stratification at diagnosis.


Blood | 2012

Diagnosis and management of acute myeloid leukemia in children and adolescents: recommendations from an international expert panel

Ursula Creutzig; Marry M. van den Heuvel-Eibrink; Brenda Gibson; Michael Dworzak; Souichi Adachi; Eveline S. J. M. de Bont; Jochen Harbott; Henrik Hasle; Donna L. Johnston; Akitoshi Kinoshita; Thomas Lehrnbecher; Guy Leverger; Ester Mejstrikova; Soheil Meshinchi; Andrea Pession; Susana C. Raimondi; Lillian Sung; Jan Stary; Christian M. Zwaan; Gertjan J. L. Kaspers; Dirk Reinhardt

Despite major improvements in outcome over the past decades, acute myeloid leukemia (AML) remains a life-threatening malignancy in children, with current survival rates of ∼70%. State-of-the-art recommendations in adult AML have recently been published in this journal by Döhner et al. The primary goal of an international expert panel of the International BFM Study Group AML Committee was to set standards for the management, diagnosis, response assessment, and treatment in childhood AML. This paper aims to discuss differences between childhood and adult AML, and to highlight recommendations that are specific to children. The particular relevance of new diagnostic and prognostic molecular markers in pediatric AML is presented. The general management of pediatric AML, the management of specific pediatric AML cohorts (such as infants) or subtypes of the disease occurring in children (such as Down syndrome related AML), as well as new therapeutic approaches, and the role of supportive care are discussed.


Nature Genetics | 2010

Germline CBL mutations cause developmental abnormalities and predispose to juvenile myelomonocytic leukemia

Charlotte M. Niemeyer; Michelle Kang; Danielle H. Shin; Ingrid Furlan; Miriam Erlacher; Nancy Bunin; Severa Bunda; Jerry Z. Finklestein; Kathleen M. Sakamoto; Thomas A. Gorr; Parinda A. Mehta; Irene Schmid; Gabriele Kropshofer; Selim Corbacioglu; Peter Lang; Christoph Klein; Paul-Gerhard Schlegel; Andrea Heinzmann; Michaela Schneider; Jan Starý; Marry M. van den Heuvel-Eibrink; Henrik Hasle; Franco Locatelli; Debbie Sakai; Sophie Archambeault; Leslie Chen; Ryan C. Russell; Stephanie S Sybingco; Michael Ohh; Benjamin S. Braun

CBL encodes a member of the Cbl family of proteins, which functions as an E3 ubiquitin ligase. We describe a dominant developmental disorder resulting from germline missense CBL mutations, which is characterized by impaired growth, developmental delay, cryptorchidism and a predisposition to juvenile myelomonocytic leukemia (JMML). Some individuals experienced spontaneous regression of their JMML but developed vasculitis later in life. Importantly, JMML specimens from affected children show loss of the normal CBL allele through acquired isodisomy. Consistent with these genetic data, the common p.371Y>H altered Cbl protein induces cytokine-independent growth and constitutive phosphorylation of ERK, AKT and S6 only in hematopoietic cells in which normal Cbl expression is reduced by RNA interference. We conclude that germline CBL mutations have developmental, tumorigenic and functional consequences that resemble disorders that are caused by hyperactive Ras/Raf/MEK/ERK signaling and include neurofibromatosis type 1, Noonan syndrome, Costello syndrome, cardiofaciocutaneous syndrome and Legius syndrome.


Blood | 2009

Mutations in CBL occur frequently in juvenile myelomonocytic leukemia

Mignon L. Loh; Debbie Sakai; Christian Flotho; Michelle Kang; Manfred Fliegauf; Sophie Archambeault; Charles G. Mullighan; Leslie Chen; Eva Bergstraesser; Carlos E. Bueso-Ramos; Peter D. Emanuel; Henrik Hasle; Jean-Pierre J. Issa; Marry M. van den Heuvel-Eibrink; Franco Locatelli; Jan Starý; Monica Trebo; Marcin W. Wlodarski; Marco Zecca; Kevin Shannon; Charlotte M. Niemeyer

Juvenile myelomonocytic leukemia is an aggressive myeloproliferative disorder characterized by malignant transformation in the hematopoietic stem cell compartment with proliferation of differentiated progeny. Seventy-five percent of patients harbor mutations in the NF1, NRAS, KRAS, or PTPN11 genes, which encode components of Ras signaling networks. Using single nucleotide polymorphism arrays, we identified a region of 11q isodisomy that contains the CBL gene in several JMML samples, and subsequently identified CBL mutations in 27 of 159 JMML samples. Thirteen of these mutations alter codon Y371. In this report, we also demonstrate that CBL and RAS/PTPN11 mutations were mutually exclusive in these patients. Moreover, the exclusivity of CBL mutations with respect to other Ras pathway-associated mutations indicates that CBL may have a role in deregulating this key pathway in JMML.


Lancet Oncology | 2009

Dexamethasone-based therapy for childhood acute lymphoblastic leukaemia : results of the prospective Dutch Childhood Oncology Group (DCOG) protocol ALL-9 (1997-2004)

Anjo J. P. Veerman; Willem A. Kamps; Henk van den Berg; Eva van den Berg; Jos P.M. Bökkerink; Marrie C. A. Bruin; Marry M. van den Heuvel-Eibrink; Carin M. Korbijn; Elisabeth T. Korthof; Karin van der Pal; Theo Stijnen; Margreet H. van Weel Sipman; J. Fransje van Weerden; Elisabeth R. van Wering; Anna van der Does-van den Berg

BACKGROUND A population-based cohort of children aged 1-18 years with acute lymphoblastic leukaemia (ALL) was treated with a dexamethasone-based protocol (Dutch Childhood Oncology Group [DCOG] ALL-9). We aimed to confirm the results of the most effective DCOG ALL protocol for non-high-risk (NHR) patients to date (ALL-6), compare results with ALL-7 and ALL-8, and study prognostic factors in a non-randomised setting. METHODS From Jan 1, 1997, until Nov 1, 2004, patients with ALL were treated according to the ALL-9 protocol in eight Dutch academic centres with their affiliated peripheral hospitals. Patients were stratified into NHR and high risk (HR) groups. HR criteria were white-blood-cell count of 50,000 cells per microL or more, T-cell phenotype, mediastinal mass, CNS or testicular involvement, and Philadelphia chromosome or MLL rearrangement; patients who did not fulfil these criteria were deemed to be NHR. The NHR group was treated with a three-drug induction (dexamethasone, vincristine, and asparaginase) for 6 weeks, medium-dose methotrexate for 3 weeks, then maintenance therapy. HR patients received a four-drug induction (as for the NHR patients plus daunorubicin) for 6 weeks, high-dose methotrexate for 8 weeks, and two intensification courses before receiving maintenance therapy. Triple intrathecal medication was given 13 times in NHR patients, 15 times in HR patients (17 times for patients with initial CNS involvement). No patient received cranial irradiation. Maintenance therapy was given until 109 weeks for all patients and consisted of mercaptopurine and methotrexate for 5 weeks, alternated with dexamethasone and vincristine for 2 weeks. Kaplan-Meier analysis was done on an intention-to-treat basis with event-free survival as the primary endpoint. This trial is registered at trialregister.nl, number NTR460/SNWLK-ALL-9. FINDINGS 859 patients were recruited to the study. Complete remission was achieved in 592 (98.5%) of the 601 patients in the NHR group and 250 (96.9%) of the 258 in the HR group. Five patients in the NHR group and four in the HR group died during induction. Median follow-up for patients alive was 72.2 (range 4.8-132.7) months as of August, 2008. 5-year event-free survival was 81% (SE 1%) in all patients: 84% (2%) in NHR patients, and 72% (3%) in HR patients. Isolated CNS relapses occurred in 22 (2.6%) of 842 patients. In a multivariate analysis, DNA index was the strongest predictor of outcome (<1.16 vs >or=1.16; relative risk 0.42, 95% CI 0.22-0.78), followed by age (1-9 vs >or=10 years; 2.23, 1.60-3.11) and white-blood-cell count (<50,000 vs >or=50,000 cells per microL; 1.60, 1.13-2.26). INTERPRETATION The overall results of the dexamethasone-based DCOG ALL-9 protocol are better than those of our previous Berlin-Frankfurt-Münster-based protocols ALL-7 and ALL-8. The results for NHR patients were achieved with high cumulative doses of dexamethasone and vincristine, but without the use of anthracyclines, etoposide, cyclophosphamide, or cranial irradiation, therefore minimising the risk of side-effects. FUNDING Dutch Health Insurers.


Blood | 2011

NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern

Iris H.I.M. Hollink; Marry M. van den Heuvel-Eibrink; Susan T.C.J.M. Arentsen-Peters; Marta Pratcorona; Saman Abbas; Jenny E. Kuipers; Janneke van Galen; H. Berna Beverloo; Edwin Sonneveld; Gertjan J. L. Kaspers; Jan Trka; André Baruchel; Martin Zimmermann; Ursula Creutzig; Dirk Reinhardt; Rob Pieters; C. Michel Zwaan

Translocations involving nucleoporin 98kD (NUP98) on chromosome 11p15 occur at relatively low frequency in acute myeloid leukemia (AML) but can be missed with routine karyotyping. In this study, high-resolution genome-wide copy number analyses revealed cryptic NUP98/NSD1 translocations in 3 of 92 cytogenetically normal (CN)-AML cases. To determine their exact frequency, we screened > 1000 well-characterized pediatric and adult AML cases using a NUP98/NSD1-specific RT-PCR. Twenty-three cases harbored the NUP98/NSD1 fusion, representing 16.1% of pediatric and 2.3% of adult CN-AML patients. NUP98/NSD1-positive AML cases had significantly higher white blood cell counts (median, 147 × 10⁹/L), more frequent FAB-M4/M5 morphology (in 63%), and more CN-AML (in 78%), FLT3/internal tandem duplication (in 91%) and WT1 mutations (in 45%) than NUP98/NSD1-negative cases. NUP98/NSD1 was mutually exclusive with all recurrent type-II aberrations. Importantly, NUP98/NSD1 was an independent predictor for poor prognosis; 4-year event-free survival was < 10% for both pediatric and adult NUP98/NSD1-positive AML patients. NUP98/NSD1-positive AML showed a characteristic HOX-gene expression pattern, distinct from, for example, MLL-rearranged AML, and the fusion protein was aberrantly localized in nuclear aggregates, providing insight into the leukemogenic pathways of these AMLs. Taken together, NUP98/NSD1 identifies a previously unrecognized group of young AML patients, with distinct characteristics and dismal prognosis, for whom new treatment strategies are urgently needed.


Medical and Pediatric Oncology | 2000

Bone mineral density, body composition, and height in long‐term survivors of acute lymphoblastic leukemia in childhood

Inge M. van der Sluis; Marry M. van den Heuvel-Eibrink; Karel Hählen; Eric P. Krenning; Sabine M.P.F. de Muinck Keizer-Schrama

BACKGROUND Childhood leukemia has increasing numbers of survivors, so more emphasis is being placed on long-term effects. The ALL-6 protocol of the Dutch Childhood Leukemia Study Group involved high-dose dexamethasone and methotrexate and no cranial irradiation. Therefore, we studied the long-term effects on bone mineral density (BMD), body composition, and growth in survivors of non-high-risk ALL treated with the ALL-6 protocol. PROCEDURE Twenty-three subjects (12.2-25.4 years) participated in this cross-sectional study. Mean follow-up was 9.6 years (range 7.9-11.4 years). BMD of lumbar spine (LS) and total body (TB) and body composition were measured by dual energy X-ray absorptiometry; results are expressed as standard deviation scores (SDS). Bone mineral apparent density (BMAD(LS)) was calculated to correct for bone size. A questionnaire was administered to determine physical activity, calcium intake, and medical history. RESULTS Mean SDS for BMD(LS), BMD(TB), and BMAD(LS) were normal. None of the subjects had BMD below -2 SDS; one subject had BMAD(LS) below -2 SDS. Mean SDS for lean body mass, percentage fat, and height were not significantly different from zero. Calcium intake correlated positively with BMD. Nine subjects reported traumatic fractures (eight during or shortly after therapy). CONCLUSIONS Ten years after ALL-6 treatment, no long-term side effects on height, BMD, or body composition were found in this small group of patients, despite high-dose dexamethasone and methotrexate. This study suggests that ALL treatment without cranial irradiation might not be associated with long-term side effects on growth and BMD.


Blood | 2008

Leukemia-associated NF1 inactivation in patients with pediatric T-ALL and AML lacking evidence for neurofibromatosis

Brian V. Balgobind; Pieter Van Vlierberghe; Ans van den Ouweland; H. Berna Beverloo; Joan N.R. Terlouw-Kromosoeto; Elisabeth R. van Wering; Dirk Reinhardt; Martin A. Horstmann; Gertjan J. L. Kaspers; Rob Pieters; C. Michel Zwaan; Marry M. van den Heuvel-Eibrink; Jules P.P. Meijerink

Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder caused by mutations in the NF1 gene. Patients with NF1 have a higher risk to develop juvenile myelomonocytic leukemia (JMML) with a possible progression toward acute myeloid leukemia (AML). In an oligo array comparative genomic hybridization-based screening of 103 patients with pediatric T-cell acute lymphoblastic leukemia (T-ALL) and 71 patients with MLL-rearranged AML, a recurrent cryptic deletion, del(17)(q11.2), was identified in 3 patients with T-ALL and 2 patients with MLL-rearranged AML. This deletion has previously been described as a microdeletion of the NF1 region in patients with NF1. However, our patients lacked clinical NF1 symptoms. Mutation analysis in 4 of these del(17)(q11.2)-positive patients revealed that mutations in the remaining NF1 allele were present in 3 patients, confirming its role as a tumor-suppressor gene in cancer. In addition, NF1 inactivation was confirmed at the RNA expression level in 3 patients tested. Since the NF1 protein is a negative regulator of the RAS pathway (RAS-GTPase activating protein), homozygous NF1 inactivation represent a novel type I mutation in pediatric MLL-rearranged AML and T-ALL with a predicted frequency that is less than 10%. NF1 inactivation may provide an additional proliferative signal toward the development of leukemia.


Pediatric Blood & Cancer | 2005

Chemotherapy and attentional dysfunction in survivors of childhood acute lymphoblastic leukemia: Effect of treatment intensity

Annemieke I. Buizer; Leo de Sonneville; Marry M. van den Heuvel-Eibrink; A. J. P. Veerman

Central nervous system (CNS) directed chemotherapy is replacing prophylactic cranial irradiation in treatment protocols for childhood acute lymphoblastic leukemia (ALL), mainly to reduce long‐term neuropsychological sequelae. We evaluated the effects of chemotherapy on attentional function in survivors of ALL and examined whether possible deficits are related to treatment intensity.


Blood | 2009

Clinical relevance of Wilms tumor 1 gene mutations in childhood acute myeloid leukemia

Iris H.I.M. Hollink; Marry M. van den Heuvel-Eibrink; Martin Zimmermann; Brian V. Balgobind; Susan T.C.J.M. Arentsen-Peters; Marielle Alders; Andre Willasch; Gertjan J. L. Kaspers; Jan Trka; André Baruchel; Siebold S.N. de Graaf; Ursula Creutzig; Rob Pieters; Dirk Reinhardt; C. Michel Zwaan

Wilms tumor 1 (WT1) mutations have recently been identified in approximately 10% of adult acute myeloid leukemia (AML) with normal cytogenetics (CN-AML) and are associated with poor outcome. Using array-based comparative genome hybridization in pediatric CN-AML samples, we detected a WT1 deletion in one sample. The other WT1 allele was mutated. This prompted us to further investigate the role of WT1 aberrations in childhood AML. Mutations were found in 35 of 298 (12%) diagnostic pediatric AML samples. In 19 of 35 (54%) samples, more than one WT1 aberration was found: 15 samples had 2 different mutations, 2 had a homozygous mutation, and 2 had a mutation plus a WT1 deletion. WT1 mutations clustered significantly in the CN-AML subgroup (22%; P < .001) and were associated with FLT3/ITD (43 vs 17%; P < .001). WT1 mutations conferred an independent poor prognostic significance (WT1 mutated vs wild-type patients: 5-year probability of overall survival [pOS] 35% vs 66%, P = .002; probability of event-free survival 22% vs 46%, P < .001; and cumulative incidence of relapse or regression 70% vs 44%, P < .001). Patients with both a WT1 mutation and a FLT3/ITD had a dismal prognosis (5-year pOS 21%). WT1 mutations occur at a significant rate in childhood AML and are a novel independent poor prognostic marker.

Collaboration


Dive into the Marry M. van den Heuvel-Eibrink's collaboration.

Top Co-Authors

Avatar

Rob Pieters

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

C. Michel Zwaan

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Stary

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

Valerie de Haas

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge