Network


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

Hotspot


Dive into the research topics where Peter J. M. Valk is active.

Publication


Featured researches published by Peter J. M. Valk.


Cancer Cell | 2010

Leukemic IDH1 and IDH2 Mutations Result in a Hypermethylation Phenotype, Disrupt TET2 Function, and Impair Hematopoietic Differentiation

Maria E. Figueroa; Omar Abdel-Wahab; Chao Lu; Patrick S. Ward; Jay Patel; Alan Shih; Yushan Li; Neha Bhagwat; Aparna Vasanthakumar; Hugo F. Fernandez; Martin S. Tallman; Zhuoxin Sun; Kristy L. Wolniak; Justine K. Peeters; Wei Liu; Sung E. Choe; Valeria Fantin; Elisabeth Paietta; Bob Löwenberg; Jonathan D. Licht; Lucy A. Godley; Ruud Delwel; Peter J. M. Valk; Craig B. Thompson; Ross L. Levine; Ari Melnick

Cancer-associated IDH mutations are characterized by neomorphic enzyme activity and resultant 2-hydroxyglutarate (2HG) production. Mutational and epigenetic profiling of a large acute myeloid leukemia (AML) patient cohort revealed that IDH1/2-mutant AMLs display global DNA hypermethylation and a specific hypermethylation signature. Furthermore, expression of 2HG-producing IDH alleles in cells induced global DNA hypermethylation. In the AML cohort, IDH1/2 mutations were mutually exclusive with mutations in the α-ketoglutarate-dependent enzyme TET2, and TET2 loss-of-function mutations were associated with similar epigenetic defects as IDH1/2 mutants. Consistent with these genetic and epigenetic data, expression of IDH mutants impaired TET2 catalytic function in cells. Finally, either expression of mutant IDH1/2 or Tet2 depletion impaired hematopoietic differentiation and increased stem/progenitor cell marker expression, suggesting a shared proleukemogenic effect.


Cancer Cell | 2010

DNA Methylation Signatures Identify Biologically Distinct Subtypes in Acute Myeloid Leukemia

Maria E. Figueroa; Sanne Lugthart; Yushan Li; Claudia Erpelinck-Verschueren; Xutao Deng; Paul J. Christos; Elizabeth D. Schifano; James G. Booth; Wim L.J. van Putten; Lucy Skrabanek; Fabien Campagne; Madhu Mazumdar; John M. Greally; Peter J. M. Valk; Bob Löwenberg; Ruud Delwel; Ari Melnick

We hypothesized that DNA methylation distributes into specific patterns in cancer cells, which reflect critical biological differences. We therefore examined the methylation profiles of 344 patients with acute myeloid leukemia (AML). Clustering of these patients by methylation data segregated patients into 16 groups. Five of these groups defined new AML subtypes that shared no other known feature. In addition, DNA methylation profiles segregated patients with CEBPA aberrations from other subtypes of leukemia, defined four epigenetically distinct forms of AML with NPM1 mutations, and showed that established AML1-ETO, CBFb-MYH11, and PML-RARA leukemia entities are associated with specific methylation profiles. We report a 15 gene methylation classifier predictive of overall survival in an independent patient cohort (p < 0.001, adjusted for known covariates).


Cell | 2014

A Single Oncogenic Enhancer Rearrangement Causes Concomitant EVI1 and GATA2 Deregulation in Leukemia

Stefan Gröschel; Mathijs A. Sanders; Remco M. Hoogenboezem; Elzo de Wit; Britta A.M. Bouwman; Claudia Erpelinck; V H J van der Velden; Marije Havermans; Roberto Avellino; Kirsten van Lom; Elwin Rombouts; Konstanze Döhner; H. Berna Beverloo; James E. Bradner; Hartmut Döhner; Bob Löwenberg; Peter J. M. Valk; Eric M. J. Bindels; Wouter de Laat; Ruud Delwel

Chromosomal rearrangements without gene fusions have been implicated in leukemogenesis by causing deregulation of proto-oncogenes via relocation of cryptic regulatory DNA elements. AML with inv(3)/t(3;3) is associated with aberrant expression of the stem-cell regulator EVI1. Applying functional genomics and genome-engineering, we demonstrate that both 3q rearrangements reposition a distal GATA2 enhancer to ectopically activate EVI1 and simultaneously confer GATA2 functional haploinsufficiency, previously identified as the cause of sporadic familial AML/MDS and MonoMac/Emberger syndromes. Genomic excision of the ectopic enhancer restored EVI1 silencing and led to growth inhibition and differentiation of AML cells, which could be replicated by pharmacologic BET inhibition. Our data show that structural rearrangements involving the chromosomal repositioning of a single enhancer can cause deregulation of two unrelated distal genes, with cancer as the outcome.


Blood | 2011

Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity.

Erdogan Taskesen; Lars Bullinger; Andrea Corbacioglu; Mathijs A. Sanders; Claudia Erpelinck; Bas J. Wouters; Sonja van der Poel-van de Luytgaarde; Jürgen Krauter; Arnold Ganser; Richard F. Schlenk; Bob Löwenberg; Ruud Delwel; Hartmut Döhner; Peter J. M. Valk; Konstanze Döhner

We evaluated concurrent gene mutations, clinical outcome, and gene expression signatures of CCAAT/enhancer binding protein alpha (CEBPA) double mutations (CEBPA(dm)) versus single mutations (CEBPA(sm)) in 1182 cytogenetically normal acute myeloid leukemia (AML) patients (16-60 years of age). We identified 151 (12.8%) patients with CEBPA mutations (91 CEBPA(dm) and 60 CEBPA(sm)). The incidence of germline mutations was 7% (5 of 71), including 3 C-terminal mutations. CEBPA(dm) patients had a lower frequency of concurrent mutations than CEBPA(sm) patients (P < .0001). Both, groups were associated with a favorable outcome compared with CEBPA(wt) (5-year overall survival [OS] 63% and 56% vs 39%; P < .0001 and P = .05, respectively). However, in multivariable analysis only CEBPA(dm) was a prognostic factor for favorable OS outcome (hazard ratio [HR] 0.36, P < .0001; event-free survival, HR 0.41, P < .0001; relapse-free survival, HR 0.55, P = .001). Outcome in CEBPA(sm) is dominated by concurrent NPM1 and/or FLT3 internal tandem duplication mutations. Unsupervised and supervised GEP analyses showed that CEBPA(dm) AML (n = 42), but not CEBPA(sm) AML (n = 18), expressed a unique gene signature. A 25-probe set prediction signature for CEBPA(dm) AML showed 100% sensitivity and specificity. Based on these findings, we propose that CEBPA(dm) should be clearly defined from CEBPA(sm) AML and considered as a separate entity in the classification of AML.


Blood | 2012

Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia

Ana Flávia Tibúrcio Ribeiro; Marta Pratcorona; Claudia Erpelinck-Verschueren; Veronika Rockova; Mathijs A. Sanders; Saman Abbas; Maria E. Figueroa; Annelieke Zeilemaker; Ari Melnick; Bob Löwenberg; Peter J. M. Valk; Ruud Delwel

The prevalence, the prognostic effect, and interaction with other molecular markers of DNMT3A mutations was studied in 415 patients with acute myeloid leukemia (AML) younger than 60 years. We show mutations in DNMT3A in 96 of 415 patients with newly diagnosed AML (23.1%). Univariate Cox regression analysis showed that patients with DNMT3A(mutant) AML show significantly worse overall survival (OS; P = .022; hazard ratio [HR], 1.38; 95% confidence interval [CI], 1.04-1.81), and relapse-free survival (RFS; P = .005; HR, 1.52; 95% CI, 1.13-2.05) than DNMT3A(wild-type) AMLs. In a multivariable analysis, DNMT3A mutations express independent unfavorable prognostic value for OS (P = .003; HR, 1.82; 95% CI, 1.2-2.7) and RFS (P < .001; HR, 2.2; 95% CI, 1.4-3.3). In a composite genotypic subset of cytogenetic intermediate-risk AML without FLT3-ITD and NPM1 mutations, this association is particularly evident (OS: P = .013; HR, 2.09; 95% CI, 1.16-3.77; RFS: P = .001; HR, 2.65; 95% CI, 1.48-4.89). The effect of DNMT3A mutations in human AML remains elusive, because DNMT3A(mutant) AMLs did not express a methylation or gene expression signature that discriminates them from patients with DNMT3A(wild-type) AML. We conclude that DNMT3A mutation status is an important factor to consider for risk stratification of patients with AML.


Blood | 2011

Integrative prognostic risk score in acute myeloid leukemia with normal karyotype.

Michael Heuser; Michael Morgan; Katharina Wagner; Kerstin Görlich; Anika Grosshennig; Iyas Hamwi; Felicitas Thol; Ewa Surdziel; Walter Fiedler; Michael Lübbert; Lothar Kanz; Christoph W. M. Reuter; Gerhard Heil; Ruud Delwel; Bob Löwenberg; Peter J. M. Valk; Jürgen Krauter; Arnold Ganser

To integrate available clinical and molecular information for cytogenetically normal acute myeloid leukemia (CN-AML) patients into one risk score, 275 CN-AML patients from multicenter treatment trials AML SHG Hannover 0199 and 0295 and 131 patients from HOVON/SAKK protocols as external controls were evaluated for mutations/polymorphisms in NPM1, FLT3, CEBPA, MLL, NRAS, IDH1/2, and WT1. Transcript levels were quantified for BAALC, ERG, EVI1, ID1, MN1, PRAME, and WT1. Integrative prognostic risk score (IPRS) was modeled in 181 patients based on age, white blood cell count, mutation status of NPM1, FLT3-ITD, CEBPA, single nucleotide polymorphism rs16754, and expression levels of BAALC, ERG, MN1, and WT1 to represent low, intermediate, and high risk of death. Complete remission (P = .005), relapse-free survival (RFS, P < .001), and overall survival (OS, P < .001) were significantly different for the 3 risk groups. In 2 independent validation cohorts of 94 and 131 patients, the IPRS predicted different OS (P < .001) and RFS (P < .001). High-risk patients with related donors had longer OS (P = .016) and RFS (P = .026) compared with patients without related donors. In contrast, intermediate-risk group patients with related donors had shorter OS (P = .003) and RFS (P = .05). Donor availability had no impact on outcome of patients in the low-risk group. Thus, the IPRS may improve consolidation treatment stratification in CN-AML patients. Study registered at www.clinicaltrials.gov as #NCT00209833.


Nature Medicine | 2016

Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia

Sheng Li; Francine E. Garrett-Bakelman; Stephen S. Chung; Mathijs A. Sanders; Todd Hricik; Franck Rapaport; Jay Patel; Richard Dillon; Priyanka Vijay; Anna L. Brown; Alexander E. Perl; Joy Cannon; Lars Bullinger; Selina M. Luger; Michael W. Becker; Ian D. Lewis; L. B. To; Ruud Delwel; Bob Löwenberg; Hartmut Döhner; Konstanze Döhner; Monica L. Guzman; Duane C. Hassane; Gail J. Roboz; David Grimwade; Peter J. M. Valk; Richard J. D'Andrea; Martin Carroll; Christopher Y. Park; Donna Neuberg

Genetic heterogeneity contributes to clinical outcome and progression of most tumors, but little is known about allelic diversity for epigenetic compartments, and almost no data exist for acute myeloid leukemia (AML). We examined epigenetic heterogeneity as assessed by cytosine methylation within defined genomic loci with four CpGs (epialleles), somatic mutations, and transcriptomes of AML patient samples at serial time points. We observed that epigenetic allele burden is linked to inferior outcome and varies considerably during disease progression. Epigenetic and genetic allelic burden and patterning followed different patterns and kinetics during disease progression. We observed a subset of AMLs with high epiallele and low somatic mutation burden at diagnosis, a subset with high somatic mutation and lower epiallele burdens at diagnosis, and a subset with a mixed profile, suggesting distinct modes of tumor heterogeneity. Genes linked to promoter-associated epiallele shifts during tumor progression showed increased single-cell transcriptional variance and differential expression, suggesting functional impact on gene regulation. Thus, genetic and epigenetic heterogeneity can occur with distinct kinetics likely to affect the biological and clinical features of tumors.


Blood | 2013

PBX3 is an important cofactor of HOXA9 in leukemogenesis

Zejuan Li; Zhiyu Zhang; Yuanyuan Li; Stephen Arnovitz; Ping Chen; Hao Huang; Xi Jiang; Gia Ming Hong; Rejani B. Kunjamma; Haomin Ren; Chunjiang He; Chong-Zhi Wang; Abdel G. Elkahloun; Peter J. M. Valk; Konstanze Döhner; Mary Beth Neilly; Lars Bullinger; Ruud Delwel; Bob Löwenberg; Paul Liu; Richard Morgan; Janet D. Rowley; Chun-Su Yuan; Jianjun Chen

Although PBX proteins are known to increase DNA-binding/transcriptional activity of HOX proteins through their direct binding, the functional importance of their interaction in leukemogenesis is unclear.We recently reported that overexpression of a 4-homeobox-gene signature (ie, PBX3/HOXA7/HOXA9/HOXA11) is an independent predictor of poor survival in patients with cytogenetically abnormal acute myeloid leukemia (CA-AML). Here we show that it is PBX3, but not PBX1 or PBX2, that is consistently coexpressed with HOXA9 in various subtypes of CA-AML, particularly MLL-rearranged AML, and thus appears as a potential pathologic cofactor of HOXA9 in CA-AML. We then show that depletion of endogenous Pbx3 expression by shRNA significantly inhibits MLL-fusion-mediated cell transformation, and coexpressed PBX3 exhibits a significantly synergistic effect with HOXA9 in promoting cell transformation in vitro and leukemogenesis in vivo. Furthermore, as a proof of concept, we show that a small peptide, namely HXR9, which was developed to specifically disrupt the interactions between HOX and PBX proteins, can selectively kill leukemic cells with overexpression of HOXA/PBX3 genes. Collectively, our data suggest that PBX3 is a critical cofactor of HOXA9 in leukemogenesis, and targeting their interaction is a feasible strategy to treat presently therapy resistant CA-AML (eg, MLL-rearranged leukemia) in which HOXA/PBX3 genes are overexpressed.


Blood | 2010

High VEGFC expression is associated with unique gene expression profiles and predicts adverse prognosis in pediatric and adult acute myeloid leukemia

Hendrik J. M. de Jonge; Peter J. M. Valk; Nic J. G. M. Veeger; Arja ter Elst; Monique L. den Boer; Jacqueline Cloos; Valerie de Haas; Marry M. van den Heuvel-Eibrink; Gertjan J. L. Kaspers; Christian M. Zwaan; Willem A. Kamps; Bob Löwenberg; Eveline S. J. M. de Bont

High VEGFC mRNA expression of acute myeloid leukemia (AML) blasts is related to increased in vitro and in vivo drug resistance. Prognostic significance of VEGFC on long-term outcome and its associated gene expression profiles remain to be defined. We studied effect of VEGFC on treatment outcome and investigated gene expression profiles associated with VEGFC using microarray data of 525 adult and 100 pediatric patients with AML. High VEGFC expression appeared strongly associated with reduced complete remission rate (P = .004), reduced overall and event-free survival (OS and EFS) in adult AML (P = .002 and P < .001, respectively). Multivariable analysis established high VEGFC as prognostic indicator independent of cytogenetic risk, FLT3-ITD, NPM1, CEBPA, age, and white blood cell count (P = .038 for OS; P = .006 for EFS). Also, in pediatric AML high VEGFC was related to reduced OS (P = .041). A unique series of differentially expressed genes was identified that distinguished AML with high VEGFC from AML with low VEGFC, that is, 331 up-regulated genes (representative of proliferation, vascular endothelial growth factor receptor activity, signal transduction) and 44 down-regulated genes (eg, related to apoptosis) consistent with a role in enhanced chemoresistance. In conclusion, high VEGFC predicts adverse long-term prognosis and provides prognostic information in addition to well-known prognostic factors.


Blood | 2009

Homing and invasiveness of MLL/ENL leukemic cells is regulated by MEF2C

Maike Schwieger; Andrea Schüler; Martin Forster; Afra Engelmann; Michael A. Arnold; Ruud Delwel; Peter J. M. Valk; Jürgen Löhler; Robert K. Slany; Eric N. Olson; Carol Stocking

Acute myelogenous leukemia is driven by leukemic stem cells (LSCs) generated by mutations that confer (or maintain) self-renewal potential coupled to an aberrant differentiation program. Using retroviral mutagenesis, we identified genes that generate LSCs in collaboration with genetic disruption of the gene encoding interferon response factor 8 (Irf8), which induces a myeloproliferation in vivo. Among the targeted genes, we identified Mef2c, encoding a MCM1-agamous-deficiens-serum response factor transcription factor, and confirmed that overexpression induced a myelomonocytic leukemia in cooperation with Irf8 deficiency. Strikingly, several of the genes identified in our screen have been reported to be up-regulated in the mixed-lineage leukemia (MLL) subtype. High MEF2C expression levels were confirmed in acute myelogenous leukemia patient samples with MLL gene disruptions, prompting an investigation of the causal interplay. Using a conditional mouse strain, we demonstrated that Mef2c deficiency does not impair the establishment or maintenance of LSCs generated in vitro by MLL/ENL fusion proteins; however, its loss led to compromised homing and invasiveness of the tumor cells. Mef2c-dependent targets included several genes encoding matrix metalloproteinases and chemokine ligands and receptors, providing a mechanistic link to increased homing and motility. Thus, MEF2C up-regulation may be responsible for the aggressive nature of this leukemia subtype.

Collaboration


Dive into the Peter J. M. Valk's collaboration.

Top Co-Authors

Avatar

Bob Löwenberg

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ruud Delwel

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mathijs A. Sanders

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gert J. Ossenkoppele

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jan J. Cornelissen

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

François G. Kavelaars

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annelieke Zeilemaker

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ivo P. Touw

Erasmus University Rotterdam

View shared research outputs
Researchain Logo
Decentralizing Knowledge