Network


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

Hotspot


Dive into the research topics where Christian Flotho is active.

Publication


Featured researches published by Christian Flotho.


Leukemia | 2009

The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells

Christian Flotho; Rainer Claus; Christiane Batz; M Schneider; Inga Sandrock; S Ihde; Christoph Plass; Charlotte M. Niemeyer; Michael Lübbert

The three DNA methyltransferase (DNMT)-inhibiting cytosine nucleoside analogues, azacitidine, decitabine and zebularine, which are currently studied as nonintensive therapy for myelodysplastic syndromes and acute myeloid leukemia (AML), differ in structure and metabolism, suggesting that they may have differential molecular activity. We investigated cellular and molecular effects of the three substances relative to cytarabine in Kasumi-1 AML blasts. Under in vitro conditions mimicking those used in clinical trials, the DNMT inhibitors inhibited proliferation and triggered apoptosis but did not induce myeloid differentiation. The DNMT inhibitors showed no interference with cell-cycle progression whereas cytarabine treatment resulted in an S-phase arrest. Quantitative methylation analysis of hypermethylated gene promoters and of genome-wide LINE1 fragments using bisulfite sequencing and MassARRAY suggested that the hypomethylating potency of decitabine was stronger than that of azacitidine; zebularine showed no hypomethylating activity. In a comparative gene expression analysis, we found that the effects of each DNMT inhibitor on gene transcription were surprisingly different, involving several genes relevant to leukemogenesis. In addition, the gene methylation and expression analyses suggested that the effects of DNMT-inhibiting cytosine nucleoside analogues on the cellular transcriptome may, in part, be unrelated to direct promoter DNA hypomethylation, as previously shown by others.


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.


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.


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.


Blood | 2008

Genotype-phenotype correlation in cases of juvenile myelomonocytic leukemia with clonal RAS mutations

Christian Flotho; Christian P. Kratz; Eva Bergsträsser; Henrik Hasle; Jan Starý; Monika Trebo; Marry M. van den Heuvel-Eibrink; Dorota Wojcik; Marco Zecca; Franco Locatelli; Charlotte M. Niemeyer

To the editor: In a recent issue of Blood , Matsuda et al reported 11 children with juvenile myelomonocytic leukemia (JMML) and clonal NRAS or KRAS mutations.[1][1] Three patients showed improvement of various clinical and laboratory features over a 2- to 4-year period without chemotherapy or


Blood | 2009

Intriguing response to azacitidine in a patient with juvenile myelomonocytic leukemia and monosomy 7

Ingrid Furlan; Christiane Batz; Christian Flotho; Brigitte Mohr; Michael Lübbert; Meinolf Suttorp; Charlotte M. Niemeyer

To the editor: In this journal and others, Ruter[1][1] and Raj[2][2] have previously reported superior response to treatment with DNA-hypomethylating agents, such as decitabine or azacitidine (AC), in adults with myelodysplastic syndrome (MDS) and clonal monosomy 7. The use of AC has not yet been


Haematologica | 2010

Mitotic recombination and compound-heterozygous mutations are predominant NF1-inactivating mechanisms in children with juvenile myelomonocytic leukemia and neurofibromatosis type 1.

Doris Steinemann; Larissa Arning; Inka Praulich; Manfred Stuhrmann; Henrik Hasle; Jan Stary; Brigitte Schlegelberger; Charlotte M. Niemeyer; Christian Flotho

Children with neurofibromatosis type 1 (NF-1), being constitutionally deficient for one allele of the NF1 gene, are at greatly increased risk of juvenile myelomonocytic leukemia (JMML). NF1 is a negative regulator of RAS pathway activity, which has a central role in JMML. To further clarify the role of biallelic NF1 gene inactivation in the pathogenesis of JMML, we investigated the somatic NF1 lesion in 10 samples from children with JMML/NF-1. We report that two-thirds of somatic events involved loss of heterozygosity (LOH) at the NF1 locus, predominantly caused by segmental uniparental disomy of large parts of chromosome arm 17q. One-third of leukemias showed compound-heterozygous NF1-inactivating mutations. A minority of cases exhibited somatic interstitial deletions. The findings reinforce the emerging role of somatic mitotic recombination as a leukemogenic mechanism. In addition, they support the concept that biallelic NF1 inactivation in hematopoietic progenitor cells is required for transformation to JMML in children with NF-1.


Current Drug Targets | 2007

Targeting RAS Signaling Pathways in Juvenile Myelomonocytic Leukemia

Christian Flotho; Christian P. Kratz; Charlotte M. Niemeyer

The RAS proteins function as fundamental signaling switches that control normal cell growth and differentiation. Deregulated activation of RAS-dependent signaling pathways constitutes a potent mechanism of malignant cell transformation. Juvenile myelomonocytic leukemia (JMML) is a rapidly fatal myeloproliferative disorder of early childhood for which no effective treatment other than hematopoietic stem cell transplantation is currently available. Many aspects of JMML pathobiology are linked to deregulated RAS signaling. Hence, targeting RAS or its interactors on a molecular level is a promising strategy in the development of novel rational therapies for this menacing disease. Here we give an overview of current concepts on the pathogenesis of JMML, present important aspects of cellular RAS biology that can be exploited for pharmacologic manipulation, and discuss mouse models that have greatly advanced our understanding of the role RAS plays in JMML. In addition, we review recent approaches to develop agents that interfere with the RAS network at the level of the granulocyte-macrophage colony-stimulating factor receptor, posttranslational RAS processing (prenylation and endoprotease cleavage), RAF serine/threonine kinase, MEK mitogen-activated protein kinase, and target of rapamycin activity. Preclinical and clinical data of these pharmaceuticals in JMML and other myeloid malignancies is discussed.

Collaboration


Dive into the Christian Flotho'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

Jan Stary

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge