76 results on '"Annika Dufour"'
Search Results
2. P457: COMPLEX CARYOTYPE IS AN INDEPENDENT RISK FACTOR IN TP53-MUTATED AML
- Author
-
Christian Rausch, Maja Rothenberg-Thurley, Annika Dufour, Stephanie Schneider, Hanna Gittinger, Cristina Sauerland, Dennis Görlich, Utz Krug, Wolfgang E. Berdel, Bernhard E. Woermann, Wolfgang Hiddemann, Jan Braess, Michael von Bergwelt-Baildon, Karsten Spiekermann, Tobias Herold, and Klaus H. Metzeler
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. P434: MYELODYSPLASIA-RELATED MUTATIONS IN THE NEW ELN CLASSIFICATION: DOES CLONAL SIZE PLAY A ROLE?
- Author
-
Christian Rausch, Maja Rothenberg-Thurley, Annika Dufour, Stephanie Schneider, Hanna Gittinger, Cristina Sauerland, Dennis Görlich, Utz Krug, Wolfgang E. Berdel, Bernhard J. Woermann, Wolfgang Hiddemann, Jan Braess, Michael von Bergwelt-Baildon, Karsten Spiekermann, Tobias Herold, and Klaus H. Metzeler
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
4. Patients with spontaneous remission of high-risk MDS and AML show persistent preleukemic clonal hematopoiesis
- Author
-
Victoria V. Grunwald, Marcus Hentrich, Xaver Schiel, Annika Dufour, Stephanie Schneider, Michaela Neusser, Marion Subklewe, Michael Fiegl, Wolfgang Hiddemann, Karsten Spiekermann, Maja Rothenberg-Thurley, and Klaus H. Metzeler
- Subjects
Specialties of internal medicine ,RC581-951 - Published
- 2019
- Full Text
- View/download PDF
5. Combination of t(4;14), del(17p13), del(1p32) and 1q21 gain FISH probes identifies clonal heterogeneity and enhances the detection of adverse cytogenetic profiles in 233 newly diagnosed multiple myeloma
- Author
-
Thomas Smol, Annika Dufour, Sabine Tricot, Mathieu Wemeau, Laure Stalnikiewicz, Franck Bernardi, Christine Terré, Benoît Ducourneau, Hervé Bisiau, and Agnès Daudignon
- Subjects
Multiple Myeloma ,Interphase FISH ,1q21 gain ,1p32 deletion ,Thresholds ,Genetics ,QH426-470 - Abstract
Abstract Background Our aim was to set the FISH combination of del(17p13), t(4;14), 1q21 gain and del(1p32), four adverse cytogenetic factors rarely evaluated together, and compare our technical thresholds with those defined in the literature. Methods Two hundred thirty-three patients with MM at diagnosis were studied using FISH to target 4 unfavorable cytogenetic abnormalities: 17p13 deletion, t(4;14) translocation, 1p32 deletion and 1q21 gain. Technical thresholds were determined for each probe using isolated CD138-expressing PC from patients without MM. Results The FISH analysis identified abnormalities in 79.0% of patients. Del(17p13) was detected in 15.0% of cases, t(4;14) in 11.5%, 1q21 gain in 37.8% and del(1p32) in 8.7%. Adding 1p32/1q21 FISH probes has enabled us to identify adverse cytogenetic profiles in 39.0% of patients without del(17p13) or t(4;14). Clonal heterogeneity was observed in 51.1% of patients as well as an increase in the number of adverse abnormalities when related clones were greater than or equal to 2 (85.1% against 45.6%). Conclusion FISH allowed detecting accumulation of adverse abnormalities and clonal heterogeneity in MM with a combination of 4 probes. The impacts of these two parameters need to be evaluated, and could be included in future cytogenetic classifications.
- Published
- 2017
- Full Text
- View/download PDF
6. Young woman with mild bone marrow dysplasia, GATA2 and ASXL1 mutation treated with allogeneic hematopoietic stem cell transplantation
- Author
-
Anna Lübking, Sebastian Vosberg, Nikola P. Konstandin, Annika Dufour, Alexander Graf, Stefan Krebs, Helmut Blum, Axel Weber, Stig Lenhoff, Mats Ehinger, Karsten Spiekermann, Philipp A. Greif, and Jörg Cammenga
- Subjects
GATA2 mutation ,Myelodysplastic syndrome ,ASXL1 mutation ,Allogeneic hematopoietic stem cell transplantation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Heterozygous mutations in GATA2 underlie different syndromes, previously described as monocytopenia and mycobacterial avium complex infection (MonoMAC); dendritic cell, monocytes, B- and NK lymphocytes deficiency (DCML); lymphedema, deafness and myelodysplasia (Emberger syndrome) and familiar myelodysplastic syndrome/acute myeloid leukemia (MDS / AML). Onset and severity of clinical symptoms vary and preceding cytopenias are not always present. We describe a case of symptomatic DCML deficiency and rather discrete bone marrow findings due to GATA2 mutation. Exome sequencing revealed a somatic ASXL1 mutation and the patient underwent allogeneic stem cell transplantation successfully.
- Published
- 2015
- Full Text
- View/download PDF
7. Molecular response assessment by quantitative real-time polymerase chain reaction after induction therapy in NPM1-mutated patients identifies those at high risk of relapse
- Author
-
Max Hubmann, Thomas Köhnke, Eva Hoster, Stephanie Schneider, Annika Dufour, Evelyn Zellmeier, Michael Fiegl, Jan Braess, Stefan K. Bohlander, Marion Subklewe, Maria-Cristina Sauerland, Wolfgang E. Berdel, Thomas Büchner, Bernhard Wörmann, Wolfgang Hiddemann, and Karsten Spiekermann
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Monitoring minimal residual disease is an important way to identify patients with acute myeloid leukemia at high risk of relapse. In this study we investigated the prognostic potential of minimal residual disease monitoring by quantitative real-time polymerase chain reaction analysis of NPM1 mutations in patients treated in the AMLCG 1999, 2004 and 2008 trials. Minimal residual disease was monitored - in aplasia, after induction therapy, after consolidation therapy, and during follow-up - in 588 samples from 158 patients positive for NPM1 mutations A, B and D (with a sensitivity of 10−6). One hundred and twenty-seven patients (80.4%) achieved complete remission after induction therapy and, of these, 56 patients (44.1%) relapsed. At each checkpoint, minimal residual disease cut-offs were calculated. After induction therapy a cut-off NPM1 mutation ratio of 0.01 was associated with a high hazard ratio of 4.26 and the highest sensitivity of 76% for the prediction of relapse. This was reflected in a cumulative incidence of relapse after 2 years of 77.8% for patients with ratios above the cut-off versus 26.4% for those with ratios below the cut-off. In the favorable subgroup according to European LeukemiaNet, the cut-off after induction therapy also separated the cohort into two prognostic groups with a cumulative incidence of relapse of 76% versus 6% after 2 years. Our data demonstrate that in addition to pre-therapeutic factors, the course of minimal residual disease in an individual is an important prognostic factor and could be included in clinical trials for the guidance of post-remission therapy. The trials from which data were obtained were registered at www.clinicaltrials.gov (#NCT01382147, #NCT00266136) and at the European Leukemia Trial Registry (#LN_AMLINT2004_230).
- Published
- 2014
- Full Text
- View/download PDF
8. RUNX1 mutations in cytogenetically normal acute myeloid leukemia are associated with a poor prognosis and up-regulation of lymphoid genes
- Author
-
Philipp A. Greif, Nikola P. Konstandin, Klaus H. Metzeler, Tobias Herold, Zlatana Pasalic, Bianka Ksienzyk, Annika Dufour, Friederike Schneider, Stephanie Schneider, Purvi M. Kakadia, Jan Braess, Maria Cristina Sauerland, Wolfgang E. Berdel, Thomas Büchner, Bernhard J. Woermann, Wolfgang Hiddemann, Karsten Spiekermann, and Stefan K. Bohlander
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background The RUNX1 (AML1) gene is a frequent mutational target in myelodysplastic syndromes and acute myeloid leukemia. Previous studies suggested that RUNX1 mutations may have pathological and prognostic implications.Design and Methods We screened 93 patients with cytogenetically normal acute myeloid leukemia for RUNX1 mutations by capillary sequencing of genomic DNA. Mutation status was then correlated with clinical data and gene expression profiles.Results We found that 15 out of 93 (16.1%) patients with cytogenetically normal acute myeloid leukemia had RUNX1 mutations. Seventy-three patients were enrolled in the AMLCG-99 trial and carried ten RUNX1 mutations (13.7%). Among these 73 patients RUNX1 mutations were significantly associated with older age, male sex, absence of NPM1 mutations and presence of MLL-partial tandem duplications. Moreover, RUNX1-mutated patients had a lower complete remission rate (30% versus 73% P=0.01), lower relapse-free survival rate (3-year relapse-free survival 0% versus 30.4%; P=0.002) and lower overall survival rate (3-year overall survival 0% versus 34.4%; P
- Published
- 2012
- Full Text
- View/download PDF
9. Germline SNPs previously implicated as prognostic biomarkers do not associate with outcomes in intensively treated AML
- Author
-
Aarif M. N. Batcha, Nele Buckup, Stefanos A. Bamopoulos, Vindi Jurinovic, Maja Rothenberg-Thurley, Hanna Gittinger, Bianka Ksienzyk, Annika Dufour, Stephanie Schneider, Mika Kontro, Joseph Saad, Caroline A. Heckmann, Cristina Sauerland, Dennis Görlich, Wolfgang E. Berdel, Bernhard J. Wörmann, Utz Krug, Jan Braess, Ulrich Mansmann, Wolfgang Hiddemann, Karsten Spiekermann, Klaus H. Metzeler, and Tobias Herold
- Subjects
Hematology - Published
- 2023
10. Validation and refinement of the 2022 European LeukemiaNet genetic risk stratification of acute myeloid leukemia
- Author
-
Christian Rausch, Maja Rothenberg-Thurley, Annika Dufour, Stephanie Schneider, Hanna Gittinger, Cristina Sauerland, Dennis Görlich, Utz Krug, Wolfgang E. Berdel, Bernhard J. Woermann, Wolfgang Hiddemann, Jan Braess, Michael von Bergwelt-Baildon, Karsten Spiekermann, Tobias Herold, and Klaus H. Metzeler
- Subjects
Cancer Research ,Oncology ,Hematology - Abstract
The revised 2022 European LeukemiaNet (ELN) AML risk stratification system requires validation in large, homogeneously treated cohorts. We studied 1118 newly diagnosed AML patients (median age, 58 years; range, 18–86 years) who received cytarabine-based induction chemotherapy between 1999 and 2012 and compared ELN-2022 to the previous ELN-2017 risk classification. Key findings were validated in a cohort of 1160 mostly younger patients. ELN-2022 reclassified 15% of patients, 3% into more favorable, and 12% into more adverse risk groups. This was mainly driven by patients reclassified from intermediate- to adverse-risk based on additional myelodysplasia-related mutations being included as adverse-risk markers. These patients (n = 79) had significantly better outcomes than patients with other adverse-risk genotypes (5-year OS, 26% vs. 12%) and resembled the remaining intermediate-risk group. Overall, time-dependent ROC curves and Harrel’s C-index controlling for age, sex, and AML type (de novo vs. sAML/tAML) show slightly worse prognostic discrimination of ELN-2022 compared to ELN-2017 for OS. Further refinement of ELN-2022 without including additional genetic markers is possible, in particular by recognizing TP53-mutated patients with complex karyotypes as “very adverse”. In summary, the ELN-2022 risk classification identifies a larger group of adverse-risk patients at the cost of slightly reduced prognostic accuracy compared to ELN-2017.
- Published
- 2023
11. A clinically applicable gene expression–based score predicts resistance to induction treatment in acute myeloid leukemia
- Author
-
Bianka Ksienzyk, Cristina Sauerland, Maja Rothenberg-Thurley, Vindi Jurinovic, Christian Moser, Annika Dufour, Jörg Kumbrink, Utz Krug, Sabine Sagebiel-Kohler, Dennis Görlich, Tobias Herold, Philipp A. Greif, Ulrich Mansmann, Wolfgang E. Berdel, Karsten Spiekermann, Klaus H. Metzeler, Sebastian Vosberg, Wolfgang Hiddemann, Aarif M. N. Batcha, Jan Braess, and Stephanie Schneider
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid Neoplasia ,Multivariate analysis ,business.industry ,Gene Expression ,Myeloid leukemia ,Hematology ,Disease ,Prognosis ,Cohort Studies ,Clinical trial ,Cytogenetics ,Leukemia, Myeloid, Acute ,Genetic marker ,Internal medicine ,Gene expression ,Cohort ,Humans ,Medicine ,business ,INDUCTION TREATMENT - Abstract
Key Points Prediction of induction failure in AML is possible using cytogenetic data and a gene expression–based classifier.Integration of PS29MRC in the clinical routine or trials may be facilitated by gene expression analysis with the NanoString platform., Visual Abstract, Prediction of resistant disease at initial diagnosis of acute myeloid leukemia (AML) can be achieved with high accuracy using cytogenetic data and 29 gene expression markers (Predictive Score 29 Medical Research Council; PS29MRC). Our aim was to establish PS29MRC as a clinically usable assay by using the widely implemented NanoString platform and further validate the classifier in a more recently treated patient cohort. Analyses were performed on 351 patients with newly diagnosed AML intensively treated within the German AML Cooperative Group registry. As a continuous variable, PS29MRC performed best in predicting induction failure in comparison with previously published risk models. The classifier was strongly associated with overall survival. We were able to establish a previously defined cutoff that allows classifier dichotomization (PS29MRCdic). PS29MRCdic significantly identified induction failure with 59% sensitivity, 77% specificity, and 72% overall accuracy (odds ratio, 4.81; P = 4.15 × 10−10). PS29MRCdic was able to improve the European Leukemia Network 2017 (ELN-2017) risk classification within every category. The median overall survival with high PS29MRCdic was 1.8 years compared with 4.3 years for low-risk patients. In multivariate analysis including ELN-2017 and clinical and genetic markers, only age and PS29MRCdic were independent predictors of refractory disease. In patients aged ≥60 years, only PS29MRCdic remained as a significant variable. In summary, we confirmed PS29MRC as a valuable classifier to identify high-risk patients with AML. Risk classification can still be refined beyond ELN-2017, and predictive classifiers might facilitate clinical trials focusing on these high-risk patients with AML.
- Published
- 2021
12. Double Drop-Off Droplet Digital PCR
- Author
-
Frank Ziemann, Annika Dufour, Klaus H. Metzeler, Karsten Spiekermann, Simon A. Buerger, Christian Rausch, Michaela Neusser, Maja Rothenberg-Thurley, Sebastian Tschuri, and Stephanie Schneider
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,NPM1 ,business.industry ,Myeloid leukemia ,Computational biology ,Gene mutation ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Real-time polymerase chain reaction ,Cell-free fetal DNA ,030220 oncology & carcinogenesis ,Molecular Medicine ,Medicine ,Digital polymerase chain reaction ,business ,Gene - Abstract
In acute myeloid leukemia (AML), somatic gene mutations are important prognostic markers and increasingly constitute therapeutic targets. Therefore, robust, sensitive, and fast diagnostic assays are needed. Current techniques for mutation screening and quantification, including next-generation sequencing and quantitative PCR, each have weaknesses that leave a need for novel diagnostic tools. We established double drop-off digital droplet PCR (DDO-ddPCR) assays for gene mutations in NPM1, IDH2, and NRAS, which can detect and quantify diverse alterations at two nearby hotspot regions present in these genes. These assays can be used for mutation screening as well as quantification and sequential monitoring. The assays were validated against next-generation sequencing and existing ddPCR assays and achieved high concordance with an overall sensitivity comparable to conventional digital PCR. In addition, the feasibility of detecting and monitoring genetic alterations in peripheral blood cell-free DNA (cfDNA) of patients with AML by DDO-ddPCR was studied. cfDNA analysis was found to have similar sensitivity compared to quantitative PCR–based analysis of peripheral blood. Finally, the cfDNA-based digital PCR in several clinical scenarios was found to be useful in long-term monitoring of target-specific therapy, early response assessment during induction chemotherapy, and identification of mutations in patients with extramedullary disease. Thus, DDO-ddPCR–based cfDNA analysis may complement existing genetic tools for diagnosis and disease monitoring in AML.
- Published
- 2021
13. Hypophosphatasia: a genetic-based nosology and new insights in genotype-phenotype correlation
- Author
-
Christine Muti, Nathalie Rousseau, Etienne Mornet, Mihelaiti Guberto, Carole Charle, Fabienne Wallon, Christelle Domingues, Emmanuelle Benaloun, Brigitte Simon-Bouy, Agnès Taillandier, Annika Dufour, and Nicole Lavaud
- Subjects
Nosology ,Heterozygote ,Adolescent ,Genotype ,Hypophosphatasia ,Biology ,Article ,Cohort Studies ,03 medical and health sciences ,Pregnancy ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Allele ,Child ,Alleles ,Genetic Association Studies ,Genetics (clinical) ,Dominance (genetics) ,0303 health sciences ,030305 genetics & heredity ,Infant ,ALPL ,Alkaline Phosphatase ,medicine.disease ,Phenotype ,Child, Preschool ,Mutation ,Female ,Haploinsufficiency ,Founder effect - Abstract
Hypophosphatasia (HPP) is caused by pathogenic variants in the ALPL gene. There is a large continuum in the severity, ranging from a lethal perinatal form to dental issues. We analyzed a cohort of 424 HPP patients from European geographic origin or ancestry. Using 3D modeling and results of functional tests we classified ALPL pathogenic variants according to their dominant negative effect (DNE) and their severity. The cohort was described by the genotypes resulting from alleles s (severe recessive), Sd (severe dominant), and m (moderate). Many recurrent variants showed a regional anchor pointing out founder effects rather than multiple mutational events. Homozygosity was an aggravating factor of the severity and moderate alleles were rare both in number and frequency. Pathogenic variants with DNE were found in both recessive and dominant HPP. Sixty percent of the adults tested were heterozygous for a variant showing no DNE, suggesting another mechanism of dominance like haploinsufficiency. Adults with dominant HPP without DNE were found statistically less severely affected than adults with DNE variants. Adults with dominant HPP without DNE represent a new clinical entity mostly diagnosed from 2010s, characterized by nonspecific signs of HPP and low alkaline phosphatase, and for which a high prevalence is expected. In conclusion, the genetic composition of our cohort suggests a nosology with 3 clinical forms: severe HPP is recessive and rare, moderate HPP is recessive or dominant and more common, and mild HPP, characterized by low alkaline phosphatase and unspecific clinical signs, is dominantly inherited and very common.
- Published
- 2020
14. Double Drop-Off Droplet Digital PCR: A Novel, Versatile Tool for Mutation Screening and Residual Disease Monitoring in Acute Myeloid Leukemia Using Cellular or Cell-Free DNA
- Author
-
Christian, Rausch, Maja, Rothenberg-Thurley, Simon A, Buerger, Sebastian, Tschuri, Annika, Dufour, Michaela, Neusser, Stephanie, Schneider, Karsten, Spiekermann, Klaus H, Metzeler, and Frank, Ziemann
- Subjects
Leukemia, Myeloid, Acute ,Neoplasm, Residual ,Molecular Diagnostic Techniques ,Mutation ,Biomarkers, Tumor ,Disease Management ,Humans ,Reproducibility of Results ,DNA, Neoplasm ,Real-Time Polymerase Chain Reaction ,Cell-Free Nucleic Acids ,Sensitivity and Specificity - Abstract
In acute myeloid leukemia (AML), somatic gene mutations are important prognostic markers and increasingly constitute therapeutic targets. Therefore, robust, sensitive, and fast diagnostic assays are needed. Current techniques for mutation screening and quantification, including next-generation sequencing and quantitative PCR, each have weaknesses that leave a need for novel diagnostic tools. We established double drop-off digital droplet PCR (DDO-ddPCR) assays for gene mutations in NPM1, IDH2, and NRAS, which can detect and quantify diverse alterations at two nearby hotspot regions present in these genes. These assays can be used for mutation screening as well as quantification and sequential monitoring. The assays were validated against next-generation sequencing and existing ddPCR assays and achieved high concordance with an overall sensitivity comparable to conventional digital PCR. In addition, the feasibility of detecting and monitoring genetic alterations in peripheral blood cell-free DNA (cfDNA) of patients with AML by DDO-ddPCR was studied. cfDNA analysis was found to have similar sensitivity compared to quantitative PCR-based analysis of peripheral blood. Finally, the cfDNA-based digital PCR in several clinical scenarios was found to be useful in long-term monitoring of target-specific therapy, early response assessment during induction chemotherapy, and identification of mutations in patients with extramedullary disease. Thus, DDO-ddPCR-based cfDNA analysis may complement existing genetic tools for diagnosis and disease monitoring in AML.
- Published
- 2021
15. Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia
- Author
-
Stefan K. Bohlander, Stephanie Schneider, Bernhard Wörmann, Marion Subklewe, Utz Krug, Tobias Herold, Victoria V. Grunwald, Wolfgang Hiddemann, Klaus H. Metzeler, Dennis Goerlich, Maria Cristina Sauerland, Andreas Faldum, Hanna Janke, Philipp A. Greif, Michaela Neusser, Karsten Spiekermann, Nikola P. Konstandin, Bianka Ksienzyk, Jan Braess, Maja Rothenberg-Thurley, Wolfgang E. Berdel, and Annika Dufour
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,Risk Assessment ,Article ,Acute myeloid leukaemia ,Young Adult ,European LeukemiaNet ,Internal medicine ,Genetics research ,CEBPA ,Humans ,Medicine ,Cancer genetics ,Survival rate ,Aged ,Aged, 80 and over ,business.industry ,Myeloid leukemia ,Induction chemotherapy ,Induction Chemotherapy ,Hematology ,Middle Aged ,Translational research ,Prognosis ,medicine.disease ,Survival Rate ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,medicine.anatomical_structure ,Risk factors ,Oncology ,Mutation ,Cohort ,Female ,business - Abstract
The revised 2017 European LeukemiaNet (ELN) recommendations for genetic risk stratification of acute myeloid leukemia have been widely adopted, but have not yet been validated in large cohorts of AML patients. We studied 1116 newly diagnosed AML patients (age range, 18–86 years) who had received induction chemotherapy. Among 771 patients not selected by genetics, the ELN-2017 classification re-assigned 26.5% of patients into a more favorable or, more commonly, a more adverse-risk group compared with the ELN-2010 recommendations. Forty percent of the cohort, and 51% of patients ≥60 years, were classified as adverse-risk by ELN-2017. In 599 patients CEBPA mutations or inv(16) had particularly favorable outcomes, while patients with mutated TP53 and a complex karyotype had especially poor prognosis. DNMT3A mutations associated with inferior OS within each ELN-2017 risk group. Our results validate the prognostic significance of the revised ELN-2017 risk classification in AML patients receiving induction chemotherapy across a broad age range. Further refinement of the ELN-2017 risk classification is possible.
- Published
- 2020
16. Combination of t(4;14), del(17p13), del(1p32) and 1q21 gain FISH probes identifies clonal heterogeneity and enhances the detection of adverse cytogenetic profiles in 233 newly diagnosed multiple myeloma
- Author
-
Christine Terré, Annika Dufour, Benoît Ducourneau, Thomas Smol, Sabine Tricot, Hervé Bisiau, Mathieu Wemeau, Franck Bernardi, Agnès Daudignon, and Laure Stalnikiewicz
- Subjects
medicine.medical_specialty ,lcsh:QH426-470 ,Chromosomal translocation ,Newly diagnosed ,Biology ,Biochemistry ,Gastroenterology ,1q21 gain ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Genetics ,medicine ,1p32 deletion ,Molecular Biology ,Genetics (clinical) ,Multiple myeloma ,Research ,Biochemistry (medical) ,Cytogenetics ,Fish analysis ,medicine.disease ,Molecular biology ,lcsh:Genetics ,030220 oncology & carcinogenesis ,Interphase FISH ,Molecular Medicine ,%22">Fish ,Thresholds ,Multiple Myeloma ,030215 immunology - Abstract
Background Our aim was to set the FISH combination of del(17p13), t(4;14), 1q21 gain and del(1p32), four adverse cytogenetic factors rarely evaluated together, and compare our technical thresholds with those defined in the literature. Methods Two hundred thirty-three patients with MM at diagnosis were studied using FISH to target 4 unfavorable cytogenetic abnormalities: 17p13 deletion, t(4;14) translocation, 1p32 deletion and 1q21 gain. Technical thresholds were determined for each probe using isolated CD138-expressing PC from patients without MM. Results The FISH analysis identified abnormalities in 79.0% of patients. Del(17p13) was detected in 15.0% of cases, t(4;14) in 11.5%, 1q21 gain in 37.8% and del(1p32) in 8.7%. Adding 1p32/1q21 FISH probes has enabled us to identify adverse cytogenetic profiles in 39.0% of patients without del(17p13) or t(4;14). Clonal heterogeneity was observed in 51.1% of patients as well as an increase in the number of adverse abnormalities when related clones were greater than or equal to 2 (85.1% against 45.6%). Conclusion FISH allowed detecting accumulation of adverse abnormalities and clonal heterogeneity in MM with a combination of 4 probes. The impacts of these two parameters need to be evaluated, and could be included in future cytogenetic classifications.
- Published
- 2017
17. Acute Myeloid Leukemia with Isocitrate Dehydrogenases (IDH) 1 and 2 Mutations. a Real-World Study from the European IDH Research Group
- Author
-
Marcus Hentrich, Jordi Esteve, Matteo G. Della Porta, Mar Tormo, Sigrid Machherndl-Spandl, Ana Garrido, Sonia Matos, Annika Dufour, Olga Salamero, Jorge Sierra, Marta Pratcorona, Lisa Pleyer, Federico Lussana, Maria Carmo-Fonseca, Francesco Passamonti, Paola Fazi, Albertina Nunes, Sonja Heibl, Susana Vives, Christoph Sippel, Nicola Stefano Fracchiolla, Alfonso Piciocchi, Joana M. P. Desterro, Montserrat Arnan, Pamela Acha, Erica Travaglino, Aida Botelho de Sousa, Friedrich Stölzel, Maria Teresa Voso, Giulia Maggioni, Christian Thiede, Claudia Basilico, Cristina Astrid Tentori, Francesc Solé, Karsten Spiekermann, Marcos Lemos, Klaus H. Metzeler, Elisabetta Todisco, Bernhard Heilmeier, Jan Moritz Middeke, Marina Diaz, Valentina Mancini, and David Gallardo
- Subjects
Response rate (survey) ,education.field_of_study ,NPM1 ,medicine.medical_specialty ,IDH1 ,business.industry ,Immunology ,Population ,Myeloid leukemia ,Context (language use) ,Cell Biology ,Hematology ,Biochemistry ,IDH2 ,Isocitrate dehydrogenase ,Internal medicine ,Medicine ,business ,education - Abstract
Introduction. Mutations in genes encoding the metabolic enzymes isocitrate dehydrogenase (IDH) 1 and 2 are found in 10-20% of patients with acute myeloid leukemia (AML). Recently, IDH inhibitors have shown good clinical response in patient's refractory to standard treatments, providing evidence for a new treatment paradigm. Comprehensive real-world studies are needed to explore genotype-to-phenotype correlations and prognosis of IDH mutated AML, which may influence targeted treatment strategies. Patients. From a retrospective, European, real-word population (ClinicalTrials.gov Identifier: NCT04369287) we studied 477 IDH mutated patients and 954 IDH wild type patients matched for age, sex and type of treatment with a 1:2 ratio. Results. Median age of IDH mutated patients was 67 years; IDH1 mutations were found in 202 patients (89% carried R132 mutation), while IDH2 mutations were found in 275 cases (51% and 28% carried R140 and R172 mutations, respectively). At diagnosis, IDH mutated patients had lower neutrophil and higher platelet count and higher percentage of marrow blasts (P Considering cytogenetic risk according to ELN criteria, the great majority of IDH1 and IDH2 mutated patients had an intermediate cytogenetic risk (84% and 86%, respectively, P We then analysed the most common co-mutational patterns in IDH mutated patients. A total of 53% of IDH1 mutated patients carried NPM1 mutations (without FLT3 mutations), while the majority of IDH2 mutated patients had wild type NPM1 gene (P Median overall survival from diagnosis (OS) was 14 months for IDH1 mutated patients, 23 for IDH2 mutated patients and 19 for IDH wild type patients (P IDH mutated patients receiving hypomethylating agents (n=211) had a lower response rate vs. wild type patients (56% vs. 36% of treatment failure, respectively, P=.04), while no significant different probability of response to intensive chemotherapy was noticed. In patients who received allogeneic transplantation (n=345), IDH1 mutated patients shower higher relapse rate vs. wild type and IDH2 mutated patients (53% vs. 34%, P Conclusion. In a real world context, AML patients with IDH1 and 2 mutations have high marrow blasts percentage, frequently present normal karyotype and show specific co-mutational patterns with respect to NPM1, FLT3 and ASXL1 genes. IDH1 mutations were an independent predictor of unfavorable outcome with high rate of disease recurrence under currently available treatment options, and could be considered as an additional marker to improve personalized prognostic assessment within ELN risk groups. Dissection of prognosis of IDH mutated AML may influence targeted treatment strategies in clinical practice. Disclosures Voso: Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Heibl:Takeda: Honoraria; AOP orphan: Consultancy, Honoraria, Research Funding; BMS/celgene: Consultancy, Honoraria, Research Funding; novartis: Consultancy, Honoraria. Metzeler:Astellas: Honoraria; Daiichi Sankyo: Honoraria; Otsuka Pharma: Consultancy; Pfizer: Consultancy; Jazz Pharmaceuticals: Consultancy; Novartis: Consultancy; Celgene: Consultancy, Honoraria, Research Funding. Thiede:AgenDix GmbH: Other: Co-owner and CEO. Fracchiolla:ABBVIE: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accommodations, expenses; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accommodations, expenses, Speakers Bureau; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accommodations, expenses, Speakers Bureau; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accommodations, expenses, Speakers Bureau. Todisco:Jannsen, Abbvie, Jazz: Membership on an entity's Board of Directors or advisory committees. Passamonti:Novartis: Speakers Bureau; BMS: Speakers Bureau; Roche: Other: Support of parent study and funding of editorial support.
- Published
- 2020
18. PTPN11 mutations and Outcomes in Adult Patients with Acute Myeloid Leukemia
- Author
-
Annika Dufour, Tobias Herold, Dennis Görlich, Jan Braess, Bernhard Wörmann, Klaus H. Metzeler, Stephanie Schneider, Maja Rothenberg-Thurley, Karsten Spiekermann, Utz Krug, Maria Cristina Sauerland, Wolfgang Hiddemann, Marion Subklewe, and Wolfgang E. Berdel
- Subjects
Oncology ,medicine.medical_specialty ,NPM1 ,business.industry ,Immunology ,Hazard ratio ,Induction chemotherapy ,Myeloid leukemia ,Cell Biology ,Hematology ,Gene mutation ,medicine.disease ,Biochemistry ,PTPN11 ,Leukemia ,Internal medicine ,medicine ,Cytarabine ,business ,medicine.drug - Abstract
Background: Mutations in the protein tyrosine phosphatase gene PTPN11 (also known as SHP2) are found in approximately 10% of adult patients with acute myeloid leukemia (AML). A recent study reported that mutated PTPN11 associates with inferior response rates and shorter survival among intensively treated AML patients, independently of the ELN prognostic groups (Alfayez et al., Leukemia 2020). Earlier analyses of the genomic landscape of AML did not uncover a similar prognostic relevance of PTPN11 mutations. Therefore, our aim was to clarify the prognostic relevance of mutated PTPN11 variants in AML patients receiving intensive front-line therapy. Patients and Methods: We studied 1116 AML patients enrolled on two subsequent multicenter phase III trials of the German AML Cooperative Group (AML-CG 1999, NCT00266136; and AML-CG 2008, NCT01382147) who were genetically characterized by amplicon-based targeted next-generation sequencing (Herold et al., Leukemia 2020). All patients had received induction chemotherapy containing cytarabine and daunorubicin or mitoxantrone. Results: We identified 146 PTPN11 mutations in 114 of 1116 patients (10%). Mutations clustered in two hotspot regions (5': codons 52-79; n=108 and 3': codons 491-512, n=38) as previously reported. Associations of PTPN11 mutations with baseline clinical and genetic patient characteristics are shown in Figure A. PTPN11 mutations were most frequent in the European LeukemiaNet (ELN) "favorable" genetic risk group, and associated with higher leukocyte counts. Patients with mutated PTPN11more commonly had mutated NPM1, IDH1 and DNMT3A, and less frequently had FLT3-ITD, IDH2 and TP53 mutations, compared to patients with wild-type PTPN11. With regard to treatment outcomes, the rate of complete remission was similar among patients with mutated and wild-type PTPN11 (65% vs. 59%, P=.25). In univariate analyses, PTPN11-mutated patients had significantly longer relapse-free survival (RFS; 5-year estimate, 55% vs 33% for PTPN11-wild type patients; P=.001; Figure B) and tended to have longer overall survival (OS; 5-year estimate, 43% vs 32%; P=.06; Figure C). However, in multivariable models adjusting for age, sex, leukocyte count, AML type (de novo/sAML/tAML) and ELN-2017 genetic risk group, mutated PTPN11 no longer associated with RFS (hazard ratio [HR], 0.89, 95% confidence interval [CI], 0.63 - 1.27; P=0.53) or OS (HR, 1.03; 95% CI, 0.80 - 1.33; P=.79). Moreover, PTPN11 mutations did not significantly associate with RFS or OS within any of the ELN genetic risk groups. Finally, we detected no significant differences in baseline characteristics or outcomes between patients with PTPN11 mutations affecting the 5' hotspot region (n=82), the 3' hotspot region (n=21), or mutations at both hotspots (n=11). Conclusion: In our cohort of newly diagnosed and intensively treated AML patients, mutations in PTPN11 occurred in 10% and associated with prognostically favorable genetic characteristics such as mutated NPM1 and absence of FLT3-ITD and TP53mutations. Consequently, PTPN11 mutations were most commonly found within the ELN-2017 favorable risk category. While patients with PTPN11 mutations had relatively favorable survival outcomes, multivariable models suggest this observation is confounded by the frequent co-occurrence of known favorable genetic markers. Our data are in disagreement with a recently published study on 880 newly diagnosed patients that found an unfavourable prognostic impact of mutated PTPN11, particularly among the 410 patients who received intensive treatment. Possible explanations for these discrepant results include differences in treatment regimens between the two cohorts, as well as the play of chance when studying a relatively rare gene mutation in medium-sized cohorts. In summary, our data do not support a role of PTPN11 mutations as an adverse prognostic biomarker in newly diagnosed, intensively treated adult AML patients. Figure Disclosures Metzeler: Daiichi Sankyo: Honoraria; Otsuka Pharma: Consultancy; Pfizer: Consultancy; Celgene: Consultancy, Honoraria, Research Funding; Novartis: Consultancy; Jazz Pharmaceuticals: Consultancy; Astellas: Honoraria. Subklewe:AMGEN: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Novartis: Consultancy, Research Funding; Janssen: Consultancy; Morphosys: Research Funding; Seattle Genetics: Research Funding; Roche AG: Consultancy, Research Funding; Gilead Sciences: Consultancy, Honoraria, Research Funding.
- Published
- 2020
19. Patients with spontaneous remission of high-risk MDS and AML show persistent preleukemic clonal hematopoiesis
- Author
-
Xaver Schiel, Klaus H. Metzeler, Maja Rothenberg-Thurley, Marion Subklewe, Annika Dufour, Stephanie Schneider, Victoria V. Grunwald, Karsten Spiekermann, Michael Fiegl, Wolfgang Hiddemann, Michaela Neusser, and Marcus Hentrich
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Myeloid ,business.industry ,Clonal hematopoiesis ,Spontaneous remission ,Hematology ,Disease ,Middle Aged ,medicine.disease ,Hematopoiesis ,Leukemia ,Haematopoiesis ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Internal medicine ,hemic and lymphatic diseases ,Myelodysplastic Syndromes ,Medicine ,Humans ,Exceptional Case Report ,business ,Aged - Abstract
Key Points We report longitudinal mutational analyses of 2 patients with high-risk MDS and AML experiencing spontaneous disease remissions. Both patients had persistent clonal hematopoiesis during remission, harboring all but 1 of the mutations from the initial diagnostic sample.
- Published
- 2019
20. Sequential high-dose cytarabine and mitoxantrone (S-HAM) versus standard double induction in acute myeloid leukemia-a phase 3 study
- Author
-
Dietrich W. Beelen, Peter Staib, Gero Massenkeil, Heidrun Hindahl, Andreas Faldum, Karsten Spiekermann, Stefan K. Bohlander, Anja Baumgartner, Christian Buske, Michael Unterhalt, Marion Subklewe, Dennis Görlich, Michael Fiegl, Gerald Meckenstock, Cristina Sauerland, Xaver Schiel, Guido Trenn, E. Roemer, Harald Biersack, Dirk Medgenberg, Birgit Braess, Raphael Koch, Bettina Zinngrebe, Jan Braess, Rainer Schwerdtfeger, Maike de Wit, Hans Walter Lindemann, Karl-Anton Kreuzer, Eva Lengfelder, Michael Uppenkamp, Dirk Behringer, Wolfgang Hiddemann, Ullrich Graeven, Ernst Spaeth-Schwalbe, Tobias Gaska, Bernhard Wörmann, Michael G. Kiehl, Stephanie Schneider, Stefan Korsten, Wolf-Dieter Ludwig, Susanne Amler, Rudolf Peceny, Martin Lablans, Yon-Dschun Ko, and Annika Dufour
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,Medizin ,Phases of clinical research ,Gastroenterology ,Article ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,immune system diseases ,Internal medicine ,hemic and lymphatic diseases ,mental disorders ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Aged ,Aged, 80 and over ,Mitoxantrone ,Leukopenia ,business.industry ,Remission Induction ,Cytarabine ,Myeloid leukemia ,food and beverages ,virus diseases ,Hematology ,Middle Aged ,medicine.disease ,Regimen ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,business ,030215 immunology ,medicine.drug - Abstract
Dose-dense induction with the S-HAM regimen was compared to standard double induction therapy in adult patients with newly diagnosed acute myeloid leukemia. Patients were centrally randomized (1:1) between S-HAM (2nd chemotherapy cycle starting on day 8 = “dose-dense”) and double induction with TAD-HAM or HAM(-HAM) (2nd cycle starting on day 21 = “standard”). 387 evaluable patients were randomly assigned to S-HAM (N = 203) and to standard double induction (N = 184). The primary endpoint overall response rate (ORR) consisting of complete remission (CR) and incomplete remission (CRi) was not significantly different (P = 0.202) between S-HAM (77%) and double induction (72%). The median overall survival was 35 months after S-HAM and 25 months after double induction (P = 0.323). Duration of critical leukopenia was significantly reduced after S-HAM (median 29 days) versus double induction (median 44 days)—P < 0.001. This translated into a significantly shortened duration of hospitalization after S-HAM (median 37 days) as compared to standard induction (median 49 days)—P < 0.001. In conclusion, dose-dense induction therapy with the S-HAM regimen shows favorable trends but no significant differences in ORR and OS compared to standard double induction. S-HAM significantly shortens critical leukopenia and the duration of hospitalization by 2 weeks.
- Published
- 2018
21. Young woman with mild bone marrow dysplasia, GATA2 and ASXL1 mutation treated with allogeneic hematopoietic stem cell transplantation
- Author
-
Helmut Blum, Alexander Graf, Stefan Krebs, Annika Dufour, Nikola P. Konstandin, Stig Lenhoff, Axel Weber, Sebastian Vosberg, Philipp A. Greif, Jörg Cammenga, Anna Lübking, Mats Ehinger, and Karsten Spiekermann
- Subjects
medicine.medical_specialty ,GATA2 mutation ,Myelodysplastic syndrome ,ASXL1 mutation ,Allogeneic hematopoietic stem cell transplantation ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Monocytopenia ,lcsh:RC254-282 ,Article ,hemic and lymphatic diseases ,Internal medicine ,Medicine ,Hematology ,business.industry ,Myeloid leukemia ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,MonoMAC ,Transplantation ,medicine.anatomical_structure ,Oncology ,Immunology ,Bone marrow ,Stem cell ,business - Abstract
Heterozygous mutations in GATA2 underlie different syndromes, previously described as monocytopenia and mycobacterial avium complex infection (MonoMAC); dendritic cell, monocytes, B- and NK lymphocytes deficiency (DCML); lymphedema, deafness and myelodysplasia (Emberger syndrome) and familiar myelodysplastic syndrome/acute myeloid leukemia (MDS / AML). Onset and severity of clinical symptoms vary and preceding cytopenias are not always present. We describe a case of symptomatic DCML deficiency and rather discrete bone marrow findings due to GATA2 mutation. Exome sequencing revealed a somatic ASXL1 mutation and the patient underwent allogeneic stem cell transplantation successfully., Highlights • Allogeneic stem cell transplantation was performed for DCML caused by GATA2 mutation. • Genetic diagnostics were done by Sanger sequencing and whole exome sequencing. • We identified an ASXL1 mutation associated with high risk for leukemic transformation.
- Published
- 2015
22. Inactivation of TP53 correlates with disease progression and low miR-34a expression in previously treated chronic lymphocytic leukemia patients
- Author
-
Philippe Solal-Celigny, Martin Weisser, Ru-Fang Yeh, Tobias Benthaus, Gudrun Mellert, John Catalano, Wolfgang Hiddemann, Stefan K. Bohlander, Debraj GuhaThakurta, Guillemette Duchateau-Nguyen, Purvi M. Kakadia, Tadeusz Robak, Lin Wu, Evelyn Zellmeier, Marco Montillo, Sabine Lohmann, David Dornan, Nancy Patten, Christian H. Geisler, Sim Truong, Giuseppe Palermo, Stephanie Schneider, Jan Braess, Galina Salogub, Annika Dufour, Anna Dmoszynska, and Karsten Spiekermann
- Subjects
Adult ,Male ,endocrine system diseases ,Chronic lymphocytic leukemia ,Immunology ,Down-Regulation ,Phases of clinical research ,Biochemistry ,Disease-Free Survival ,stomatognathic system ,Downregulation and upregulation ,Recurrence ,Biomarkers, Tumor ,Humans ,Medicine ,Gene Silencing ,Treatment Failure ,neoplasms ,Aged ,Regulation of gene expression ,medicine.diagnostic_test ,Gene Expression Regulation, Leukemic ,business.industry ,Point mutation ,Cell Biology ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,MicroRNAs ,Leukemia ,Disease Progression ,Cancer research ,Female ,Rituximab ,Tumor Suppressor Protein p53 ,business ,medicine.drug ,Fluorescence in situ hybridization - Abstract
In chronic lymphocytic leukemia (CLL) patients, disruptions of the TP53 tumor suppressor pathway by 17p13 deletion (del17p), somatic TP53 mutations, or downregulation of microRNA-34a have been associated with a poor prognosis. So far, the impact of the various TP53 defects has not been evaluated in a large cohort of previously treated and relapsed CLL patients. Here, we present the results of TP53 gene sequencing and fluorescence in situ hybridization for del17p in a phase 3 clinical trial (REACH [Rituximab in the Study of Relapsed Chronic Lymphocytic Leukemia]). Of the 457 patients, 52 had TP53 mutations and 37 had del17p. In 24 (46%) of the TP53 mutated patients, no del17p was found and in 9 of the del17p patients, no TP53 mutation was identified. Based on a predicted proportion of TP53 disruption, a complete disruption of TP53 function, either by a combination of point mutations and/or del17p, was associated with a high risk for disease progression. Progression-free survival of patients with a heterozygous TP53 mutation was not significantly different from patients with a completely intact TP53 locus. In addition, only a complete loss of TP53 function correlated with low microRNA-34a expression levels. This trial was registered at www.clinicaltrials.gov as #NCT00090051.
- Published
- 2013
23. The new and recurrent FLT3 juxtamembrane deletion mutation shows a dominant negative effect on the wild-type FLT3 receptor
- Author
-
Evelyn Zellmeier, Nikola P. Konstandin, Maja Rothenberg, Nadine Sandhöfer, Annika Dufour, Klaus H. Metzeler, Belay Tizazu, Karsten Spiekermann, Katrin Reiter, Wolfgang Hiddemann, Julia Bauer, Harald Polzer, and Philipp A. Greif
- Subjects
Male ,0301 basic medicine ,Biology ,Tropomyosin receptor kinase C ,Article ,03 medical and health sciences ,fluids and secretions ,0302 clinical medicine ,Growth factor receptor ,Cell Line, Tumor ,hemic and lymphatic diseases ,Humans ,5-HT5A receptor ,Receptor ,Genes, Dominant ,Multidisciplinary ,hemic and immune systems ,Ligand (biochemistry) ,Molecular biology ,Leukemia, Myeloid, Acute ,030104 developmental biology ,fms-Like Tyrosine Kinase 3 ,030220 oncology & carcinogenesis ,Interleukin-21 receptor ,Mutation ,embryonic structures ,Fms-Like Tyrosine Kinase 3 ,ROR1 ,Female - Abstract
In acute myeloid leukemia (AML), the Fms-like tyrosine kinase 3 (FLT3) is one of the most frequently mutated genes. Recently, a new and recurrent juxtamembrane deletion mutation (p.Q569Vfs*2) resulting in a truncated receptor was identified. The mutated receptor is expressed on the cell surface and still binds its ligand but loses the ability to activate ERK signaling. FLT3 p.Q569fs-expressing Ba/F3 cells show no proliferation after ligand stimulation. Furthermore, coexpressed with the FLT3 wild-type (WT) receptor, the truncated receptor suppresses stimulation and activation of the WT receptor. Thus, FLT3 p.Q569Vfs*2, to our knowledge, is the first FLT3 mutation with a dominant negative effect on the WT receptor.
- Published
- 2016
24. Identification of recurring tumor-specific somatic mutations in acute myeloid leukemia by transcriptome sequencing
- Author
-
Stefan K. Bohlander, Bettina Lorenz-Depiereux, T. M. Strom, Bianka Ksienzyk, Anna Benet-Pagès, Philipp A. Greif, A T Vetter, Thomas Meitinger, Nikola P. Konstandin, Annika Dufour, Sebastian Eck, and Henning D. Popp
- Subjects
Cancer Research ,Nonsense mutation ,Biology ,Polymorphism, Single Nucleotide ,Transcriptome ,Germline mutation ,Bone Marrow ,hemic and lymphatic diseases ,Biomarkers, Tumor ,Humans ,Missense mutation ,RNA, Messenger ,Aged ,COLD-PCR ,Whole genome sequencing ,Genetics ,Reverse Transcriptase Polymerase Chain Reaction ,Sequence Analysis, RNA ,Point mutation ,High-Throughput Nucleotide Sequencing ,Myeloid leukemia ,Hematology ,acute myeloid leukemia ,point mutations ,TLE4 ,SHKBP1 ,RUNX1 ,transcriptome sequencing ,Leukemia, Myeloid, Acute ,Oncology ,Mutation ,Cancer research - Abstract
Genetic lesions are crucial for cancer initiation. Recently, whole genome sequencing, using next generation technology, was used as a systematic approach to identify mutations in genomes of various types of tumors including melanoma, lung and breast cancer, as well as acute myeloid leukemia (AML). Here, we identify tumor-specific somatic mutations by sequencing transcriptionally active genes. Mutations were detected by comparing the transcriptome sequence of an AML sample with the corresponding remission sample. Using this approach, we found five non-synonymous mutations specific to the tumor sample. They include a nonsense mutation affecting the RUNX1 gene, which is a known mutational target in AML, and a missense mutation in the putative tumor suppressor gene TLE4, which encodes a RUNX1 interacting protein. Another missense mutation was identified in SHKBP1, which acts downstream of FLT3, a receptor tyrosine kinase mutated in about 30% of AML cases. The frequency of mutations in TLE4 and SHKBP1 in 95 cytogenetically normal AML patients was 2%. Our study demonstrates that whole transcriptome sequencing leads to the rapid detection of recurring point mutations in the coding regions of genes relevant to malignant transformation.
- Published
- 2011
25. Minimal Residual Disease (MRD) Detection By Flow Cytometry Complements Molecular MRD Assessment in AML
- Author
-
Thomas Köhnke, Marion Subklewe, Cristina Sauerland, Frauke M. Schnorfeil, Wolfgang Hiddemann, Nikola P. Konstandin, Veit Bücklein, Annika Dufour, Karsten Spiekermann, and Klaus H. Metzeler
- Subjects
Oncology ,medicine.medical_specialty ,Surrogate endpoint ,business.industry ,Immunology ,Induction chemotherapy ,Cell Biology ,Hematology ,Biochemistry ,Minimal residual disease ,Chemotherapy regimen ,body regions ,Clinical trial ,Immunophenotyping ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Risk factor ,business ,Survival analysis - Abstract
The prognosis of patients with AML is determined by a multitude of recurrent genetic alterations, and treatment algorithms heavily rely on risk stratification by genetic characterization of the disease at the time of diagnosis. However, this a priori risk stratification does not integrate information on treatment susceptibility of the individual patient. Assessment of Minimal Residual Disease (MRD) aims to implement this information in the patient-specific treatment management. AML cells with aberrant phenotypes can be detected at sensitivities below 1:104 by flow cytometry in the majority of patients. Therefore, flow cytometry MRD assessment (flow MRD) enables determination of MRD status in patients without suitable molecular markers (e.g. NPM1, CBFß-MYH11, and RUNX1-RUNX1T1). Here, we validate the role of flow MRD in AML patients receiving intensive chemotherapy with and without available molecular markers. Flow MRD was analyzed in patients with AML (excluding APL) diagnosed between 2012 and 2017 receiving intensive induction chemotherapy (sHAM or 7+3). Flow MRD analysis was performed during aplasia (on day 16 after treatment initiation) as well as post induction. Presence of ≥0.1% Leukemia-associated immunophenotype (LAIP)-positive cells was defined as flow MRD positivity. Molecular MRD was analyzed post induction for NPM1 and CBF, and post consolidation for RUNX1-RUNX1T1. Kaplan-Meier estimators and log-rank tests were used to analyze survival data. Cox's proportional hazards regression model was used to determine the influence of individual factors in multivariate analyses. A total of 161 patients were included. In 5 cases (3.1% of all cases), no LAIP could be identified, and these patients were excluded from further analyses. Flow MRD assessment during aplasia was available in 145 cases. 122 patients had flow MRD assessments available post induction. 114 patients achieving CR or CRi after induction therapy had flow MRD assessments available at both time points. Flow MRD positivity during aplasia was associated with shorter event free survival (EFS, 6.1 months vs. 19.1 months, p0.05 for all endpoints). 64/161 patients (39%) had molecular MRD assessment available for analysis. In these patients, molecular MRD positivity predicted a significantly shorter EFS (9.3 months vs. median not reached, p=0.01). Indeed, molecular MRD positivity was an independent risk factor for adverse EFS and RFS (HR 1.7 and 1.6, p=0.008 and p=0.018, respectively). In this subgroup, flow MRD was not an independent prognostic factor. However, for patients without available molecular MRD marker (97/161 patients), flow MRD positivity at aplasia (p=0.004), post induction (p=0.015) or as combined status (p=0.004) was associated with a significantly shorter EFS and remained an independent risk factor in multivariate analysis (HR 2.5 and 2.6, p=0.016 and p=0.012 for EFS and RFS, respectively). Taken together, we demonstrate that both flow MRD as well as molecular MRD strongly correlate with survival. While molecular MRD assessment was only available in 39% of patients, MRD assessment by flow cytometry was feasible in >95% of AML patients. Flow MRD positivity both during aplasia and post induction was an independent risk factor, confirming the superiority of flow MRD compared to early morphologic response assessment. In conclusion, molecular and flow cytometric MRD assessment are complementing methods for the estimation of treatment response, and will be integrated in clinical trials to validate their significance for patient-specific treatment management. Disclosures Metzeler: Celgene: Consultancy, Research Funding; Novartis: Consultancy. Hiddemann:F. Hoffman-La Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer: Consultancy, Research Funding; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Subklewe:Gilead Sciences: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees; Roche AG: Research Funding; AMGEN: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2018
26. NPM1 Variant Allele Frequency and Outcomes in AML
- Author
-
Stefan K. Bohlander, Marion Subklewe, Bernhard Wörmann, Karsten Spiekermann, Cristina Sauerland, Tobias Herold, Stephanie Schneider, Wolfgang E. Berdel, Michael von Bergwelt, Jan Braess, Klaus H. Metzeler, Dennis Görlich, Wolfgang Hiddemann, Annika Dufour, Nikola P. Konstandin, Andreas Faldum, Hanna Janke, Evelyn Packeiser, Bianka Ksienzyk, Utz Krug, Maja Rothenberg-Thurley, and Victoria V. Prassek
- Subjects
Genetics ,NPM1 ,business.industry ,Immunology ,Cell Biology ,Hematology ,Variant allele ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Medicine ,business ,030215 immunology - Abstract
Background: Mutations in the NPM1 gene are among the most common genetic alterations in patients with acute myeloid leukemia (AML). NPM1 mutations predominantly occur in patients with normal or intermediate-risk abnormal cytogenetics, and define a distinct subgroup of AML patients recognized in the 2016 WHO classification. Overall, mutated NPM1 associates with favorable response to induction chemotherapy and relatively favorable overall survival. However, this prognostic impact is modulated by the presence of other gene mutations including FLT3 internal tandem duplications (ITD) and DNMT3A mutations. Recently, Patel and colleagues reported that a high variant allele frequency (VAF) of mutated NPM1 at the time of initial diagnosis associates with unfavorable outcomes in de novo AML (Blood 131:2816-25). This interesting and unexpected observation prompted us to investigate the association between NPM1 VAF and outcomes in a large AML patient cohort. Patients and Methods: We studied NPM1 mutated AML patients who had been enrolled on two successive multicenter phase III trials of the German AML Cooperative Group (AML-CG 1999, NCT00266136; AML-CG 2008, NCT01382147) and genetically characterized by amplicon-based targeted next-generation sequencing (NGS, Agilent Haloplex; Metzeler et al., Blood 128:686-98 and unpublished data). All patients had received induction chemotherapy containing cytarabine with either daunorubicin plus thioguanine or mitoxantrone. The minimum VAF for calling of insertion/deletion variants was 0.05, and samples with NPM1 coverage Results: We identified 417 NPM1-mutated patients (type A mutations, 316; type B, 28; type D, 35; and other types, 38). Median patient age was 56 years (range, 19 - 86 years), and 31/414 patients (7.5%) with cytogenetic data had abnormal karyotypes. The median NPM1 VAF was 0.43 (range, 0.05 to 1.0). Type A NPM1 mutations had significantly higher VAFs than non-type A mutations (median, 0.43 vs. 0.41; P=.0002), while type B mutations had lower VAFs than non-type B mutations (median, 0.34 vs. 0.43; P=.0025) (Figure A). Age or karyotype did not associate with NPM1 VAF. NPM1 VAF, as a continuous variable, did not associate with response to induction chemotherapy (P=.6) or relapse-free survival (P=.22). A higher NPM1 VAF did, however, associate with shorter OS (hazard ratio for an increase in NPM1 VAF equal to the interquartile range, 1.14; 95% confidence interval, 1.00-1.30; P=.049). In particular, patients in the lowest quartile of NPM1 allele burden ('low NPM1 VAF') had longer OS than patients with allele burdens above the 25th percentile ('high NPM1 VAF') (median OS, 63.7 vs. 27.0 months; 5-year OS, 51% vs 42%; P=.05; Figure B). Patients with high NPM1 VAF had higher leukocyte counts (median, 46000/µl vs. 9300/µl; P Conclusion: Our study confirms the recent report that adult AML patients with high NPM1 mutant allele burden have shorter survival. In our cohort, however, higher NPM1 VAF also associated with higher leukocyte counts and marrow blast percentage, and with prognostically adverse FLT3-ITD and DNMT3A mutations. After adjusting for these confounders, NPM1 allelic burden did not independently predict survival in our analysis. We therefore suspect that high NPM1 VAF may be a surrogate marker of highly proliferative AML subsets, for example those with high allelic ratio FLT3-ITD, rather than a novel independent prognostic factor. Figure. Figure. Disclosures Prassek: Jannsen: Other: Travel support; Celgene: Other: Travel support. Subklewe:Roche: Consultancy, Research Funding; Gilead: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Celgene: Consultancy, Honoraria. Hiddemann:Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; F. Hoffman-La Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer: Consultancy, Research Funding; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Metzeler:Celgene: Consultancy, Research Funding; Novartis: Consultancy.
- Published
- 2018
27. Acute Myeloid Leukemia With Biallelic CEBPA Gene Mutations and Normal Karyotype Represents a Distinct Genetic Entity Associated With a Favorable Clinical Outcome
- Author
-
Wolfgang Hiddemann, Tobias Benthaus, Thomas Büchner, Stephanie Schneider, Bernhard Wörmann, Eva Hoster, Jan Braess, Annika Dufour, Wolfgang E. Berdel, Maria-Cristina Sauerland, Evelyn Zellmeier, Stefan K. Bohlander, Friederike Schneider, Karsten Spiekermann, and Klaus H. Metzeler
- Subjects
Adult ,Male ,Cancer Research ,NPM1 ,Myeloid ,Adolescent ,medicine.disease_cause ,Young Adult ,CEBPA ,Biomarkers, Tumor ,medicine ,Humans ,Survival rate ,Alleles ,Aged ,Neoplasm Staging ,Oligonucleotide Array Sequence Analysis ,Aged, 80 and over ,Mutation ,Gene Expression Regulation, Leukemic ,business.industry ,Gene Expression Profiling ,Nuclear Proteins ,Myeloid leukemia ,MINIMAL RESIDUAL DISEASE ,BINDING-PROTEIN-ALPHA ,C/EBP-ALPHA ,PROGNOSTIC-SIGNIFICANCE ,EXPRESSION PROFILE ,TANDEM DUPLICATION ,RELAPSE SAMPLES ,AML ,FLT3 ,CYTOGENETICS ,Histone-Lysine N-Methyltransferase ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,Gene expression profiling ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Oncology ,Karyotyping ,CCAAT-Enhancer-Binding Proteins ,Cancer research ,Female ,business ,Nucleophosmin ,Myeloid-Lymphoid Leukemia Protein - Abstract
Purpose CEBPA mutations are found as either biallelic (biCEBPA) or monoallelic (moCEBPA). We set out to explore whether the kind of CEBPA mutation is of prognostic relevance in cytogenetically normal (CN) acute myeloid leukemia (AML). Patients and Methods Four hundred sixty-seven homogeneously treated patients with CN-AML were subdivided into moCEBPA, biCEBPA, and wild-type (wt) CEBPA patients. The subgroups were analyzed for clinical parameters and for additional mutations in the NPM1, FLT3, and MLL genes. Furthermore, we obtained gene expression profiles using oligonucleotide microarrays. Results Only patients with biCEBPA had an improved median overall survival when compared with patients with wtCEBPA (not reached v 20.4 months, respectively; P = .018), whereas patients with moCEBPA (20.9 months) and wtCEBPA had a similar outcome (P = .506). Multivariable analysis confirmed biCEBPA, but not moCEBPA, mutations as an independent favorable prognostic factor. Interestingly, biCEBPA mutations, compared with wtCEBPA, were never associated with mutated NPM1 (0% v 43%, respectively; P < .001) and rarely associated with FLT3 internal tandem duplication (ITD; 5% v 23%, respectively; P = .059), whereas patients with moCEBPA had a similar frequency of mutated NPM1 and a significantly higher association with FLT3-ITD compared with patients with wtCEBPA (44% v 23%, respectively; P = .037). Furthermore, patients with biCEBPA showed a homogeneous gene expression profile that was characterized by downregulation of HOX genes, whereas patients with moCEBPA showed greater heterogeneity in their gene expression profiles. Conclusion Biallelic disruption of the N and C terminus of CEBPA is required for the favorable clinical outcome of CEBPA-mutated patients and represents a distinct molecular subtype of CN-AML with a different frequency of associated gene mutations. These findings are of great significance for risk-adapted therapeutic strategies in AML.
- Published
- 2010
28. ERG Expression Is an Independent Prognostic Factor and Allows Refined Risk Stratification in Cytogenetically Normal Acute Myeloid Leukemia: A Comprehensive Analysis of ERG, MN1, and BAALC Transcript Levels Using Oligonucleotide Microarrays
- Author
-
Wolfgang E. Berdel, Jan Braess, Christian Buske, Ulrich Mansmann, Klaus H. Metzeler, Maria-Cristina Sauerland, Achim Heinecke, Annika Dufour, Karsten Spiekermann, Bernhard Wörmann, Tobias Benthaus, Thomas Büchner, Wolfgang Hiddemann, Manuela Hummel, and Stefan K. Bohlander
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,NPM1 ,Myeloid ,Adolescent ,Transcription, Genetic ,Risk Assessment ,Cohort Studies ,Young Adult ,Transcriptional Regulator ERG ,Risk Factors ,Internal medicine ,CEBPA ,medicine ,Humans ,Survival analysis ,BAALC ,Aged ,Oligonucleotide Array Sequence Analysis ,Aged, 80 and over ,internal tandem duplications ,high-dose cytarabine ,ets-related gene ,group-b ,normal karyotype ,prolonged maintenance ,favorable prognosis ,microrna expression ,prostate-cancer ,cebpa mutations ,business.industry ,Gene Expression Profiling ,Tumor Suppressor Proteins ,Middle Aged ,Prognosis ,medicine.disease ,Survival Analysis ,Neoplasm Proteins ,Gene expression profiling ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Immunology ,Trans-Activators ,Female ,business ,Nucleophosmin - Abstract
Purpose Recently, several novel molecular prognostic markers were identified in cytogenetically normal acute myeloid leukemia (CN-AML). In addition to the well-known influence of FLT3, NPM1, and CEBPA mutations, high transcript levels of the ERG, BAALC, and MN1 genes have been associated with inferior outcomes, but the relative importance of these risk markers remains to be defined. Patients and Methods We analyzed ERG, BAALC, and MN1 expression levels in a cohort of 210 patients with CN-AML who received intensive chemotherapy. Expression levels of ERG, BAALC, and MN1 were determined in bone marrow samples by using oligonucleotide microarrays. Results High transcript levels of ERG, BAALC, and MN1 were predictors for inferior overall survival (OS) and a lower rate of complete remissions (CRs). There were significant positive correlations between the expression levels of all three genes. ERG expression levels predicted OS in elderly patients (ie, age 60 years or older) with CN-AML (P = .006) as well as in younger patients (P = .013). In multivariate analyses, high ERG expression was independently associated with a lower CR rate (P = .013), shorter event-free survival (P = .008), and shorter OS (P = .005). Patients who had low ERG levels and absent FLT3 internal tandem duplication (ITD) had a 5-year OS of 44%, and patients who had high ERG expression and FLT3 ITD had a 5-year OS of only 5%. Conclusion We analyzed a comprehensive set of molecular risk factors in a large, homogeneous CN-AML patient cohort. In this study, high ERG expression levels emerged as a strong negative prognostic factor and provided prognostic information in addition to established molecular markers.
- Published
- 2009
29. NPM1 but not FLT3-ITD mutations predict early blast cell clearance and CR rate in patients with normal karyotype AML (NK-AML) or high-risk myelodysplastic syndrome (MDS)
- Author
-
Wolfgang Hiddemann, Bernhard J. Woermann, Eva Hoster, Christian Buske, Tobias Benthaus, Stephanie Schneider, Karsten Spiekermann, Thomas Buechner, Achim Heinecke, Maria Cristina Sauerland, Friederike Schneider, Michaela Feuring-Buske, Stefan K. Bohlander, Wolfgang E. Berdel, Susanne Fritsch, Jan Braess, Evelin Zellmeier, Gudrun Mellert, Michael Unterhalt, and Annika Dufour
- Subjects
Oncology ,medicine.medical_specialty ,NPM1 ,Myeloid ,Immunology ,Antineoplastic Agents ,Biology ,medicine.disease_cause ,Biochemistry ,hemic and lymphatic diseases ,Internal medicine ,Precursor cell ,medicine ,Humans ,Mutation ,Inverted Repeat Sequences ,Remission Induction ,Nuclear Proteins ,Myeloid leukemia ,Induction chemotherapy ,Cancer ,Cell Biology ,Hematology ,Prognosis ,medicine.disease ,Killer Cells, Natural ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Karyotyping ,Myelodysplastic Syndromes ,Cancer research ,Blast Crisis ,Nucleophosmin - Abstract
Mutations in the NPM1 gene represent the most frequent genetic alterations in patients with acute myeloid leukemia (AML) and are associated with a favorable outcome. In 690 normal karyotype (NK) AML patients the complete remission rates (CRs) and the percentage of patients with adequate in vivo blast cell reduction 1 week after the end of the first induction cycle were significantly higher in NPM1+ (75% and 80%, respectively) than in NPM1− (57% and 57%, respectively) patients, but were unaffected by the FLT3-ITD status. Multivariate analyses revealed the presence of a NPM1 mutation as an independent positive prognostic factor for the achievement of an adequate day-16 blast clearance and a CR. In conclusion, NPM1+ blast cells show a high in vivo sensitivity toward induction chemotherapy irrespective of the FLT3-ITD mutation status. These findings provide insight into the pathophysiology and help to understand the favorable clinical outcome of patients with NPM1+ AML.
- Published
- 2009
30. Gene expression of INPP5F as an independent prognostic marker in fludarabine-based therapy of chronic lymphocytic leukemia
- Author
-
Martin Weisser, Ru-Fang Yeh, Tadeusz Robak, Tri Quang Nguyen, Giuseppe Palermo, Helen Smith, David Dornan, D Maisel, Guillemette Duchateau-Nguyen, Martin Barrett, and Annika Dufour
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Cyclophosphamide ,Chronic lymphocytic leukemia ,Antineoplastic Agents ,Kaplan-Meier Estimate ,Disease-Free Survival ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Survival analysis ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,Hematology ,business.industry ,Hazard ratio ,Inositol Polyphosphate 5-Phosphatases ,Middle Aged ,medicine.disease ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,Phosphoric Monoester Hydrolases ,Fludarabine ,Leukemia ,Immunology ,Rituximab ,Female ,Original Article ,business ,Transcriptome ,Vidarabine ,medicine.drug - Abstract
Chronic lymphocytic leukemia (CLL) is a heterogeneous disease. Various disease-related and patient-related factors have been shown to influence the course of the disease. The aim of this study was to identify novel biomarkers of significant clinical relevance. Pretreatment CD19-separated lymphocytes (n=237; discovery set) and peripheral blood mononuclear cells (n=92; validation set) from the REACH trial, a randomized phase III trial in relapsed CLL comparing rituximab plus fludarabine plus cyclophosphamide with fludarabine plus cyclophosphamide alone, underwent gene expression profiling. By using Cox regression survival analysis on the discovery set, we identified inositol polyphosphate-5-phosphatase F (INPP5F) as a prognostic factor for progression-free survival (PPINPP5F may serve as a novel, easy-to-assess future prognostic biomarker for fludarabine-based therapy in CLL.
- Published
- 2015
31. Early assessment of minimal residual disease in aml by flow cytometry during aplasia identifies patients at increased risk of relapse
- Author
-
Thomas Köhnke, Annika Dufour, Th. Büchner, Max Hubmann, Marion Subklewe, Laubender Rp, Maria-Cristina Sauerland, Stephanie Schneider, Jan Braess, Eva Hoster, Katharina Ringel, Wolfgang E. Berdel, Stefan K. Bohlander, Daniela Sauter, Karsten Spiekermann, Purvi M. Kakadia, Wolfgang Hiddemann, and Bernhard Wörmann
- Subjects
Male ,Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Pathology ,Neoplasm, Residual ,Bone Marrow Cells ,Disease-Free Survival ,Flow cytometry ,Bone Marrow ,Risk Factors ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,neoplasms ,Aged ,Proportional Hazards Models ,medicine.diagnostic_test ,business.industry ,Remission Induction ,Nuclear Proteins ,Hematology ,Aplasia ,Middle Aged ,Flow Cytometry ,Prognosis ,medicine.disease ,Minimal residual disease ,body regions ,Leukemia, Myeloid, Acute ,stomatognathic diseases ,Treatment Outcome ,Increased risk ,Karyotyping ,Leukocytes, Mononuclear ,Female ,Neoplasm Recurrence, Local ,business ,Nucleophosmin - Abstract
In acute myeloid leukemia (AML), assessment of minimal residual disease (MRD) by flow cytometry (flow MRD) after induction and consolidation therapy has been shown to provide independent prognostic information. However, data on the value of earlier flow MRD assessment are lacking. Therefore, the value of flow MRD detection was determined during aplasia in 178 patients achieving complete remission after treatment according to AMLCG (AML Cooperative Group) induction protocols. Flow MRD positivity during aplasia predicted poor outcome (5-year relapse-free survival (RFS) 16% vs 43%, P0.001) independently from age and cytogenetic risk group (hazard ratio for MRD positivity 1.71; P=0.009). Importantly, the prognosis of patients without detectable MRD was neither impacted by morphological blast count during aplasia nor by MRD status postinduction. Early flow MRD was also evaluated in the context of existing risk factors. Flow MRD was prognostic within the intermediate cytogenetic risk group (5-year RFS 15% vs 37%, P=0.016) as well as for patients with normal karyotype and NPM1 mutations (5-year RFS 13% vs 49%, P=0.02) or FLT3-ITD (3-year RFS rates 9% vs 44%, P=0.016). Early flow MRD assessment can improve current risk stratification approaches by prediction of RFS in AML and might facilitate adaptation of postremission therapy for patients at high risk of relapse.
- Published
- 2015
32. Atrial fibrosis in heart surgery patients
- Author
-
Johannes Böhm, Matthias R. Benz, J Grammer, Robert Bauernschmitt, Annika Dufour, and Rüdiger Lange
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Collagen Type I ,Collagen Type III ,Postoperative Complications ,Risk Factors ,Fibrosis ,Physiology (medical) ,Internal medicine ,Atrial Fibrillation ,Humans ,Medicine ,Sinus rhythm ,Heart Atria ,RNA, Messenger ,Osteopontin ,Cardiac Surgical Procedures ,Aged ,biology ,business.industry ,Myocardium ,Atrial fibrillation ,Angiotensin-converting enzyme ,Middle Aged ,medicine.disease ,Cardiac surgery ,biology.protein ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers ,Type I collagen - Abstract
In chronic atrial fibrillation, increased expression of angiotensinconverting enzyme (ACE) promotes upregulation of profibrotic proteins. Atrial fibrillation early after cardiac surgery (poAF) is common but unpredictable, and is regarded as a different entity. Therefore, the present study tested whether atrial expression of ACE, osteopontin (OPN), and collagen is elevated in patients with no history of AF but who develop poAF. Thus, 19 patients (66 +/- 9 years) with postoperative sinus rhythm (poSR) were compared to 14 patients (68 +/- 10) who experienced poAF. mRNA and protein expression were determined by RT-PCR and Western blotting. Picrosirius red was used to stain collagen. The medians for ACE, OPN, type I collagen (Col I), and type III collagen (Col III) mRNA and protein expression did not significantly differ between poSR and poAF (U-test). However, the Col III/I protein ratio was significantly lower in patients with poAF (2.62 vs. 1.09; poSR vs. poAF; p < 0.05). Our data suggest that the occurrence of poAF is not dependent on increased ACE and OPN expression, rendering the determination of preoperative OPN plasma levels inadequate to estimate the risk for the occurrence of poAF. Contrarily, a shift in atrial collagen expression levels in favor of Col I is linked to the occurrence of poAF.
- Published
- 2005
33. Isolated trisomy 13 defines a homogeneous AML subgroup with high frequency of mutations in spliceosome genes and poor prognosis
- Author
-
Alexander Graf, Max Hubmann, Ulrich Mansmann, Stefan Krebs, Zlatana Pasalic, Bianka Ksienzyk, Annika Dufour, Tobias Herold, Wolfgang E. Berdel, Bernhard J. Woermann, Gerhard Ehninger, Vindi Jurinovic, Martin Bornhäuser, Friedrich Stölzel, Maria Cristina Sauerland, Philipp A. Greif, Sebastian Vosberg, Luise Hartmann, Helmut Blum, C. Röllig, Stefan K. Bohlander, Stephanie Schneider, Klaus H. Metzeler, Purvi M. Kakadia, Karsten Spiekermann, Evelyn Zellmeier, Thomas Büchner, and Wolfgang Hiddemann
- Subjects
Adult ,Male ,Poor prognosis ,Spliceosome ,Adolescent ,Immunology ,Trisomy ,Biology ,Bioinformatics ,Biochemistry ,Disease-Free Survival ,hemic and lymphatic diseases ,Germany ,medicine ,Humans ,neoplasms ,Gene ,Exome sequencing ,Aged ,Aged, 80 and over ,Chromosomes, Human, Pair 13 ,Gene Expression Regulation, Leukemic ,Clinical course ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Neoplasm Proteins ,Up-Regulation ,Survival Rate ,Leukemia, Myeloid, Acute ,Homogeneous ,Cancer research ,Female - Abstract
Isolated trisomy 13 (AML+13) is a rare chromosomal abnormality in acute myeloid leukemia (AML), and its prognostic relevance is poorly characterized. We analyzed the clinical course of 34 AML+13 patients enrolled in the German AMLCG-1999 and SAL trials and studied their biological characteristics by exome sequencing, targeted sequencing of candidate genes and gene expression profiling. Relapse-free (RFS) and overall survival (OS) of AML+13 patients were inferior compared to other ELN Intermediate-II patients (n=855) (median RFS, 7.8 vs 14.1 months, p=0.006; median OS 9.3 vs. 14.8 months, p=0.004). Besides the known high frequency of RUNX1 mutations (75%), we identified mutations in spliceosome components in 88%, including SRSF2 codon 95 mutations in 81%, of AML+13 patients. Moreover, recurring mutations were detected in ASXL1 (44%) and BCOR (25%). Two patients carried mutations in CEBPZ, suggesting that CEBPZ is a novel recurrently mutated gene in AML. Gene expression analysis revealed a homogenous expression profile including upregulation of FOXO1 and FLT3 and downregulation of SPRY2. This is the most comprehensive clinical and biological characterization of AML+13 to date, and reveals a striking clustering of lesions in a few genes, defining AML+13 as a genetically homogenous leukemia subgroup with alterations in a few critical cellular pathways. These studies were registered at clinicaltrials.gov, identifiers: AMLCG-1999: NCT00266136; AML96: NCT00180115; AML2003: NCT00180102; and AML60+: NCT00893373.
- Published
- 2014
34. PTK2 expression and immunochemotherapy outcome in chronic lymphocytic leukemia
- Author
-
Michael Wenger, Loree Larratt, Nancy Yu, Christian H. Geisler, Galina Salogub, Viktor Rossiev, Johannes Bloehdorn, Javier Loscertales, Martin Weisser, Ru-Fang Yeh, Anna Dmoszynska, Giuseppe Palermo, David Dornan, Xiaoyan Shi, Tri Quang Nguyen, Annika Dufour, Tadeusz Robak, Marco Montillo, Axel Benner, Kirsten Fischer, Anna-Maria Fink, Philippe Solal-Celigny, Krzysztof Warzocha, John Catalano, Isabelle Bence-Bruckler, Susanna Stinson, Hartmut Döhner, Stephan Stilgenbauer, Michael Hallek, Raymonde Busch, Nancy Valente, and Guillemette Duchateau-Nguyen
- Subjects
Oncology ,medicine.medical_specialty ,Cyclophosphamide ,Chronic lymphocytic leukemia ,Immunology ,Gene Expression ,Biochemistry ,Disease-Free Survival ,Antibodies, Monoclonal, Murine-Derived ,Recurrence ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,RNA, Messenger ,RNA, Neoplasm ,Proportional Hazards Models ,Retrospective Studies ,business.industry ,Proportional hazards model ,Cell Biology ,Hematology ,medicine.disease ,Combined Modality Therapy ,Leukemia, Lymphocytic, Chronic, B-Cell ,Fludarabine ,Leukemia ,Focal Adhesion Kinase 1 ,Monoclonal ,Cancer research ,Biomarker (medicine) ,Rituximab ,Immunotherapy ,business ,Vidarabine ,medicine.drug - Abstract
Addition of rituximab (R) to fludarabine and cyclophosphamide (FC) has significantly improved patient outcomes in chronic lymphocytic leukemia (CLL). Whether baseline gene expression can identify patients who will benefit from immunochemotherapy over chemotherapy alone has not been determined. We assessed genome-wide expression of 300 pretreatment specimens from a subset of 552 patients in REACH, a study of FC or R-FC in relapsed CLL. An independent test set was derived from 282 pretreatment specimens from CLL8, a study of FC or R-FC in treatment-naive patients. Genes specific for benefit from R-FC were determined by assessing treatment-gene interactions in Cox proportional hazards models. REACH patients with higher pretreatment protein tyrosine kinase 2 (PTK2) messenger RNA levels derived greater benefit from R-FC, with significant improvements in progression-free survival, independent of known prognostic factors in a multivariate model. Examination of PTK2 gene expression in CLL8 patients yielded similar results. Furthermore, PTK2 inhibition blunted R-dependent cell death in vitro. This retrospective analysis from 2 independent trials revealed that increased PTK2 expression is associated with improved outcomes for CLL patients treated with R-FC vs FC. PTK2 expression may be a useful biomarker for patient selection in future trials. These trials were registered at www.clinicaltrials.gov as #NCT00090051 (REACH) and #NCT00281918 (CLL8).
- Published
- 2014
35. Long-term follow-up of cytogenetically normal CEBPA-mutated AML
- Author
-
Friederike, Pastore, Daniela, Kling, Eva, Hoster, Annika, Dufour, Nikola P, Konstandin, Stephanie, Schneider, Maria C, Sauerland, Wolfgang E, Berdel, Thomas, Buechner, Bernhard, Woermann, Jan, Braess, Wolfgang, Hiddemann, and Karsten, Spiekermann
- Subjects
Male ,Acute Myeloid Leukemia ,Gene Expression Profiling ,Research ,Antineoplastic Agents ,Biallelic CEBPA mutation ,Kaplan-Meier Estimate ,Middle Aged ,Prognosis ,Disease-Free Survival ,Monoallelic CEBPA mutation ,Cytogenetically normal AML ,Leukemia, Myeloid, Acute ,Treatment Outcome ,Mutation ,Biomarkers, Tumor ,CCAAT-Enhancer-Binding Proteins ,Humans ,Female ,Aged ,Follow-Up Studies - Abstract
Background The aim of this study was to analyze the long-term survival of AML patients with CEBPA mutations. Patients and methods We investigated 88 AML patients with a median age of 61 years and (1) cytogenetically normal AML (CN-AML), (2) monoallelic (moCEBPA) or biallelic (biCEBPA) CEBPA mutation, and (3) intensive induction treatment. 60/88 patients have been described previously with a shorter follow-up. Results Median follow-up time was 9.8 years (95% CI: 9.4-10.1 years) compared to 3.2 and 5.2 years in our former analyses. Patients with biCEBPA mutations survived significantly longer compared to those with moCEBPA (median overall survival (OS) 9.6 years vs. 1.7 years, p = 0.008). Patients ≤ 60 years and biCEBPA mutations showed a favorable prognosis with a 10-year OS rate of 81%. Both, bi- and moCEBPA-mutated groups had a low early death (d60) rate of 7% and 9%, respectively. Complete remission (CR) rates for biCEBPA- and moCEBPA-mutated patients were 82% vs. 70% (p = 0.17). biCEBPA-mutated patients showed a longer relapse free survival (RFS) (median RFS 9.4 years vs. 1.5 years, p = 0.021) and a lower cumulative incidence of relapse (CIR) compared to moCEBPA-mutated patients. These differences in OS and RFS were confirmed after adjustment for known clinical and molecular prognostic factors. Conclusions In this long-term observation we confirmed the favorable prognostic outcome of patients with biCEBPA mutations compared to moCEBPA-mutated CN-AML. The high probability of OS (81%) in younger patients is helpful to guide intensity of postremission therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13045-014-0055-7) contains supplementary material, which is available to authorized users.
- Published
- 2014
36. Molecular response assessment by quantitativerael-time polymerase chain reaction after induction therapy in NPM1-mutated patients identifies patients at high risk for relapse
- Author
-
Marion Subklewe, Thomas Büchner, Michael Fiegl, Wolfgang E. Berdel, Wolfgang Hiddemann, Annika Dufour, Max Hubmann, Karsten Spiekermann, Maria-Cristina Sauerland, Stephanie Schneider, Thomas Köhnke, Jan Braess, Evelyn Zellmeier, Bernhard Wörmann, Stefan K. Bohlander, and Eva Hoster
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Adolescent ,Real-Time Polymerase Chain Reaction ,Acute Myeloid Leukemia ,Mrd ,Minimal Residual Disease ,Npm1 ,Rt-pcr ,Young Adult ,Recurrence ,Risk Factors ,Internal medicine ,medicine ,Humans ,Cumulative incidence ,Prospective Studies ,Prospective cohort study ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Hazard ratio ,Nuclear Proteins ,Myeloid leukemia ,Retrospective cohort study ,Induction Chemotherapy ,Articles ,Hematology ,Middle Aged ,medicine.disease ,Minimal residual disease ,Surgery ,Survival Rate ,Clinical trial ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,Mutation ,Female ,business ,Nucleophosmin ,Follow-Up Studies - Abstract
Monitoring minimal residual disease is an important way to identify patients with acute myeloid leukemia at high risk of relapse. In this study we investigated the prognostic potential of minimal residual disease monitoring by quantitative real-time polymerase chain reaction analysis of NPM1 mutations in patients treated in the AMLCG 1999, 2004 and 2008 trials. Minimal residual disease was monitored - in aplasia, after induction therapy, after consolidation therapy, and during follow-up - in 588 samples from 158 patients positive for NPM1 mutations A, B and D (with a sensitivity of 10(-6)). One hundred and twenty-seven patients (80.4%) achieved complete remission after induction therapy and, of these, 56 patients (44.1%) relapsed. At each checkpoint, minimal residual disease cut-offs were calculated. After induction therapy a cut-off NPM1 mutation ratio of 0.01 was associated with a high hazard ratio of 4.26 and the highest sensitivity of 76% for the prediction of relapse. This was reflected in a cumulative incidence of relapse after 2 years of 77.8% for patients with ratios above the cut-off versus 26.4% for those with ratios below the cut-off. In the favorable subgroup according to European LeukemiaNet, the cut-off after induction therapy also separated the cohort into two prognostic groups with a cumulative incidence of relapse of 76% versus 6% after 2 years. Our data demonstrate that in addition to pre-therapeutic factors, the course of minimal residual disease in an individual is an important prognostic factor and could be included in clinical trials for the guidance of post-remission therapy. The trials from which data were obtained were registered at www.clinicaltrials.gov (#NCT01382147, #NCT00266136) and at the European Leukemia Trial Registry (#LN_AMLINT2004_230).
- Published
- 2014
37. Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia
- Author
-
Annika Dufour, Gudrun Mellert, Guido Marcucci, Eva Hoster, Kati Maharry, Bernhard J. Woermann, Wolfgang E. Berdel, Stefan K. Bohlander, Maria Cristina Sauerland, Michaela Feuring-Buske, Bianka Ksienzyk, Utz Krug, Stephanie Schneider, Evelyn Zellmeier, Christian Buske, Clara D. Bloomfield, Purvi M. Kakadia, Michael Unterhalt, Tobias Benthaus, Klaus H. Metzeler, Karsten Spiekermann, Achim Heinecke, Wolfgang Hiddemann, Thomas Buechner, Jan Braess, and Friederike Pastore
- Subjects
Oncology ,Male ,Cancer Research ,Myeloid ,Time Factors ,DNA Mutational Analysis ,Kaplan-Meier Estimate ,Recurrence ,Risk Factors ,Germany ,CEBPA ,Medicine ,Aged, 80 and over ,Age Factors ,Myeloid leukemia ,Nuclear Proteins ,ORIGINAL REPORTS ,Middle Aged ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Phenotype ,Treatment Outcome ,Cytogenetic Analysis ,Female ,Nucleophosmin ,Adult ,medicine.medical_specialty ,NPM1 ,Adolescent ,Risk Assessment ,Disease-Free Survival ,Decision Support Techniques ,Young Adult ,Predictive Value of Tests ,Internal medicine ,Humans ,Genetic Predisposition to Disease ,Aged ,Proportional Hazards Models ,Performance status ,business.industry ,Proportional hazards model ,Cancer ,Reproducibility of Results ,medicine.disease ,fms-Like Tyrosine Kinase 3 ,Immunology ,Mutation ,CCAAT-Enhancer-Binding Proteins ,business - Abstract
Purpose Cytogenetically normal (CN) acute myeloid leukemia (AML) is the largest and most heterogeneous cytogenetic AML subgroup. For the practicing clinician, it is difficult to summarize the prognostic information of the growing number of clinical and molecular markers. Our purpose was to develop a widely applicable prognostic model by combining well-established pretreatment patient and disease characteristics. Patients and Methods Two prognostic indices for CN-AML (PINA), one regarding overall survival (OS; PINAOS) and the other regarding relapse-free survival (RFS; PINARFS), were derived from data of 572 patients with CN-AML treated within the AML Cooperative Group 99 study ( www.aml-score.org ). Results On the basis of age (median, 60 years; range, 17 to 85 years), performance status, WBC count, and mutation status of NPM1, CEBPA, and FLT3-internal tandem duplication, patients were classified into the following three risk groups according to PINAOS and PINARFS: 29% of all patients and 32% of 381 responding patients had low-risk disease (5-year OS, 74%; 5-year RFS, 55%); 56% of all patients and 39% of responding patients had intermediate-risk disease (5-year OS, 28%; 5-year RFS, 27%), and 15% of all patients and 29% of responding patients had high-risk disease (5-year OS, 3%; 5-year RFS, 5%), respectively. PINAOS and PINARFS stratified outcome within European LeukemiaNet genetic groups. Both indices were confirmed on independent data from Cancer and Leukemia Group B/Alliance trials. Conclusion We have developed and validated, to our knowledge, the first prognostic indices specifically designed for adult patients of all ages with CN-AML that combine well-established molecular and clinical variables and that are easily applicable in routine clinical care. The integration of both clinical and molecular markers could provide a basis for individualized patient care through risk-adapted therapy of CN-AML.
- Published
- 2014
38. Comparison of FLT3-ITD Detection By High-Throughput Amplicon Sequencing to Routine Diagnostics - a Retrospective Analysis of AMLCG Study Patients
- Author
-
Christina Sauerland, Evelyn Zellmeier, Karsten Spiekermann, Annika Dufour, Bianka Ksienzyk, Stephanie Schneider, Stefan K. Bohlander, Wolfgang E. Berdel, Kathrin Bräundl, Max Hubmann, Jan Braess, Philipp A. Greif, Wolfgang Hiddemann, Bernhard Wörmann, Egor Harin, Katrin Reiter, Sebastian Vosberg, and Thomas Büchner
- Subjects
Oncology ,medicine.medical_specialty ,biology ,business.industry ,Immunology ,Cell Biology ,Hematology ,Amplicon ,Biochemistry ,Relapse free survival ,body regions ,KMT2A ,Internal medicine ,Induction therapy ,CEBPA ,biology.protein ,Retrospective analysis ,Amplicon sequencing ,medicine ,business ,psychological phenomena and processes ,Flt3 itd - Abstract
Introduction In Acute Myeloid Leukemia (AML) internal tandem duplications (ITD) in the fms-related tyrosine kinase 3 (FLT3) are a frequent event associated with an unfavorable prognosis. At diagnosis, the FLT3-ITD status is routinely assessed by fragment analysis of PCR-amplified cDNA. However, this assay only provides information on the length but not on the position and sequence of the insertion. Therefore, it is attractive to overcome this limitation by the use of high-throughput amplicon sequencing (HTAS) as an alternative strategy for FLT3-ITD detection. To proof the feasibility and accuracy of this approach we performed HTAS on 260 AML patients, of which 250 were FLT3-ITD positive according to routine diagnostics. Patients and Methods All samples were obtained from patients treated on the German Acute Myeloid Leukemia Cooperative Group (AMLCG) trials 1999, 2004 and 2008. All patients received intensive induction therapy with curative intent. At diagnosis, patients had a median age of 60 years (range, 18-80 years). Additional molecular marker were screened routinely: NPM1 mutation (62%), KMT2A-PTD (8%), CEBPA mutations (8%). According to the ELN-classification patients clustered into the following groups: ELN intermediate I (70%), intermediate II (21%) or adverse (9%). A normal karyotype was observed in 71%, while 29% had an aberrant karyotpye. All patient samples were analyzed routinely by FLT3 fragment analysis. For HTAS of FLT3 we used custom FLT3 cDNA primers including barcode and adapter-sequences enabling a one-step PCR-protocol. Sequencing was performed (2x250bp paired end) on a MiSeq (Illumina). Per run, up to 96 samples were sequenced, yielding a median of 79,110 reads per amplicon (range: 31,996 - 259,783). As controls, cDNA from FLT3-ITD positive (Molm-13) and negative (HL60) cell lines were used. Sequencing data were aligned to the FLT3 cDNA reference (NM_004119.2) and FLT3-ITDs were called using Pindel software (version 0.2.5a7). Results Based on the HTAS results obtained from the FLT3 wild-type control HL60, we set the cut-off for ITD detection at a variant allele frequency (VAF) of 0.5%. Using this threshold, all patients which were assessed to be FLT3-ITD negative in diagnostic routine, were also negative in HTAS. FLT3-ITDs were detected by HTAS in 242 out of 250 (97%) patients who were FLT3-ITD positive patients according to routine fragment analysis. For six out of the 8 remaining patients, no valid ITD was detected by HTAS possibly due to one of the following reasons: a low mutational burden resulting in a VAF below the cut-off level (n=4) or a deletion near the ITD (n=2) potentially interfering with ITD-detection. In total, 308 ITDs were detected in 242 patients by HTAS while in these patients 282 ITDs were detected by routine diagnostics. Of note, HTAS missed 13 subclonal ITDs reported in routine, while 39 additional subclonal ITDs were detected by HTAS only. Overall, HTAS detected a higher number of ITDs per patient (mean: 1.27; range: 1-4) compared to fragment analysis (mean: 1.17; range: 1-3). Patients with more than one ITD according to HTAS showed a trend towards shorter overall and relapse free survival (p=0.105, p=0.104 respectively; Figure 1A). The ITD position (i.e. affected FLT3 domain) based on HTAS did not impact on clinical outcome. There was a significant correlation between the FLT3-ITD levels detected by fragment analysis and HTAS (Pearson, R=0.801, p=0.01; Figure 1B). High FLT3-ITD levels measured by fragment analysis had a significant impact on RFS, whereas this effect was not observed for FLT3-ITD levels measured by HTAS. The quantification of FLT3-ITD by HTAS might be hampered by underestimation of the VAFs of long ITDs that are less likely to be mapped correctly compared to shorter ITDs. Conclusion In summary, our study demonstrates the feasibility of HTAS for FLT3-ITD detection in AML. In particular, the identification of subclonal ITDs with high sensitivity provides additional information with potential prognostic value. Thus, HTAS may serve as a robust tool that could be implemented in future diagnostic routines. However, bioinformatic algorithms for ITD detection may need further improvement, e.g. to optimize ITD quantification and to facilitate the detection of ITDs in combination with deletions. Disclosures Hiddemann: Roche: Other: Grants; Genentech: Other: Grants; Roche: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2016
39. Exome sequencing identifies recurring FLT3 N676K mutations in core-binding factor leukemia
- Author
-
Bianka Ksienzyk, Sabrina Opatz, Helmut Blum, Tobias Herold, Philipp A. Greif, Nikola P. Konstandin, Annika Dufour, Sebastian Vosberg, Maria Cristina Sauerland, Wolfgang E. Berdel, Wolfgang Hiddemann, Thomas Büchner, Jan Braess, Bernhard J. Woermann, Evelyn Zellmeier, Karl-Peter Hopfner, Stefan Krebs, Harald Polzer, Stephanie Schneider, Alexander Graf, Karsten Spiekermann, Purvi M. Kakadia, and Stefan K. Bohlander
- Subjects
Adult ,Male ,Models, Molecular ,Adolescent ,Oncogene Proteins, Fusion ,Immunology ,DNA Mutational Analysis ,Molecular Sequence Data ,Apoptosis ,Biology ,medicine.disease_cause ,Biochemistry ,Core Binding Factor beta Subunit ,Fusion gene ,chemistry.chemical_compound ,hemic and lymphatic diseases ,medicine ,MYH11 ,Humans ,Exome ,Benzothiazoles ,Protein Kinase Inhibitors ,Exome sequencing ,Cell Proliferation ,Genetics ,Gene Rearrangement ,Mutation ,Leukemia ,Base Sequence ,Gene Expression Regulation, Leukemic ,Phenylurea Compounds ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Staurosporine ,Cell Transformation, Neoplastic ,RUNX1 ,chemistry ,Amino Acid Substitution ,fms-Like Tyrosine Kinase 3 ,embryonic structures ,Cancer research ,Cytokines ,Female ,Tyrosine kinase - Abstract
The t(8;21) and inv(16)/t(16;16) rearrangements affecting the core-binding factors RUNX1 and CBFB, respectively, are found in 15% to 20% of adult de novo acute myeloid leukemia (AML) cases and are associated with a favorable prognosis. Since the expression of the fusion genes CBFB / MYH11 or RUNX1 / RUNX1T1 alone is not sufficient to cause leukemia, we performed exome sequencing of an AML sample with an inv(16) to identify mutations, which may collaborate with the CBFB/MYH11 fusion during leukemogenesis. We discovered an N676K mutation in the adenosine triphosphate (ATP)-binding domain (tyrosine kinase domain 1 [TKD1]) of the fms-related tyrosine kinase 3 ( FLT3 ) gene. In a cohort of 84 de novo AML patients with a CBFB/MYH11 rearrangement and in 36 patients with a RUNX1/RUNX1T1 rearrangement, the FLT3 N676K mutation was identified in 5 and 1 patients, respectively (5 [6%] of 84; 1 [3%] of 36). The FLT3-N676K mutant alone leads to factor-independent growth in Ba/F3 cells and, together with a concurrent FLT3-ITD (internal tandem duplication), confers resistance to the FLT3 protein tyrosine kinase inhibitors (PTKIs) PKC412 and AC220. Gene expression analysis of AML patients with CBFB / MYH11 rearrangement and FLT3 N676K mutation showed a trend toward a specific expression profile. Ours is the first report of recurring FLT3 N676 mutations in core-binding factor (CBF) leukemias and suggests a defined subgroup of CBF leukemias. This trial was registered at www.clinicaltrials.gov as #NCT00266136.
- Published
- 2013
40. High expression of MZB1 predicts adverse prognosis in chronic lymphocytic leukemia, follicular lymphoma and diffuse large B-cell lymphoma and is associated with a unique gene expression signature
- Author
-
Vindi Jurinovic, Christian Buske, Tobias Herold, Martin Dreyling, Wolfgang Hiddemann, Purvi M. Kakadia, Annika Dufour, Klaus H. Metzeler, Medhanie A. Mulaw, Till Seiler, Karsten Spiekermann, Stefan K. Bohlander, Stephanie Schneider, and Ulrich Mansmann
- Subjects
Cancer Research ,Chronic lymphocytic leukemia ,Follicular lymphoma ,Biology ,hemic and lymphatic diseases ,medicine ,Humans ,Lymphoma, Follicular ,B cell ,Adaptor Proteins, Signal Transducing ,Germinal center ,Myeloid leukemia ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,Lymphoma ,Gene Expression Regulation, Neoplastic ,Leukemia ,medicine.anatomical_structure ,Oncology ,Immunology ,Cancer research ,Cytokines ,Lymphoma, Large B-Cell, Diffuse ,Multiple Myeloma ,Transcriptome ,Diffuse large B-cell lymphoma - Abstract
We recently identified the marginal zone B and B1 cell-specific protein (MZB1) as part of a gene expression signature associated with outcomes in chronic lymphocytic leukemia (CLL). MZB1 is important for B cell function as a key regulator of antibody secretion, calcium homeostasis and adhesion. Therefore, we analyzed the role of MZB1 expression levels in 139 patients with CLL using quantitative real-time polymerase chain reaction (qRT-PCR) and microarray data sets in CLL, follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), multiple myeloma (MM) and acute myeloid leukemia (AML). High MZB1 expression was associated with inferior survival in CLL (hazard ratio [HR]: 1.63 [confidence interval (CI): 1.14-2.33], p = 0.007), FL (221286_s_at HR: 1.16 [CI: 0.98-1.37], p = 0.086; 223565_at: HR: 1.3 [CI: 1.1-1.61], p = 0.015) and DLBCL (221286_s_at: HR: 1.17 [CI: 1.06-1.3], p = 0.003; 223565_at: HR: 1.21 [CI: 1.08-1.35], p = 0.001). In DLBCL MZB1 expression was an additive prognostic marker in a multivariate model including activated B-cell like (ABC) versus germinal center (GCB) subtype. Additionally, MZB1 expression correlated with a unique gene expression pattern. This study is the first to show that the expression level of a single gene has prognostic significance in different lymphoma subtypes. Due to its biological function, MZB1 may play a central role in B cell neoplasms and is a potential target for future therapeutic interventions.
- Published
- 2012
41. GATA2 zinc finger 1 mutations associated with biallelic CEBPA mutations define a unique genetic entity of acute myeloid leukemia
- Author
-
Friederike Schneider, Jan Braess, Andreas Hauser, Helmut Blum, Martin Stanulla, Tobias Herold, Jutta Sturm, Eva Hoster, Wolfgang Hiddemann, Purvi M. Kakadia, Nikola P. Konstandin, Wolfgang E. Berdel, Belay Tizazu, Tobias Benthaus, Thomas Büchner, Evelyn Zellmeier, Stefan K. Bohlander, Karl-Peter Hopfner, Maria Cristina Sauerland, Stefan Krebs, Petra Dörge, Bianka Ksienzyk, Annika Dufour, Alexander Graf, Stephanie Schneider, Bernhard J. Woermann, Karsten Spiekermann, Philipp A. Greif, and Marjan Yaghmaie
- Subjects
Adult ,Models, Molecular ,Transcriptional Activation ,Immunology ,DNA Mutational Analysis ,Karyotype ,Molecular Sequence Data ,Biology ,Biochemistry ,Gene Frequency ,CEBPA ,medicine ,Humans ,Exome ,Amino Acid Sequence ,Gene ,Exome sequencing ,Alleles ,Genetics ,Zinc finger ,Base Sequence ,GATA2 ,Myeloid leukemia ,Zinc Fingers ,Cell Biology ,Hematology ,DNA, Neoplasm ,medicine.disease ,Prognosis ,GATA2 Transcription Factor ,Leukemia ,Leukemia, Myeloid, Acute ,Cytogenetic Analysis ,Mutation ,Cancer research ,CCAAT-Enhancer-Binding Proteins - Abstract
Cytogenetically normal acute myeloid leukemia (CN-AML) with biallelic CEBPA gene mutations (biCEPBA) represents a distinct disease entity with a favorable clinical outcome. So far, it is not known whether other genetic alterations cooperate with biCEBPA mutations during leukemogenesis. To identify additional mutations, we performed whole exome sequencing of 5 biCEBPA patients and detected somatic GATA2 zinc finger 1 (ZF1) mutations in 2 of 5 cases. Both GATA2 and CEBPA are transcription factors crucial for hematopoietic development. Inherited or acquired mutations in both genes have been associated with leukemogenesis. Further mutational screening detected novel GATA2 ZF1 mutations in 13 of 33 biCEBPA-positive CN-AML patients (13/33, 39.4%). No GATA2 mutations were found in 38 CN-AML patients with a monoallelic CEBPA mutation and in 89 CN-AML patients with wild-type CEBPA status. The presence of additional GATA2 mutations (n=10) did not significantly influence the clinical outcome of 26 biCEBPA-positive patients. In reporter gene assays, all tested GATA2 ZF1 mutants showed reduced capacity to enhance CEBPA-mediated activation of transcription, suggesting that the GATA2 ZF1 mutations may collaborate with biCEPBA mutations to deregulate target genes during malignant transformation. We thus provide evidence for a genetically distinct subgroup of CN-AML. The German AML cooperative group trials 1999 and 2008 are registered with the identifiers NCT00266136 and NCT01382147 at www.clinicaltrials.gov.
- Published
- 2012
42. The FLT3ITD mRNA level has a high prognostic impact in NPM1 mutated, but not in NPM1 unmutated, AML with a normal karyotype
- Author
-
Gudrun Mellert, Annika Dufour, Michaela Feuring-Buske, Stefan K. Bohlander, Karsten Spiekermann, Friederike Schneider, Maria Cristina Sauerland, Achim Heinecke, Wolfgang E. Berdel, Michael Unterhalt, Jan Braess, Evelyn Zellmeier, Bernhard Wörmann, Stephanie Schneider, Tobias Benthaus, Thomas Büchner, Wolfgang Hiddemann, Eva Hoster, Purvi M. Kakadia, and Christian Buske
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,NPM1 ,Myeloid ,Immunology ,Karyotype ,Biology ,Biochemistry ,Internal medicine ,medicine ,Humans ,RNA, Messenger ,Survival analysis ,Aged ,Aged, 80 and over ,Proportional hazards model ,Hazard ratio ,Myeloid leukemia ,Nuclear Proteins ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Combined Modality Therapy ,Survival Analysis ,Confidence interval ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,fms-Like Tyrosine Kinase 3 ,Tandem Repeat Sequences ,Mutation ,Female ,Nucleophosmin - Abstract
The impact of a FLT3-internal tandem duplication (FLT3ITD) on prognosis of patients with acute myeloid leukemia (AML) is dependent on the ratio of mutated to wild-type allele. In 648 normal karyotype (NK) AML patients, we found a significant independent effect of the quantitative FLT3ITD mRNA level—measured as (FLT3ITD/wtFLT3)/(FLT3ITD/wtFLT3 + 1)—on outcome. Moreover, this effect was clearly seen in 329 patients with a mutated NPM1 gene (NPM1+), but not in 319 patients without a NPM1 mutation (wtNPM1). In a multivariate Cox regression model, the quantitative FLT3ITD mRNA level showed an independent prognostic impact on overall survival (OS) and relapse-free survival (RFS) only in the NPM1+ subgroup (OS: hazard ratio, 5.9; [95% confidence interval [CI]: 3.1-11.2]; RFS: hazard ratio, 7.5 [95% CI: 3.4-16.5]). The FLT3ITD mRNA level contributes to relapse risk stratification and might help to guide postremission therapy in NPM1-mutated AML.
- Published
- 2012
43. RUNX1 mutations in cytogenetically normal acute myeloid leukemia are associated with poor prognosis and up-regulation of lymphoid genes
- Author
-
Thomas Büchner, Zlatana Pasalic, Jan Braess, Wolfgang E. Berdel, Friederike Schneider, Wolfgang Hiddemann, Nikola P. Konstandin, Purvi M. Kakadia, Tobias Herold, Bianka Ksienzyk, Maria Cristina Sauerland, Bernhard J. Woermann, Stephanie Schneider, Philipp A. Greif, Stefan K. Bohlander, Karsten Spiekermann, Klaus H. Metzeler, and Annika Dufour
- Subjects
Adult ,Male ,NPM1 ,Lineage (genetic) ,Biology ,medicine.disease_cause ,Pathogenesis ,Cohort Studies ,chemistry.chemical_compound ,hemic and lymphatic diseases ,medicine ,Biomarkers, Tumor ,Humans ,Survival rate ,In Situ Hybridization, Fluorescence ,Adaptor Proteins, Signal Transducing ,Aged ,Oligonucleotide Array Sequence Analysis ,Aged, 80 and over ,Mutation ,Myelodysplastic syndromes ,Gene Expression Profiling ,Remission Induction ,Myeloid leukemia ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Survival Rate ,Leukemia, Myeloid, Acute ,RUNX1 ,chemistry ,Core Binding Factor Alpha 2 Subunit ,embryonic structures ,Runx1 ,Mutations ,Acute Myeloid Leukemia ,Cancer research ,Female ,Original Articles and Brief Reports ,Nucleophosmin ,Follow-Up Studies - Abstract
Background The RUNX1 ( AML1 ) gene is a frequent mutational target in myelodysplastic syndromes and acute myeloid leukemia. Previous studies suggested that RUNX1 mutations may have pathological and prognostic implications. Design and Methods We screened 93 patients with cytogenetically normal acute myeloid leukemia for RUNX1 mutations by capillary sequencing of genomic DNA. Mutation status was then correlated with clinical data and gene expression profiles. Results We found that 15 out of 93 (16.1%) patients with cytogenetically normal acute myeloid leukemia had RUNX1 mutations. Seventy-three patients were enrolled in the AMLCG-99 trial and carried ten RUNX1 mutations (13.7%). Among these 73 patients RUNX1 mutations were significantly associated with older age, male sex, absence of NPM1 mutations and presence of MLL -partial tandem duplications. Moreover, RUNX1 -mutated patients had a lower complete remission rate (30% versus 73% P =0.01), lower relapse-free survival rate (3-year relapse-free survival 0% versus 30.4%; P =0.002) and lower overall survival rate (3-year overall survival 0% versus 34.4%; P
- Published
- 2012
44. Age-dependent frequencies of NPM1 mutations and FLT3-ITD in patients with normal karyotype AML (NK-AML)
- Author
-
Purvi M. Kakadia, Christian Buske, Christian Thiede, Thomas Buechner, Jan Braess, Stefan K. Bohlander, Karsten Spiekermann, Friederike Schneider, Michaela Feuring-Buske, Annika Dufour, Tobias Benthaus, Ursula Creutzig, Michel Zwaan, Stephanie Schneider, Dirk Reinhardt, Eva Hoster, Achim Heinecke, Wolfgang E. Berdel, Marry M. van den Heuvel-Eibrink, Wolfgang Hiddemann, Bernhard J. Woermann, Maria Cristina Sauerland, and Pediatrics
- Subjects
Oncology ,Adult ,NPM1 ,medicine.medical_specialty ,Adolescent ,DNA Mutational Analysis ,Karyotype ,Gene mutation ,Young Adult ,hemic and lymphatic diseases ,Internal medicine ,CEBPA ,Medicine ,Humans ,In patient ,Child ,Aged ,Hematology ,business.industry ,Age Factors ,Infant ,Nuclear Proteins ,General Medicine ,Middle Aged ,Prognosis ,Survival Rate ,Leukemia, Myeloid, Acute ,fms-Like Tyrosine Kinase 3 ,Tandem Repeat Sequences ,Child, Preschool ,Cohort ,Immunology ,Mutation ,business ,Nucleophosmin ,Flt3 itd - Abstract
Prognosis of AML in elderly patients is poor due to adverse patient characteristics and comorbidities. In addition, disease-associated parameters reveal differences between older and younger patients with AML. Survival in normal karyotype AML (NK-AML) is influenced by different clinical and molecular markers. The aim of this work was to investigate the frequencies of molecular markers in patients with NK-AML with a focus on NPM1 mutations and FLT3-ITD in different age groups. In the present study, we analyzed the frequencies of mutations of NPM1 and FLT3-ITD in a cohort of 1,321 adult patients and 148 children with AML treated within the AMLCG99, the AML98, and AML04 trials and their distribution in different age groups. Additionally, the frequencies of mutations in CEBPA genes, FLT3-TKD, and MLL-PTD were analyzed in the cohort with NK-AML (n = 729). Our data show that the presence of mutations of NPM1 (from 60% to 40%) and FLT3-ITD (from 50% to 20%) significantly decreased with age in adult AML. Consequently, the proportion of NPM1−/FLT3-ITD− patients increased with age. The decreasing frequency of NPM1 mutations in elderly patients was paralleled by a reduced complete remission (CR) rate in the elderly of 55% compared to 80% in the younger patients. By contrast, the frequencies of other gene mutations, like FLT3-TKD and MLL-PTD, and mutations in CEBPA were not age-dependent. The decreasing frequency of the favorable NPM1 mutations with increasing age may partially explain the worse outcome in the elderly patients. Furthermore, the increasing amount of elderly patients without NPM1 mutations or FLT3-ITD suggests that other molecular and clinical risk factors may influence prognosis in this age group.
- Published
- 2012
45. DNMT3A Mutations Associate with Shorter Survival and Modulate the Prognostic Impact of Mutated NPM1: an Analysis Based on Comprehensive Mutational Screening of 660 AML Patients Treated on German AML Cooperative Group (AMLCG) Trials
- Author
-
Luise Hartmann, Marion Subklewe, Susanne Amler, Andreas Faldum, Annika Dufour, Cristina Sauerland, Michael Fiegl, Tobias Herold, Jan Braess, Kathrin Bräundl, Wolfgang E. Berdel, Evelyn Zellmeier, Bianka Ksienzyk, Stefan K. Bohlander, Klaus H. Metzeler, Bernhard Wörmann, Nikola P. Konstandin, Karsten Spiekermann, Stephanie Schneider, Thomas Büchner, Philipp A. Greif, Utz Krug, Maja Rothenberg-Thurley, and Wolfgang Hiddemann
- Subjects
Oncology ,medicine.medical_specialty ,NPM1 ,Myeloid ,Immunology ,Hazard ratio ,Cytogenetics ,Induction chemotherapy ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,Bioinformatics ,Biochemistry ,medicine.anatomical_structure ,Internal medicine ,Genotype ,medicine ,Missense mutation - Abstract
Background: Mutations in DNA methyltransferase 3A (DNMT3A) are common in acute myeloid leukemia (AML), affecting ~20% of patients (pts) and 30-40% of those with cytogenetically normal (CN-) AML. Although several groups have investigated their prognostic relevance, most studies focused on younger adults (=60 y) AML pts treated on German AML Cooperative Group (AMLCG) protocols, and studied the association between DNMT3A mutations and outcomes. Patients and Methods: We analyzed pretreatment blood or bone marrow specimens from 660 adult AML pts who received intensive induction chemotherapy on two consecutive phase III trials (AMLCG-1999, n=388, and AMLCG-2008, n=272; median age, 57y, range, 18-86y). Sequence variants in DNMT3A exons 7-23 and other genes known to be mutated in myeloid neoplasms were analyzed by multiplexed amplicon resequencing (Agilent Haloplex). Sequencing was performed on an Illumina MiSeq instrument using 2x250bp paired-end reads. Variants were classified as known/putative driver mutations, variants of unknown significance, or known germline polymorphisms based on published data including dbSNP, the Catalogue Of Somatic Mutations In Cancer (COSMIC) and The Cancer Genome Atlas (TCGA). Cytogenetic analyses were performed centrally. Results: We identified 223 DNMT3A mutations in 207/660 pts (31%), including 180/449 pts (40%) with intermediate-risk cytogenetics according to the MRC classification (P 1 type of DNMT3A mutation. DNMT3A mutations tended to be more frequent in older compared to younger pts (35% vs. 28%, P =.08) and were associated with female sex (38% vs 26% in males; P Conclusion: In our cohort of intensively treated AML pts covering a broad age range, we found that DNMT3A mutations associate with inferior survival and modulate the prognostic impact of mutated NPM1, confirming data recently reported by the MRC group (Gale et al., J Clin Oncol 33:2072). In contrast to this and other published reports, we observed no outcome differences between different types of DNMT3A mutations. Information on DNMT3A mutation status further refined the risk stratification of CN-AML based on the NPM1 mutated / FLT3-ITD negative genotype, supporting a role for DNMT3A mutations as a prognostic marker. Figure 1. Figure 1. Disclosures Subklewe: AMGEN Research (Munich): Research Funding. Krug:Boehringer Ingelheim: Research Funding; Novartis; BMS; Roche; Boehringer Ingelheim; Bayer: Honoraria; Sunesis: Speakers Bureau; Sunesis; Clavis Pharma; usa Pharma, Catapult Cell Therapy, Gilead, Roche: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2015
46. Monoallelic CEBPA mutations in normal karyotype acute myeloid leukemia: independent favorable prognostic factor within NPM1 mutated patients
- Author
-
Annika, Dufour, Friederike, Schneider, Eva, Hoster, Tobias, Benthaus, Bianka, Ksienzyk, Stephanie, Schneider, Purvi M, Kakadia, Maria-Cristina, Sauerland, Wolfgang E, Berdel, Thomas, Büchner, Bernhard, Wörmann, Jan, Braess, Marion, Subklewe, Wolfgang, Hiddemann, Stefan K, Bohlander, Karsten, Spiekermann, and X, Schiel
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,NPM1 ,Myeloid ,Adolescent ,Karyotype ,Context (language use) ,Biology ,Young Adult ,Risk Factors ,Internal medicine ,CEBPA ,Genotype ,medicine ,Humans ,Genetic Predisposition to Disease ,Alleles ,Aged ,Aged, 80 and over ,Hematology ,Proportional hazards model ,Homozygote ,Nuclear Proteins ,Epistasis, Genetic ,General Medicine ,Middle Aged ,medicine.disease ,Prognosis ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Mutation ,Cancer research ,CCAAT-Enhancer-Binding Proteins ,Female ,Nucleophosmin - Abstract
We and others have shown that cytogenetically normal (CN)-AML patients with biallelic CEBPA gene mutations (biCEBPA) represent a molecularly distinct group with a favorable prognosis. Patients carrying a monoallelic CEBPA mutation (moCEBPA), however, show no different outcome compared to patients with wildtype CEBPA, and these mutations are frequently associated with mutated NPM1 or FLT3-ITD. So far, no molecular or clinical hallmark has been identified to prognostically distinguish moCEBPA patients from patients with wildtype CEBPA. Therefore, we used the data of 663 CN-AML patients treated within the AMLCG 1999 trial to explore the prognostic value of moCEBPA in the context of concomitant clinical and molecular markers (mutated NPM1, FLT3-ITD). Multiple Cox regression in 515 patients adjusting for all available potential confounders revealed that the NPM1 mutation modified the prognostic value of moCEBPA with respect to overall survival (OS, p = 0.017) and event-free survival (EFS, p = 0.011). MoCEBPA was beneficial in NPM1 mutated patients: adjusted OS–hazard ratio (HR) 0.09, 95% confidence interval (CI) 0.01–0.63, p = 0.016; EFS–HR (95% CI) 0.16 (0.04–0.65), p = 0.010. In contrast, moCEBPA had no prognostic impact in patients with wildtype NPM1: OS–HR (95% CI) 1.08 (0.59–1.97), p = 0.804; EFS–HR (95% CI) 1.12 (0.64–1.96), p = 0.682. We found no prognostic effect modification for moCEBPA by FLT3-ITD. The presence of a moCEBPA mutation was shown to be associated with prolonged survival in NPM1 mutated CN-AML patients. Confirmation of these results in larger studies will clarify whether an additional moCEBPA mutation influences the risk stratification of patients with an NPM1 mutated/FLT3-ITD positive genotype.
- Published
- 2011
47. An eight-gene expression signature for the prediction of survival and time to treatment in chronic lymphocytic leukemia
- Author
-
Wolfgang Hiddemann, Christian Buske, Jan Braess, Silvia Thoene, Markus Schmidberger, Tobias Herold, Till Seiler, Klaus H. Metzeler, Zlatana Pasalic, Stefan K. Bohlander, Michael Schmidt, Stephanie Schneider, Michaela Feuring-Buske, Purvi M. Kakadia, Ulrich Mansmann, Vindi Jurinovic, Karsten Spiekermann, Manuela Bergmann, Anne-Laure Boulesteix, Annika Dufour, and Medhanie A. Mulaw
- Subjects
Adult ,Male ,Cancer Research ,Chronic lymphocytic leukemia ,Expression Signature ,Time to treatment ,Real-Time Polymerase Chain Reaction ,hemic and lymphatic diseases ,Gene expression ,Heterogeneity ,Risk ,Humans ,Medicine ,Survival analysis ,Aged ,Aged, 80 and over ,business.industry ,Gene Expression Profiling ,Hematology ,Middle Aged ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Survival Analysis ,Gene expression profiling ,Leukemia ,Real-time polymerase chain reaction ,Oncology ,Immunology ,Female ,business - Abstract
An eight-gene expression signature for the prediction of survival and time to treatment in chronic lymphocytic leukemia
- Published
- 2011
48. Genomic 5-hydroxymethylcytosine levels correlate with TET2 mutations and a distinct global gene expression pattern in secondary acute myeloid leukemia
- Author
-
Nikola P. Konstandin, Stefan K. Bohlander, Bianka Ksienzyk, A Szwagierczak, Stephanie Schneider, Heinrich Leonhardt, Purvi M. Kakadia, Medhanie A. Mulaw, Tobias Herold, Friederike Schneider, Sebastian Bultmann, Annika Dufour, and Karsten Spiekermann
- Subjects
Genetics ,5-Hydroxymethylcytosine ,Cancer Research ,MLL ,T(10/11)(Q22,Q23) ,5-methylcytosine ,Protein ,Cancers ,DNA ,Gene Expression Profiling ,RUNX1T1 ,Hematology ,Biology ,Dioxygenases ,DNA-Binding Proteins ,chemistry.chemical_compound ,Cytosine ,Leukemia, Myeloid, Acute ,Oncology ,chemistry ,hemic and lymphatic diseases ,Proto-Oncogene Proteins ,Gene expression ,Mutation ,5-Methylcytosine ,Secondary Acute Myeloid Leukemia ,Humans ,In Situ Hybridization, Fluorescence - Abstract
Genomic 5-hydroxymethylcytosine levels correlate with TET2 mutations and a distinct global gene expression pattern in secondary acute myeloid leukemia
- Published
- 2011
49. Expression analysis of genes located in the minimally deleted regions of 13q14 and 11q22-23 in chronic lymphocytic leukemia-unexpected expression pattern of the RHO GTPase activator ARHGAP20
- Author
-
Till Seiler, Michaela Feuring-Buske, Stefan K. Bohlander, Christian Buske, Wolfgang Hiddemann, Ulrich Mansmann, Tobias Herold, Purvi M. Kakadia, Annika Dufour, Stephanie Schneider, Karsten Spiekermann, Medhanie A. Mulaw, Vindi Jurinovic, and Jan Braess
- Subjects
Genetics ,Cancer Research ,Microarray ,GTPase-activating protein ,Chromosomes, Human, Pair 13 ,Chronic lymphocytic leukemia ,Chromosomes, Human, Pair 11 ,GTPase-Activating Proteins ,Gene Dosage ,Biology ,medicine.disease ,Molecular biology ,Gene dosage ,Leukemia, Lymphocytic, Chronic, B-Cell ,Gene Expression Regulation, Neoplastic ,Real-time polymerase chain reaction ,microRNA ,Gene expression ,medicine ,Humans ,Gene ,Sequence Deletion ,Transcription Factors - Abstract
In chronic lymphocytic leukemia (CLL), 13q14 and 11q22-23 deletions are found in 2/3 of the cases. 11q22-23 deletions are associated with poor survival, whereas 13q14 deletions as single abnormality are often found in indolent disease forms. The molecular basis for this difference in prognosis is not known. We examined the 13q14 and 11q22-23 minimally deleted regions (MDRs) for differentially expressed genes by analyzing 154 microarray CLL gene expression data sets. We were able to generate a detailed gene expression map of the MDRs demonstrating a gene dosage effect. Surprisingly, ARHGAP20 encoding the RHO GTPase activating protein 20, which is located in the 11q22-23 MDR, showed-counterintuitively-a significantly higher expression in cases with 11q22-23 deletions compared with cases with no detectable genetic lesion or trisomy 12. Interestingly, cases with 13q14 deletions also had higher ARHGAP20 expression. These expression level changes were confirmed by quantitative PCR in 110 additional CLL samples. The ARHGAP20 gene encodes an evolutionarily conserved protein. In the zebra fish (Danio rerio) genome the syntenic regions of human chromosomal bands 13q14 and 11q22-23 are juxtaposed. The similar expression profiles of ARHGAP20 in 13q14 and 11q22-23 deleted CLL cases suggest a molecular connection and an intriguing mechanism of regulation.
- Published
- 2010
50. Effect of FCGR2A and FCGR3A variants on CLL outcome
- Author
-
David Dornan, Philippe Solal-Celigny, Anna Dmoszynska, Olivia Spleiss, Viktor Rossiev, Marco Montillo, Tadeusz Robak, Annika Dufour, Sergey I. Moiseev, Isabelle Bence-Bruckler, Guillemette Duchateau-Nguyen, Christian H. Geisler, Michael Wenger, Ru Fang Yeh, Loree Larratt, Krzysztof Warzocha, Javier Loscertales, Martin Weisser, Boris V. Afanasiev, Jianguo Zhi, and John Catalano
- Subjects
Adult ,Male ,medicine.medical_specialty ,Genotype ,Chronic lymphocytic leukemia ,Immunology ,FCGR2A ,Biochemistry ,Gastroenterology ,Polymorphism, Single Nucleotide ,Disease-Free Survival ,Antibodies, Monoclonal, Murine-Derived ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Progression-free survival ,Cyclophosphamide ,Aged ,Antibody-dependent cell-mediated cytotoxicity ,Aged, 80 and over ,business.industry ,Hazard ratio ,Receptors, IgG ,FCGR3A ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,Fludarabine ,Treatment Outcome ,Rituximab ,Female ,business ,Vidarabine ,medicine.drug - Abstract
Polymorphisms of activating Fc-γ receptors (FCGRs) on natural killer cells and macrophages result in variable affinity for immunoglobulin G1 monoclonal antibodies and subsequently modulate antibody-dependent cellular cytotoxicity (ADCC) activity. Whether single-nucleotide polymorphisms of FCGRs correlate with survival of chronic lymphocytic leukemia (CLL) patients treated with a monoclonal antibody containing regimen is unclear. We assessed the FCGR3A and FCGR2A genotype of patients enrolled in the REACH trial, where patients received fludarabine and cyclophosphamide (FC) or rituximab plus FC (R-FC). FCGR3A and FCGR2A polymorphisms did not demonstrate prognostic significance in the FC arm (P = .42 and P = .64, respectively) or R-FC arm (P = .41 and P = .88, respectively) with respect to progression free survival. Patients with intermediate affinity genotypes (FV and HR) benefited significantly from addition of rituximab (hazard ratio = 0.55 [0.37-0.8 CI]; P = .0017 and hazard ratio = 0.63 [0.44-0.9 CI]; P = .011, respectively). Similar benefit was suggested for patients with high- affinity VV and HH (hazard ratio = 0.86 [0.4-1.84 CI]; P = .7 and hazard ratio = 0.7 [0.41-1.18 CI]; P = .18, respectively) and low-affinity FF and RR (hazard ratio = 0.85 [0.56-1.29 CI]; P = .44 and hazard ratio = 0.82 [0.47-1.42 CI]; P = .48, respectively). Overall, our results suggest that FCGR2A and FCGR3A polymorphisms do not significantly influence the outcomes of relapsed or refractory CLL patients treated with FC or the monoclonal antibody regimen R-FC.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.