91 results on '"Renate Kirschner"'
Search Results
2. The KRAS-G12D mutation induces metabolic vulnerability in B-cell acute lymphoblastic leukemia
- Author
-
Yan Xu, Houshun Fang, Yao Chen, Yabin Tang, Huiying Sun, Ziqing Kong, Fan Yang, Renate Kirschner-Schwabe, Liang Zhu, Alex Toker, Ning Xiao, Bin-Bing S. Zhou, and Hui Li
- Subjects
Biological sciences ,Biochemistry ,Molecular biology ,Science - Abstract
Summary: Mutations in RAS pathway genes are highly prevalent in acute lymphoblastic leukemia (ALL). However, the effects of RAS mutations on ALL cell growth have not been experimentally characterized, and effective RAS-targeting therapies are being sought after. Here, we found that Reh ALL cells bearing the KRAS-G12D mutation showed increased proliferation rates in vitro but displayed severely compromised growth in mice. Exploring this divergence, proliferation assays with multiple ALL cell lines revealed that the KRAS-G12D rewired methionine and arginine metabolism. Isotope tracing results showed that KRAS-G12D promotes catabolism of methionine and arginine to support anabolism of polyamines and proline, respectively. Chemical inhibition of polyamine biosynthesis selectively killed KRAS-G12D B-ALL cells. Finally, chemically inhibiting AKT/mTOR signaling abrogated the altered amino acid metabolism and strongly promoted the in vivo growth of KRAS-G12D cells in B-ALL xenograft. Our study thus illustrates how hyperactivated AKT/mTOR signaling exerts distinct impacts on hematological malignancies vs. solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
3. Chromatin accessibility landscape of pediatric T‐lymphoblastic leukemia and human T‐cell precursors
- Author
-
Büşra Erarslan‐Uysal, Joachim B Kunz, Tobias Rausch, Paulina Richter‐Pechańska, Ianthe AEM van Belzen, Viktoras Frismantas, Beat Bornhauser, Diana Ordoñez‐Rueada, Malte Paulsen, Vladimir Benes, Martin Stanulla, Martin Schrappe, Gunnar Cario, Gabriele Escherich, Kseniya Bakharevich, Renate Kirschner‐Schwabe, Cornelia Eckert, Tsvetomir Loukanov, Matthias Gorenflo, Sebastian M Waszak, Jean‐Pierre Bourquin, Martina U Muckenthaler, Jan O Korbel, and Andreas E Kulozik
- Subjects
ATAC‐Seq ,chromatin accessibility ,T‐cell development ,T‐cell leukemia ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract We aimed at identifying the developmental stage at which leukemic cells of pediatric T‐ALLs are arrested and at defining leukemogenic mechanisms based on ATAC‐Seq. Chromatin accessibility maps of seven developmental stages of human healthy T cells revealed progressive chromatin condensation during T‐cell maturation. Developmental stages were distinguished by 2,823 signature chromatin regions with 95% accuracy. Open chromatin surrounding SAE1 was identified to best distinguish thymic developmental stages suggesting a potential role of SUMOylation in T‐cell development. Deconvolution using signature regions revealed that T‐ALLs, including those with mature immunophenotypes, resemble the most immature populations, which was confirmed by TF‐binding motif profiles. We integrated ATAC‐Seq and RNA‐Seq and found DAB1, a gene not related to leukemia previously, to be overexpressed, abnormally spliced and hyper‐accessible in T‐ALLs. DAB1‐negative patients formed a distinct subgroup with particularly immature chromatin profiles and hyper‐accessible binding sites for SPI1 (PU.1), a TF crucial for normal T‐cell maturation. In conclusion, our analyses of chromatin accessibility and TF‐binding motifs showed that pediatric T‐ALL cells are most similar to immature thymic precursors, indicating an early developmental arrest.
- Published
- 2020
- Full Text
- View/download PDF
4. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia
- Author
-
Alva Rani James, Michael P. Schroeder, Martin Neumann, Lorenz Bastian, Cornelia Eckert, Nicola Gökbuget, Jutta Ortiz Tanchez, Cornelia Schlee, Konstandina Isaakidis, Stefan Schwartz, Thomas Burmeister, Arend von Stackelberg, Michael A. Rieger, Stefanie Göllner, Martin Horstman, Martin Schrappe, Renate Kirschner-Schwabe, Monika Brüggemann, Carsten Müller-Tidow, Hubert Serve, Altuna Akalin, and Claudia D. Baldus
- Subjects
BCP-ALL subtypes ,DUX4 ,Ph-like ,NH-HeH ,Subtype-specific lncRNAs ,Key signaling pathways ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Long non-coding RNAs (lncRNAs) have emerged as a novel class of RNA due to its diverse mechanism in cancer development and progression. However, the role and expression pattern of lncRNAs in molecular subtypes of B cell acute lymphoblastic leukemia (BCP-ALL) have not yet been investigated. Here, we assess to what extent lncRNA expression and DNA methylation is driving the progression of relapsed BCP-ALL subtypes and we determine if the expression and DNA methylation profile of lncRNAs correlates with established BCP-ALL subtypes. Methods We performed RNA sequencing and DNA methylation (Illumina Infinium microarray) of 40 diagnosis and 42 relapse samples from 45 BCP-ALL patients in a German cohort and quantified lncRNA expression. Unsupervised clustering was applied to ascertain and confirm that the lncRNA-based classification of the BCP-ALL molecular subtypes is present in both our cohort and an independent validation cohort of 47 patients. A differential expression and differential methylation analysis was applied to determine the subtype-specific, relapse-specific, and differentially methylated lncRNAs. Potential functions of subtype-specific lncRNAs were determined by using co-expression-based analysis on nearby (cis) and distally (trans) located protein-coding genes. Results Using an integrative Bioinformatics analysis, we developed a comprehensive catalog of 1235 aberrantly dysregulated BCP-ALL subtype-specific and 942 relapse-specific lncRNAs and the methylation profile of three subtypes of BCP-ALL. The 1235 subtype-specific lncRNA signature represented a similar classification of the molecular subtypes of BCP-ALL in the independent validation cohort. We identified a strong correlation between the DUX4-specific lncRNAs and genes involved in the activation of TGF-β and Hippo signaling pathways. Similarly, Ph-like-specific lncRNAs were correlated with genes involved in the activation of PI3K-AKT, mTOR, and JAK-STAT signaling pathways. Interestingly, the relapse-specific lncRNAs correlated with the activation of metabolic and signaling pathways. Finally, we found 23 promoter methylated lncRNAs epigenetically facilitating their expression levels. Conclusion Here, we describe a set of subtype-specific and relapse-specific lncRNAs from three major BCP-ALL subtypes and define their potential functions and epigenetic regulation. The subtype-specific lncRNAs are reproducible and can effectively stratify BCP-ALL subtypes. Our data uncover the diverse mechanism of action of lncRNAs in BCP-ALL subtypes defining which lncRNAs are involved in the pathogenesis of disease and are relevant for the stratification of BCP-ALL subtypes.
- Published
- 2019
- Full Text
- View/download PDF
5. PDX models recapitulate the genetic and epigenetic landscape of pediatric T‐cell leukemia
- Author
-
Paulina Richter‐Pechańska, Joachim B Kunz, Beat Bornhauser, Caroline von Knebel Doeberitz, Tobias Rausch, Büşra Erarslan‐Uysal, Yassen Assenov, Viktoras Frismantas, Blerim Marovca, Sebastian M Waszak, Martin Zimmermann, Julia Seemann, Margit Happich, Martin Stanulla, Martin Schrappe, Gunnar Cario, Gabriele Escherich, Kseniya Bakharevich, Renate Kirschner‐Schwabe, Cornelia Eckert, Martina U Muckenthaler, Jan O Korbel, Jean‐Pierre Bourquin, and Andreas E Kulozik
- Subjects
ATAC‐Seq ,PDX stability ,T‐ALL ,T‐cell leukemia ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract We compared 24 primary pediatric T‐cell acute lymphoblastic leukemias (T‐ALL) collected at the time of initial diagnosis and relapse from 12 patients and 24 matched patient‐derived xenografts (PDXs). DNA methylation profile was preserved in PDX mice in 97.5% of the promoters (ρ = 0.99). Similarly, the genome‐wide chromatin accessibility (ATAC‐Seq) was preserved remarkably well (ρ = 0.96). Interestingly, both the ATAC regions, which showed a significant decrease in accessibility in PDXs and the regions hypermethylated in PDXs, were associated with immune response, which might reflect the immune deficiency of the mice and potentially the incomplete interaction between murine cytokines and human receptors. The longitudinal approach of this study allowed an observation that samples collected from patients who developed a type 1 relapse (clonal mutations maintained at relapse) preserved their genomic composition; whereas in patients who developed a type 2 relapse (subset of clonal mutations lost at relapse), the preservation of the leukemia's composition was more variable. In sum, this study underlines the remarkable genomic stability, and for the first time documents the preservation of the epigenomic landscape in T‐ALL‐derived PDX models.
- Published
- 2018
- Full Text
- View/download PDF
6. Intragenic amplification of PAX5: a novel subgroup in B-cell precursor acute lymphoblastic leukemia?
- Author
-
Claire Schwab, Karin Nebral, Lucy Chilton, Cristina Leschi, Esmé Waanders, Judith M. Boer, Markéta Žaliová, Rosemary Sutton, Ingegerd Ivanov Öfverholm, Kentaro Ohki, Yuka Yamashita, Stefanie Groeneveld-Krentz, Eva Froňková, Marleen Bakkus, Joelle Tchinda, Thayana da Conceição Barbosa, Grazia Fazio, Wojciech Mlynarski, Agata Pastorczak, Giovanni Cazzaniga, Maria S. Pombo-de-Oliveira, Jan Trka, Renate Kirschner-Schwabe, Toshihiko Imamura, Gisela Barbany, Martin Stanulla, Andishe Attarbaschi, Renate Panzer-Grümayer, Roland P. Kuiper, Monique L. den Boer, Hélène Cavé, Anthony V. Moorman, Christine J. Harrison, and Sabine Strehl
- Subjects
Specialties of internal medicine ,RC581-951 - Published
- 2017
- Full Text
- View/download PDF
7. NOTCH1 mutation, TP53 alteration and myeloid antigen expression predict outcome heterogeneity in children with first relapse of T-cell acute lymphoblastic leukemia
- Author
-
Jana Hof, Corinne Kox, Stefanie Groeneveld-Krentz, Obul R. Bandapalli, Leonid Karawajew, Katharina Schedel, Joachim B. Kunz, Cornelia Eckert, Wolf-Dieter Ludwig, Richard Ratei, Peter Rhein, Günter Henze, Martina U. Muckenthaler, Andreas E. Kulozik, Arend von Stackelberg, and Renate Kirschner-Schwabe
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2017
- Full Text
- View/download PDF
8. Pediatric T-cell lymphoblastic leukemia evolves into relapse by clonal selection, acquisition of mutations and promoter hypomethylation
- Author
-
Joachim B. Kunz, Tobias Rausch, Obul R. Bandapalli, Juliane Eilers, Paulina Pechanska, Stephanie Schuessele, Yassen Assenov, Adrian M. Stütz, Renate Kirschner-Schwabe, Jana Hof, Cornelia Eckert, Arend von Stackelberg, Martin Schrappe, Martin Stanulla, Rolf Koehler, Smadar Avigad, Sarah Elitzur, Rupert Handgretinger, Vladimir Benes, Joachim Weischenfeldt, Jan O. Korbel, Martina U. Muckenthaler, and Andreas E. Kulozik
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Relapsed precursor T-cell acute lymphoblastic leukemia is characterized by resistance against chemotherapy and is frequently fatal. We aimed at understanding the molecular mechanisms resulting in relapse of T-cell acute lymphoblastic leukemia and analyzed 13 patients at first diagnosis, remission and relapse by whole exome sequencing, targeted ultra-deep sequencing, multiplex ligation dependent probe amplification and DNA methylation array. Compared to primary T-cell acute lymphoblastic leukemia, in relapse the number of single nucleotide variants and small insertions and deletions approximately doubled from 11.5 to 26. Targeted ultra-deep sequencing sensitively detected subclones that were selected for in relapse. The mutational pattern defined two types of relapses. While both are characterized by selection of subclones and acquisition of novel mutations, ‘type 1’ relapse derives from the primary leukemia whereas ‘type 2’ relapse originates from a common pre-leukemic ancestor. Relapse-specific changes included activation of the nucleotidase NT5C2 resulting in resistance to chemotherapy and mutations of epigenetic modulators, exemplified by SUZ12, WHSC1 and SMARCA4. While mutations present in primary leukemia and in relapse were enriched for known drivers of leukemia, relapse-specific changes revealed an association with general cancer-promoting mechanisms. This study thus identifies mechanisms that drive progression of pediatric T-cell acute lymphoblastic leukemia to relapse and may explain the characteristic treatment resistance of this condition.
- Published
- 2015
- Full Text
- View/download PDF
9. The activating STAT5B N642H mutation is a common abnormality in pediatric T-cell acute lymphoblastic leukemia and confers a higher risk of relapse
- Author
-
Obul R. Bandapalli, Stephanie Schuessele, Joachim B. Kunz, Tobias Rausch, Adrian M. Stütz, Noa Tal, Ifat Geron, Nava Gershman, Shai Izraeli, Juliane Eilers, Nina Vaezipour, Renate Kirschner-Schwabe, Jana Hof, Arend von Stackelberg, Martin Schrappe, Martin Stanulla, Martin Zimmermann, Rolf Koehler, Smadar Avigad, Rupert Handgretinger, Viktoras Frismantas, Jean Pierre Bourquin, Beat Bornhauser, Jan O. Korbel, Martina U. Muckenthaler, and Andreas E. Kulozik
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2014
- Full Text
- View/download PDF
10. High VLA-4 expression is associated with adverse outcome and distinct gene expression changes in childhood B-cell precursor acute lymphoblastic leukemia at first relapse
- Author
-
Shabnam Shalapour, Jana Hof, Renate Kirschner-Schwabe, Lorenz Bastian, Cornelia Eckert, Javier Prada, Günter Henze, Arend von Stackelberg, and Karl Seeger
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background Resistance to therapy and subsequent relapse remain major challenges in the clinical management of relapsed childhood acute lymphoblastic leukemia. As the bone marrow environment plays an important role in survival and chemotherapy resistance of leukemia cells by activating different signaling pathways, such as the VLA-4 and PI3K/Akt pathways, we studied the prognostic and biological impact of VLA-4 expression in leukemia cells from children with relapsed B-cell precursor acute lymphoblastic leukemia and its influence on the sensitivity of the leukemia cells to drugs.Design and Methods VLA-4 expression was quantified by real-time polymerase chain reaction in leukemia cells from 56 patients with relapsed acute lymphoblastic leukemia enrolled in the ALL-REZ BFM 2002 trial of the Berlin-Frankfurt-Münster study group. Gene expression changes related to VLA-4 expression were investigated by microarray-based mRNA profiling. The effect of VLA-4 signaling on proliferation and drug resistance was studied in co-cultures of leukemia and stromal cells.Results High expression of VLA-4 at first relapse was associated with adverse prognostic factors, poor molecular response to therapy and significantly worse probabilities of event-free and overall survival. VLA-4 expression was an independent prognostic parameter. Comparing gene expression profiles of leukemia cells with high versus low VLA-4 expression, we identified 27 differentially expressed genes primarily involved in the PI3K/Akt, ephrin and Rho GTPase pathways. Blocking of VLA-4 signaling in combination with cytarabine treatment abolished the growth supportive effect of stromal cells.Conclusions Our results show that high VLA-4 expression is a marker of poor prognosis and a potential therapeutic target in children with relapsed acute lymphoblastic leukemia and confirm that cellular interactions and biological effects related to VLA-4 play a decisive role in the survival of leukemia cells and response to therapy.
- Published
- 2011
- Full Text
- View/download PDF
11. Supplementary Figure 1 from Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Author
-
Karl Seeger, Christian Hagemeier, Rainer Spang, Günter Henze, Wolf-Dieter Ludwig, Andreas E. Kulozik, Dennis Kostka, Ute Ungethüm, Arend von Stackelberg, Cornelia Eckert, Leonid Karawajew, Peter Rhein, Jörn Tödling, Claudio Lottaz, and Renate Kirschner-Schwabe
- Abstract
Supplementary Figure 1 from Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Published
- 2023
12. Supplementary Figure 2 from Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Author
-
Karl Seeger, Christian Hagemeier, Rainer Spang, Günter Henze, Wolf-Dieter Ludwig, Andreas E. Kulozik, Dennis Kostka, Ute Ungethüm, Arend von Stackelberg, Cornelia Eckert, Leonid Karawajew, Peter Rhein, Jörn Tödling, Claudio Lottaz, and Renate Kirschner-Schwabe
- Abstract
Supplementary Figure 2 from Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Published
- 2023
13. Supplementary Table 1 from Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Author
-
Karl Seeger, Christian Hagemeier, Rainer Spang, Günter Henze, Wolf-Dieter Ludwig, Andreas E. Kulozik, Dennis Kostka, Ute Ungethüm, Arend von Stackelberg, Cornelia Eckert, Leonid Karawajew, Peter Rhein, Jörn Tödling, Claudio Lottaz, and Renate Kirschner-Schwabe
- Abstract
Supplementary Table 1 from Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Published
- 2023
14. Data from Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Author
-
Karl Seeger, Christian Hagemeier, Rainer Spang, Günter Henze, Wolf-Dieter Ludwig, Andreas E. Kulozik, Dennis Kostka, Ute Ungethüm, Arend von Stackelberg, Cornelia Eckert, Leonid Karawajew, Peter Rhein, Jörn Tödling, Claudio Lottaz, and Renate Kirschner-Schwabe
- Abstract
Purpose: In childhood acute lymphoblastic leukemia (ALL), ∼25% of patients suffer from relapse. In recurrent disease, despite intensified therapy, overall cure rates of 40% remain unsatisfactory and survival rates are particularly poor in certain subgroups. The probability of long-term survival after relapse is predicted from well-established prognostic factors (i.e., time and site of relapse, immunophenotype, and minimal residual disease). However, the underlying biological determinants of these prognostic factors remain poorly understood.Experimental Design: Aiming at identifying molecular pathways associated with these clinically well-defined prognostic factors, we did gene expression profiling on 60 prospectively collected samples of first relapse patients enrolled on the relapse trial ALL-REZ BFM 2002 of the Berlin-Frankfurt-Münster study group.Results: We show here that patients with very early relapse of ALL are characterized by a distinctive gene expression pattern. We identified a set of 83 genes differentially expressed in very early relapsed ALL compared with late relapsed disease. The vast majority of genes were up-regulated and many were late cell cycle genes with a function in mitosis. In addition, samples from patients with very early relapse showed a significant increase in the percentage of S and G2-M phase cells and this correlated well with the expression level of cell cycle genes.Conclusions: Very early relapse of ALL is characterized by an increased proliferative capacity of leukemic blasts and up-regulated mitotic genes. The latter suggests that novel drugs, targeting late cell cycle proteins, might be beneficial for these patients that typically face a dismal prognosis.
- Published
- 2023
15. The combination of subclonal and clonal TP53 and KRAS alterations and poor response to first line therapy identifies ultra-high-risk patients with pediatric T-lymphoblastic leukemia (T-ALL)
- Author
-
Tamara Kempter, Paulina Richter-Pechanska, Katarzyna Michel, Tobias Rausch, Busra Erarslan-Uysal, Cornelia Eckert, Martin Zimmermann, Martin Stanulla, Martin Schrappe, Gunnar Cario, Stefan Köhrer, Renate Kirschner-Schwabe, Jan Korbel, Joachim Kunz, and Andreas Kulozik
- Abstract
It has been the hypothesis of this study that clonal or subclonal variants of TP53 and KRAS that signify ultra-high risk in relapsed pediatric T-ALL indicate a poor prognosis at initial disease. We analyzed two cohorts of 160 and 226 BFM-patients, one consisting of 81 patients who later relapsed and 79 matched non-relapsing controls. Cohort-2 consisted of consecutive patients of whom 30 relapsed. In cohort-1 targeted sequencing revealed TP53 clonal and subclonal variants in 6/81 relapsing but in none of the non-relapsing patients (p=0.014). KRAS alterations occurred in 9/81 relapsing and 2/79 non-relapsing patients (p=0.032). No relapsing patient with TP53 and/or KRAS alterations survived, whereas 19/67 relapsing patients without such variants did (p=0.023). In cohort-2, 196 patients without relapse included 10 with TP53 or KRAS variants, all of whom were non-high risk based on treatment response. However, in the entire group of 386 patients the 188 patients with poor treatment response included 9 with such a variant of whom 8 relapsed and died. Of these, 5 had not undergone stem-cell-transplantation. In conclusion, these data show that subclonal and clonal TP53 and KRAS alterations at initial diagnosis indicate a dismal prognosis in children with T-ALL and poor treatment response.
- Published
- 2023
16. Chemotherapy and mismatch repair deficiency cooperate to fuel TP53 mutagenesis and ALL relapse
- Author
-
Lijuan Du, Pandurang Kolekar, Jiaoyang Cai, Jeffery M. Klco, Houshun Fang, Xiaotu Ma, Bin-Bing S. Zhou, Omkar Pathak, Huiying Sun, Samuel W. Brady, Hong-Zhuan Chen, Yu Liu, Benshang Li, Hui Li, Xiaomeng Li, Lixia Ding, Cornelia Eckert, Renate Kirschner-Schwabe, Malwine J. Barz, Shuhong Shen, Jinghui Zhang, Cai-Wen Duan, Fan Yang, Chao Tang, Arend von Stackelberg, Yao Chen, and Tianyi Wang
- Subjects
Cancer Research ,Chemotherapy ,Thiopurine methyltransferase ,biology ,Mechanism (biology) ,business.industry ,medicine.medical_treatment ,Standard treatment ,Mutagenesis (molecular biology technique) ,Somatic evolution in cancer ,Oncology ,medicine ,biology.protein ,Cancer research ,MISMATCH REPAIR DEFICIENCY ,DNA mismatch repair ,business - Abstract
Chemotherapy is a standard treatment for pediatric acute lymphoblastic leukemia (ALL), which sometimes relapses with chemoresistant features. However, whether acquired drug-resistance mutations in relapsed ALL pre-exist or are induced by treatment remains unknown. Here we provide direct evidence of a specific mechanism by which chemotherapy induces drug-resistance-associated mutations leading to relapse. Using genomic and functional analysis of relapsed ALL we show that thiopurine treatment in mismatch repair (MMR)-deficient leukemias induces hotspot TP53 R248Q mutations through a specific mutational signature (thio-dMMR). Clonal evolution analysis reveals sequential MMR inactivation followed by TP53 mutation in some patients with ALL. Acquired TP53 R248Q mutations are associated with on-treatment relapse, poor treatment response and resistance to multiple chemotherapeutic agents, which could be reversed by pharmacological p53 reactivation. Our findings indicate that TP53 R248Q in relapsed ALL originates through synergistic mutagenesis from thiopurine treatment and MMR deficiency and suggest strategies to prevent or treat TP53-mutant relapse. Zhou and colleagues demonstrate that thiopurine chemotherapy in mismatch repair-deficient ALL cells leads to R248Q TP53 mutations and clonal selection that favors on-treatment ALL relapse and chemoresistance.
- Published
- 2021
17. Subclonal NT5C2 mutations are associated with poor outcomes after relapse of pediatric acute lymphoblastic leukemia
- Author
-
Arend von Stackelberg, Hossein Khiabanian, Jui Wan Loh, Stefanie Groeneveld-Krentz, Adolfo A. Ferrando, Malwine J. Barz, Annabell Szymansky, Jana Hof, Cornelia Eckert, Kathy Astrahantseff, and Renate Kirschner-Schwabe
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Adolescent ,DNA Mutational Analysis ,Immunology ,Purine analogue ,medicine.disease_cause ,Biochemistry ,law.invention ,Young Adult ,Gene Frequency ,Recurrence ,law ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Young adult ,Allele ,Child ,5'-Nucleotidase ,Allele frequency ,Alleles ,Polymerase chain reaction ,Mutation ,business.industry ,Hazard ratio ,Wild type ,Infant ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Child, Preschool ,Female ,business ,Biomarkers - Abstract
Activating mutations in cytosolic 5′-nucleotidase II (NT5C2) are considered to drive relapse formation in acute lymphoblastic leukemia (ALL) by conferring purine analog resistance. To examine the clinical effects of NT5C2 mutations in relapsed ALL, we analyzed NT5C2 in 455 relapsed B-cell precursor ALL patients treated within the ALL-REZ BFM 2002 relapse trial using sequencing and sensitive allele-specific real-time polymerase chain reaction. We detected 110 NT5C2 mutations in 75 (16.5%) of 455 B-cell precursor ALL relapses. Two-thirds of relapses harbored subclonal mutations and only one-third harbored clonal mutations. Event-free survival after relapse was inferior in patients with relapses with clonal and subclonal NT5C2 mutations compared with those without (19% and 25% vs 53%, P < .001). However, subclonal, but not clonal, NT5C2 mutations were associated with reduced event-free survival in multivariable analysis (hazard ratio, 1.89; 95% confidence interval, 1.28-2.69; P = .001) and with an increased rate of nonresponse to relapse treatment (subclonal 32%, clonal 12%, wild type 9%, P < .001). Nevertheless, 27 (82%) of 33 subclonal NT5C2 mutations became undetectable at the time of nonresponse or second relapse, and in 10 (71%) of 14 patients subclonal NT5C2 mutations were undetectable already after relapse induction treatment. These results show that subclonal NT5C2 mutations define relapses associated with high risk of treatment failure in patients and at the same time emphasize that their role in outcome is complex and goes beyond mutant NT5C2 acting as a targetable driver during relapse progression. Sensitive, prospective identification of NT5C2 mutations is warranted to improve the understanding and treatment of this aggressive ALL relapse subtype.
- Published
- 2020
18. Improving Stratification for Children With Late Bone Marrow B-Cell Acute Lymphoblastic Leukemia Relapses With Refined Response Classification and Integration of Genetics
- Author
-
Stefanie Groeneveld-Krentz, Cornelia Eckert, Angelika Eggert, Jean-Pierre Bourquin, Andishe Attarbaschi, Lucie Sramkova, Christina Peters, Arend von Stackelberg, Peter Bader, Günter Henze, Christiane Chen-Santel, Renate Panzer-Grümayer, Renate Kirschner-Schwabe, Kathy Astrahantseff, Nikola Hagedorn, Gunnar Cario, Arndt Borkhardt, and Gabriele Escherich
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Adolescent ,Lymphoblastic Leukemia ,medicine.medical_treatment ,Antineoplastic Agents ,Hematopoietic stem cell transplantation ,Disease ,Disease-Free Survival ,Neoplasm Recurrence ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,hemic and lymphatic diseases ,Internal medicine ,Humans ,Medicine ,Neoplasm ,Child ,business.industry ,Patient Selection ,Hematopoietic Stem Cell Transplantation ,B-cell acute lymphoblastic leukemia ,medicine.disease ,Clinical trial ,medicine.anatomical_structure ,Female ,Bone marrow ,Neoplasm Recurrence, Local ,business - Abstract
PURPOSE Minimal residual disease (MRD) helps to accurately assess when children with late bone marrow relapses of B-cell precursor (BCP) acute lymphoblastic leukemia (ALL) will benefit from allogeneic hematopoietic stem-cell transplantation (allo-HSCT). More detailed dissection of MRD response heterogeneity and the specific genetic aberrations could improve current practice. PATIENTS AND METHODS MRD was assessed after induction treatment and at different times during relapse treatment until allo-HSCT (indicated in poor responders to induction; MRD ≥ 10−3) for patients being treated for late BCP-ALL bone marrow relapses (n = 413; median follow-up, 9.4 years) in the ALL-REZ BFM 2002 trial/registry (ClinicalTrials.gov identifier: NCT00114348 ). RESULTS Patients with both good (MRD < 10−3) and poor responses to induction treatment reached excellent event-free survival (EFS; 72% v 65%) and overall survival (OS; 82% v 74%). Patients with MRD of 10−2 or greater after induction had reduced EFS (56%), and their MRD persisted until allo-HSCT more frequently than it did in patients with MRD of 10−3 or greater to less than 10−2 ( P = .037). Patients with 25% or more leukemic blasts after induction (early nonresponders) had the poorest prognosis (EFS, 22%). Interestingly, patients with MRD of 10−3 or greater before allo-HSCT (late nonresponders) still had an EFS of 50% and OS of 63%, which in principle justifies allo-HSCT in these patients. From a panel of selected candidate genes, TP53 alterations (frequency, 8%) were the only genetic alteration with independent prognostic value in any MRD-based response subgroup. CONCLUSION After induction treatment, MRD-based treatment stratification resulted in excellent survival in patients with late relapsed BCP-ALL. Prognosis could be further improved in very poor responders by intensifying treatment directly after induction. TP53 alterations can be defined as a novel genetic high-risk marker in all MRD response groups in late relapsed BCP-ALL. Here we identified early and late nonresponders to be considered as events in future trials.
- Published
- 2019
19. Risk factors and outcomes in children with high-risk B-cell precursor and T-cell relapsed acute lymphoblastic leukaemia: combined analysis of ALLR3 and ALL-REZ BFM 2002 clinical trials
- Author
-
Lucie Sramkova, Vaskar Saha, Christiane Chen-Santel, Anthony V. Moorman, Andishe Attarbaschi, Renate Kirschner-Schwabe, Stefanie Groeneveld-Krentz, Shekhar Krishnan, Arend von Stackelberg, Peter M. Hoogerbrugge, Martin Zimmermann, Guenter Henze, Catriona Parker, Jean-Pierre Bourquin, Tamas Revesz, Rosemary Sutton, Jeremy Hancock, Cornelia Eckert, Julie Ae Irving, University of Zurich, Eckert, Cornelia, and Saha, Vaskar
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Neoplasm, Residual ,Time Factors ,medicine.medical_treatment ,High-risk ,Gene Dosage ,Graft vs Host Disease ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Targeted therapy ,0302 clinical medicine ,Recurrence ,Risk Factors ,Antineoplastic Combined Chemotherapy Protocols ,1306 Cancer Research ,Child ,Clinical Trials as Topic ,Manchester Cancer Research Centre ,Acute lymphoblastic leukaemia ,Hematopoietic Stem Cell Transplantation ,Stem cell transplantation ,Progression-Free Survival ,medicine.anatomical_structure ,Child, Preschool ,030220 oncology & carcinogenesis ,Disease Progression ,2730 Oncology ,Female ,medicine.medical_specialty ,Adolescent ,T cell ,Karyotype ,610 Medicine & health ,Outcomes ,Risk Assessment ,Disease-Free Survival ,03 medical and health sciences ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,B cell ,Proportional hazards model ,business.industry ,Minimal residual disease ,ResearchInstitutes_Networks_Beacons/mcrc ,Transplantation ,Clinical trial ,030104 developmental biology ,10036 Medical Clinic ,Mutation ,Lymphoblastic leukaemia ,business - Abstract
Aim: Outcomes of children with high-risk (HR) relapsed acute lymphoblastic leukaemia (ALL) (N = 393), recruited to ALLR3 and ALL-REZ BFM 2002 trials, were analysed. Minimal residual disease (MRD) was assessed after induction and at predetermined time points until haematopoietic stem cell transplantation (SCT). Methods: Genetic analyses included karyotype, copy-number alterations and mutation analyses. Ten-year survivals were analysed using Kaplan-Meier and Cox models for multivariable analyses. Results: Outcomes of patients were comparable in ALLR3 and ALL-REZ BFM 2002. The event-free survival of B-cell precursor (BCP) and T-cell ALL (T-ALL) was 22.6% and 26.2% (P = 0.94), respectively, and the overall survival (OS) was 32.6% and 28.2% (P = 0.11), respectively. Induction failures (38%) were associated with deletions of NR3C1 (P = 0.002) and BTG1 (P = 0.03) in BCP-ALL. The disease-free survival (DFS) and OS in patients with good vs poor MRD responses were 57.4% vs 22.6% (P < 0.0001) and 57.8% vs 32.0% (P = 0.0004), respectively. For BCP- and T-ALL, the post-SCT DFS and OS were 42.1% and 56.8% (P = 0.26) and 51.6% and 55.4% (P = 0.67), respectively. The cumulative incidences of post-SCT relapse for BCP- and T-ALL were 36.9% and 17.8% (P = 0.012) and of death were 10.7% and 25.5% (P = 0.013), respectively. Determinants of outcomes after SCT were acute graft versus host disease, pre-SCT MRD (≥10 −3), HR cytogenetics and TP53 alterations in BCP-ALL. Conclusion: Improvements in outcomes for HR ALL relapses require novel compounds in induction therapy to improve remission rates and immune targeted therapy after induction to maintain remission after SCT. Trial registration: ALLR3: NCT00967057; ALL REZ-BFM 2002: NCT00114348
- Published
- 2021
20. Aneuploidy in children with relapsed B‐cell precursor acute lymphoblastic leukaemia: clinical importance of detecting a hypodiploid origin of relapse
- Author
-
Claudia D. Baldus, Jana Hof, Helia J Pimentel-Gutiérrez, Seval Türkmen, Malwine J Pogodzinski, Stefanie Groeneveld-Krentz, Stefan Gattenlöhner, Michael Reiter, Leonid Karawajew, Cornelia Eckert, Christiane Chen-Santel, Arend von Stackelberg, Michael P Schroeder, Oskar A. Haas, Jutta Bradtke, Renia Vagkopoulou, Karin Nebral, and Renate Kirschner-Schwabe
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Adolescent ,Centromere ,Clone (cell biology) ,Aneuploidy ,Dna index ,Kaplan-Meier Estimate ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Risk Factors ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Cluster Analysis ,Humans ,Genetic Predisposition to Disease ,Child ,Survival rate ,B cell ,business.industry ,Infant, Newborn ,Infant ,DNA, Neoplasm ,Hematology ,Prognosis ,medicine.disease ,medicine.anatomical_structure ,Child, Preschool ,030220 oncology & carcinogenesis ,Lymphoblastic leukaemia ,Hypodiploidy ,Female ,Hyperdiploidy ,business ,Multiplex Polymerase Chain Reaction ,030215 immunology - Abstract
Aneuploidy is common in paediatric B-cell precursor acute lymphoblastic leukaemia (ALL). Specific subgroups, such as high hyperdiploidy (>50 chromosomes or DNA Index ≥1·16) and hypodiploidy (
- Published
- 2019
21. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia
- Author
-
Hubert Serve, Renate Kirschner-Schwabe, Alva Rani James, Cornelia Eckert, Carsten Müller-Tidow, Jutta Ortiz Tanchez, Claudia D. Baldus, Martin Schrappe, Stefanie Göllner, Konstandina Isaakidis, Arend von Stackelberg, Michael P Schroeder, Michael A. Rieger, Stefan Schwartz, Monika Brüggemann, Cornelia Schlee, Lorenz Bastian, Nicola Gökbuget, Martin Horstman, Altuna Akalin, Thomas Burmeister, and Martin Neumann
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Microarray ,DUX4 ,Epigenesis, Genetic ,Transcriptome ,Cohort Studies ,0302 clinical medicine ,Bone Marrow ,Recurrence ,BCP-ALL subtypes ,Child ,Promoter Regions, Genetic ,Epigenetically altered lncRNAs ,Subtype-specific lncRNAs ,Hematology ,Methylation ,Ph-like ,lcsh:Diseases of the blood and blood-forming organs ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Berlin ,medicine.anatomical_structure ,Oncology ,Hippo signaling ,030220 oncology & carcinogenesis ,DNA methylation ,Female ,RNA, Long Noncoding ,Key signaling pathways ,Signal transduction ,Technology Platforms ,Metabolic Networks and Pathways ,Adult ,Computational biology ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,medicine ,Biomarkers, Tumor ,Humans ,Epigenetics ,Molecular Biology ,Gene ,PI3K/AKT/mTOR pathway ,B cell ,Integrative bioinformatics ,Base Sequence ,lcsh:RC633-647.5 ,Research ,RNA ,DNA Methylation ,Relapse-specific lncRNAs ,030104 developmental biology ,Cardiovascular and Metabolic Diseases ,NH-HeH - Abstract
Recent studies implicated that long non-coding RNAs (lncRNAs) may play a role in the progression and development of acute lymphoblastic leukemia, however, this role is not yet clear. In order to unravel the role of lncRNAs associated with B-cell precursor Acute Lymphoblastic Leukemia (BCP-ALL) subtypes, we performed transcriptome sequencing and DNA methylation array across 82 BCP-ALL samples from three molecular subtypes (DUX4, Ph-like, and Near Haploid or High Hyperdiploidy). Unsupervised clustering of BCP-ALL samples on the basis of their lncRNAs on transcriptome and DNA methylation profiles revealed robust clusters separating three molecular subtypes. Using extensive computational analysis, we developed a comprehensive catalog of 1235 aberrantly dysregulated BCP-ALL subtype-specific lncRNAs with altered expression and methylation patterns from three subtypes of BCP-ALL. By analyzing the co-expression of subtype-specific lncRNAs and protein-coding genes, we inferred key molecular processes in BCP-ALL subtypes. A strong correlation was identified between the DUX4 specific lncRNAs and activation of TGF-β and Hippo signaling pathways. Similarly, Ph-like specific lncRNAs were correlated with genes involved in activation of PI3K-AKT, mTOR, and JAK-STAT signaling pathways. Interestingly, the relapse-specific differentially expressed lncRNAs correlated with the activation of metabolic and signaling pathways. Finally, we showed a set of epigenetically altered lncRNAs facilitating the expression of tumor genes located at theircislocation. Overall, our study provides a comprehensive set of novel subtype and relapse-specific lncRNAs in BCP-ALL. Our findings suggest a wide range of molecular pathways are associated with lncRNAs in BCP-ALL subtypes and provide a foundation for functional investigations that could lead to new therapeutic approaches.Author SummaryAcute lymphoblastic leukemia is a heterogeneous blood cancer, with multiple molecular subtypes, and with high relapse rate. We are far from the complete understanding of the rationale behind these subtypes and high relapse rate. Long non-coding (lncRNAs) has emerged as a novel class of RNA due to its diverse mechanism in cancer development and progression. LncRNAs does not code for proteins and represent around 70% of human transcripts. Recently, there are a number of studies used lncRNAs expression profile in the classification of various cancers subtypes and displayed their correlation with genomic, epigenetic, pathological and clinical features in diverse cancers. Therefore, lncRNAs can account for heterogeneity and has independent prognostic value in various cancer subtypes. However, lncRNAs defining the molecular subtypes of BCP-ALL are not portrayed yet. Here, we describe a set of relapse and subtype-specific lncRNAs from three major BCP-ALL subtypes and define their potential functions and epigenetic regulation. Our data uncover the diverse mechanism of action of lncRNAs in BCP-ALL subtypes defining how lncRNAs are involved in the pathogenesis of disease and the relevance in the stratification of BCP-ALL subtypes.
- Published
- 2019
22. Advanced Minimal Residual Disease Monitoring for Acute Lymphoblastic Leukemia with Multiplex Mediator Probe PCR
- Author
-
Roland Zengerle, Elena Kipf, Franziska Schlenker, Cornelia Eckert, Michael Lehnert, Felix von Stetten, Renate Kirschner-Schwabe, Marion Fillies, and Nadine Borst
- Subjects
Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,Lymphoblastic Leukemia ,Malignancy ,Real-Time Polymerase Chain Reaction ,Pathology and Forensic Medicine ,Mediator ,hemic and lymphatic diseases ,Internal medicine ,Medicine ,Humans ,Multiplex ,Prospective Studies ,biology ,business.industry ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Minimal residual disease ,body regions ,Duplex (building) ,Cancer cell ,biology.protein ,Molecular Medicine ,Antibody ,business ,Multiplex Polymerase Chain Reaction - Abstract
Acute lymphoblastic leukemia (ALL) is the most frequent malignancy in childhood. Minimal residual disease (MRD) monitoring is an important prognostic factor for ALL treatment response and patient stratification. MRD monitoring uses personalized real-time PCR to measure the amount of cancer cells among normal cells. Due to clonal tumor evolution or secondary rearrangement processes, MRD markers can disappear during treatment, leading to false-negative MRD results and wrong decision-making in personalized treatments. Therefore, monitoring of multiple MRD markers per patient is required. For the first time, the authors present personalized multiplex mediator probe PCR (MP PCR) for MRD monitoring in ALL. These assays can precisely quantify more MRD markers in less sample material. Therefore, clinical outcomes will be less affected by clonal tumor evolution. Personalized duplex MP PCR assays were developed for different genomic MRD markers, including immunoglobulin/T-cell receptor gene rearrangements, gene fusions, and gene deletions. One duplex assay was successfully applied in a prospective patient case and compared with hydrolysis probes. Moreover, the authors increased the multiplex level from duplex to 4-plex and still met the EuroMRD requirements for reliable quantification. In addition, the authors' MRD-MP design guidelines and multiplex workflow facilitate and accelerate MP PCR assay development. This helps the standardization of personal diagnostics.
- Published
- 2021
23. Clinical and genetic characteristics of children with acute lymphoblastic leukemia and Li–Fraumeni syndrome
- Author
-
Stefanie Groeneveld-Krentz, Renate Kirschner-Schwabe, Nienke van Engelen, Marjolijn C.J. Jongmans, Karin Wadt, Simon Bailey, Arend von Stackelberg, Martin Stanulla, Gabriele Escherich, Christian P. Kratz, Cornelia Eckert, Anja Möricke, Tim Ripperger, Lisa Richter, Doris Steinemann, Greta Winter, Olli Lohi, Adela Escudero, and Roula Farah
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Letter ,Adolescent ,Lymphoblastic Leukemia ,Medizin ,Li-Fraumeni Syndrome ,Internal medicine ,Genetics research ,Humans ,Medicine ,Genetic Predisposition to Disease ,Child ,Risk factors ,Acute lymphocytic leukaemia ,Germ-Line Mutation ,business.industry ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Li–Fraumeni syndrome ,Child, Preschool ,Female ,business - Published
- 2021
24. The KRAS-G12D mutation induces metabolic vulnerability in B-cell acute lymphoblastic leukemia
- Author
-
Renate Kirschner-Schwabe, Yan Xu, Houshun Fang, Hui Li, Ziqing Kong, Huiying Sun, Bin-Bing S. Zhou, Yao Chen, Yabin Tang, Liang Zhu, Alex Toker, Ning Xiao, and Fan Yang
- Subjects
Mutation ,Methionine ,Multidisciplinary ,Arginine ,Cell growth ,Mutant ,medicine.disease_cause ,Ornithine decarboxylase ,chemistry.chemical_compound ,chemistry ,Cell culture ,Cancer research ,medicine ,Protein kinase B - Abstract
Mutations in RAS pathway genes are highly prevalent in acute lymphoblastic leukemia (ALL). However, the effects of RAS mutations on ALL cell growth have not been experimentally characterized, and effective RAS-targeting therapies are being sought after. Here, we found that Reh ALL cells bearing the KRAS-G12D mutation showed increased proliferation rates in vitro but displayed severely compromised growth in mice. Exploring this divergence, proliferation assays with multiple ALL cell lines revealed that the KRAS-G12D mutation drives rewiring of methionine and arginine metabolism. Our isotope tracing results showed that mutant KRAS promotes catabolism of methionine and arginine to support anabolism of polyamines and proline, respectively. Chemical inhibition of polyamine biosynthesis selectively killed KRAS-G12D B-ALL cells, implicating the ornithine decarboxylase 1 (ODC1) enzyme as a potential vulnerability for RAS mutant B-ALL. Finally, chemically inhibiting AKT/mTOR signaling abrogated the altered amino acid metabolism and strongly promoted the in vivo growth of KRAS-G12D cells in B-ALL xenograft model. Our study thus illustrates how hyperactivated AKT/mTOR signaling exerts distinct impacts on hematological malignancies vs. solid tumors.
- Published
- 2022
25. Chemotherapy and mismatch repair deficiency cooperate to fuel TP53 mutagenesis and ALL relapse
- Author
-
Fan, Yang, Samuel W, Brady, Chao, Tang, Huiying, Sun, Lijuan, Du, Malwine J, Barz, Xiaotu, Ma, Yao, Chen, Houshun, Fang, Xiaomeng, Li, Pandurang, Kolekar, Omkar, Pathak, Jiaoyang, Cai, Lixia, Ding, Tianyi, Wang, Arend, von Stackelberg, Shuhong, Shen, Cornelia, Eckert, Jeffery M, Klco, Hongzhuan, Chen, Caiwen, Duan, Yu, Liu, Hui, Li, Benshang, Li, Renate, Kirschner-Schwabe, Jinghui, Zhang, and Bin-Bing S, Zhou
- Subjects
Mutagenesis ,Neoplastic Syndromes, Hereditary ,Recurrence ,Primary Immunodeficiency Diseases ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Tumor Suppressor Protein p53 ,Child - Abstract
Chemotherapy is a standard treatment for pediatric acute lymphoblastic leukemia (ALL), which sometimes relapses with chemoresistant features. However, whether acquired drug-resistance mutations in relapsed ALL pre-exist or are induced by treatment remains unknown. Here we provide direct evidence of a specific mechanism by which chemotherapy induces drug-resistance-associated mutations leading to relapse. Using genomic and functional analysis of relapsed ALL we show that thiopurine treatment in mismatch repair (MMR)-deficient leukemias induces hotspot TP53 R248Q mutations through a specific mutational signature (thio-dMMR). Clonal evolution analysis reveals sequential MMR inactivation followed by TP53 mutation in some patients with ALL. Acquired TP53 R248Q mutations are associated with on-treatment relapse, poor treatment response and resistance to multiple chemotherapeutic agents, which could be reversed by pharmacological p53 reactivation. Our findings indicate that TP53 R248Q in relapsed ALL originates through synergistic mutagenesis from thiopurine treatment and MMR deficiency and suggest strategies to prevent or treat TP53-mutant relapse.
- Published
- 2020
26. Chromatin accessibility landscape of pediatric T‐lymphoblastic leukemia and human T‐cell precursors
- Author
-
Gabriele Escherich, Beat Bornhauser, Martin Stanulla, Kseniya Bakharevich, Tsvetomir Loukanov, Matthias Gorenflo, Joachim B. Kunz, V. Frismantas, Jean-Pierre Bourquin, Sebastian M. Waszak, Paulina Richter-Pechanska, Cornelia Eckert, Martina U. Muckenthaler, Jan O. Korbel, Martin Schrappe, Büşra Erarslan-Uysal, Tobias Rausch, Andreas E. Kulozik, Malte Paulsen, Vladimir Benes, Ianthe Aem van Belzen, Renate Kirschner-Schwabe, Gunnar Cario, and Diana Ordoñez-Rueada
- Subjects
0301 basic medicine ,Medicine (General) ,T-cell leukemia ,SUMO protein ,ATAC-seq ,Biology ,QH426-470 ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Chromatin, Epigenetics, Genomics & Functional Genomics ,Article ,03 medical and health sciences ,0302 clinical medicine ,Prophase ,R5-920 ,T‐cell development ,ATAC‐Seq ,medicine ,Genetics ,Humans ,Child ,Gene ,Cancer ,Precursor Cells, T-Lymphoid ,SPI1 ,Articles ,Oncogenes ,medicine.disease ,Chromatin ,Cell biology ,Leukemia ,030104 developmental biology ,chromatin accessibility ,Molecular Medicine ,T‐cell leukemia ,030217 neurology & neurosurgery ,Protein Binding - Abstract
We aimed at identifying the developmental stage at which leukemic cells of pediatric T‐ALLs are arrested and at defining leukemogenic mechanisms based on ATAC‐Seq. Chromatin accessibility maps of seven developmental stages of human healthy T cells revealed progressive chromatin condensation during T‐cell maturation. Developmental stages were distinguished by 2,823 signature chromatin regions with 95% accuracy. Open chromatin surrounding SAE1 was identified to best distinguish thymic developmental stages suggesting a potential role of SUMOylation in T‐cell development. Deconvolution using signature regions revealed that T‐ALLs, including those with mature immunophenotypes, resemble the most immature populations, which was confirmed by TF‐binding motif profiles. We integrated ATAC‐Seq and RNA‐Seq and found DAB1, a gene not related to leukemia previously, to be overexpressed, abnormally spliced and hyper‐accessible in T‐ALLs. DAB1‐negative patients formed a distinct subgroup with particularly immature chromatin profiles and hyper‐accessible binding sites for SPI1 (PU.1), a TF crucial for normal T‐cell maturation. In conclusion, our analyses of chromatin accessibility and TF‐binding motifs showed that pediatric T‐ALL cells are most similar to immature thymic precursors, indicating an early developmental arrest., Analysis of chromatin accessibility of human T‐cell precursors revealed progressive chromatin condensation during maturation. Pediatric T‐ALLs resemble the most immature populations indicating that the epigenetic landscape of this type of leukemia is most similar to the earliest thymic precursors.
- Published
- 2020
27. Clonal evolution mechanisms in NT5C2 mutant relapsed acute lymphoblastic leukemia
- Author
-
Alberto Ambesi-Impiombato, Hossein Khiabanian, Mignon L. Loh, Gannie Tzoneva, Koichi Oshima, Chelsea L. Dieck, Giuseppe Basso, Maria Luisa Sulis, Julie M. Gastier-Foster, Maddalena Paganin, Ilaria Iacobucci, Chioma J. Madubata, Adolfo A. Ferrando, Esmé Waanders, Jiangyan Yu, Raul Rabadan, Charles G. Mullighan, Marta Sanchez-Martin, Motohiro Kato, Katsuyoshi Koh, and Renate Kirschner-Schwabe
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Mutant ,Drug Resistance ,medicine.disease_cause ,Somatic evolution in cancer ,Mice ,0302 clinical medicine ,IMP Dehydrogenase ,Recurrence ,hemic and lymphatic diseases ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Receptor, Notch1 ,Cytotoxicity ,5-Nucleotidase ,5'-Nucleotidase ,Cancer ,Pediatric ,Mutation ,Multidisciplinary ,Guanosine ,Mercaptopurine ,Lymphoblast ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Phenotype ,3. Good health ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Gain of Function Mutation ,Female ,Development of treatments and therapeutic interventions ,Receptor ,Pediatric Cancer ,Childhood Leukemia ,General Science & Technology ,Biology ,Article ,Clonal Evolution ,03 medical and health sciences ,Rare Diseases ,All institutes and research themes of the Radboud University Medical Center ,Nucleotidase ,medicine ,Genetics ,Animals ,Humans ,Cell Proliferation ,Chemotherapy ,Notch1 ,Animal ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,HEK293 Cells ,Drug Resistance, Neoplasm ,Purines ,Disease Models ,Cancer research ,Neoplasm - Abstract
Relapsed acute lymphoblastic leukaemia (ALL) is associated with resistance to chemotherapy and poor prognosis. Gain-of-function mutations in the 5'-nucleotidase, cytosolic II (NT5C2) gene induce resistance to 6-mercaptopurine and are selectively present in relapsed ALL. Yet, the mechanisms involved in NT5C2 mutation-driven clonal evolution during the initiation of leukaemia, disease progression and relapse remain unknown. Here we use a conditional-and-inducible leukaemia model to demonstrate that expression of NT5C2(R367Q), a highly prevalent relapsed-ALL NT5C2 mutation, induces resistance to chemotherapy with 6-mercaptopurine at the cost of impaired leukaemia cell growth and leukaemia-initiating cell activity. The loss-of-fitness phenotype of NT5C2+/R367Q mutant cells is associated with excess export of purines to the extracellular space and depletion of the intracellular purine-nucleotide pool. Consequently, blocking guanosine synthesis by inhibition of inosine-5'-monophosphate dehydrogenase (IMPDH) induced increased cytotoxicity against NT5C2-mutant leukaemia lymphoblasts. These results identify the fitness cost of NT5C2 mutation and resistance to chemotherapy as key evolutionary drivers that shape clonal evolution in relapsed ALL and support a role for IMPDH inhibition in the treatment of ALL.
- Published
- 2018
28. TP53 and KRAS Variants at Initial Diagnosis Identify an Ultra-High Risk Group of Pediatric T-Lymphoblastic Leukemia (T-ALL)
- Author
-
Katarzyna Tomska, Martin Stanulla, Joachim B. Kunz, Andreas E. Kulozik, Renate Kirschner-Schwabe, Tamara Kempter, Cornelia Eckert, Martina U. Muckenthaler, Jan O. Korbel, Gunnar Cario, Gabriele Escherich, Martin Schrappe, Büşra Erarslan-Uysal, Martin Zimmermann, Tobias Rausch, and Paulina Richter-Pechanska
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Lymphoblastic Leukemia ,Immunology ,Cell Biology ,Hematology ,Ultra high risk ,medicine.disease_cause ,Biochemistry ,Internal medicine ,Medicine ,KRAS ,business - Abstract
Introduction Patients who suffer a relapse of pediatric T-cell acute lymphoblastic leukemia (T-ALL) face a dismal prognosis. Prognostic molecular biomarkers that reliably predict the risk of relapse at the time of first diagnosis are not available. Inactivating mutations in TP53 were previously detected in approximately 10% of relapsed patients (Hof et al. J Clin Oncol. 2011) and are invariably associated with fatal outcome (Richter-Pechanska et al. Blood Cancer J. 2017). Mutations in other genes were identified to be either specific for relapse (NT5C2 and CCDC88A) or to be associated with a poor prognosis in relapse (IL7R, KRAS, NRAS, USP7, CNOT3 and MSH6) (Meyer et al. Nat Genet. 2013; Richter-Pechanska et al. Blood Cancer J. 2017). We hypothesized that subclones bearing such mutations can give rise to relapse and analyzed these 9 genes at initial diagnosis of T-ALL with targeted ultra-deep sequencing. Methods Leukemia samples collected at initial diagnosis of 81 children with T-ALL who later relapsed were analyzed. As a control group, we selected 79 children with T-ALL who remained in first remission for at least three years and were matched with regard to treatment response, treatment, age and sex. Targeted deep sequencing was performed by using the Agilent Haloplex High Sensitivity kit with unique molecular identifiers for reliable detection of mutations with very low allele frequencies (average read depth: 1,012x). Results Overall, we detected 75 mutations among 7 targeted genes in 33 / 81 relapsing and 21 / 79 non-relapsing patients. The average allele frequency (AF) of the identified mutations was 25% (0.8% - 83%; SD ± 18%). More than half of the variants (43/75) showed AFs below 30% and were thus classified as subclonal. Interestingly, 7 pathogenic TP53 mutations (subclonal: n=5, clonal: n=2) with AFs of 4.4% - 49.4% were exclusively discovered in 6 patients who experienced a relapse. While 2 of these patients received an allogeneic stem cell transplantation in first remission because of poor treatment response, the remaining 4 patients were treated by chemotherapy in the high-risk (n=1) or medium-risk (n=3) arm. None of the 79 non-relapsing control patients carried TP53 mutations. Consistent with the hypothesis of clonal evolution as a mechanism of relapse in T-ALL, Sanger Sequencing of the relapse sample of one TP53-positive patient confirmed that the subclone harboring the TP53 mutation A159D at initial diagnosis (AF 5.4%) expanded to a major clone (AF 42%) in relapse. The presence of TP53 mutations in two further TP53-positive patients in at least one available post-remission sample is also compatible with clonal selection. However, in a fourth patient the low allele frequency of the TP53 mutation at relapse indicates that the TP53 subclone persisted but did not expand during the development of relapse. In addition to TP53, we identified pathogenic KRAS mutations to be significantly enriched in relapsing patients (9 / 81) compared to non-relapsing patients (2 / 79) at the time of initial diagnosis (chi-squared test, p= 0.032; Table 1). Conclusion Subclonal and clonal mutations in TP53 and KRAS at initial diagnosis were enriched in T-ALL patients who later relapsed and identified approximately 17% of patients suffering a relapse. We thus propose that (subclonal) mutations of TP53 and KRAS may define a subgroup of high-risk T-ALL patients already at the time of first diagnosis. The identification of such mutations may complement the current risk stratification which depends on treatment response and may determine a new molecularly defined subgroup of T-ALLs that may benefit from intensified treatment strategies. Figure 1 Figure 1. Disclosures Schrappe: SigmaTau: Other: research support; Amgen: Other: research support; Servier: Honoraria; Novartis: Honoraria; JazzPharma: Honoraria; Servier: Honoraria, Other: research support; JazzPharma: Honoraria, Other: research support; SHIRE: Other: research support; Novartis: Honoraria, Other: research support. Cario: Novartis: Other: Lecture Fee. Muckenthaler: Silence Therapeutics: Research Funding. Kulozik: Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; BioMedX: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; bluebird bio, Inc.: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Sanofi: Consultancy, Honoraria.
- Published
- 2021
29. Abstract 633: Thiopurines and mismatch repair deficiency cooperate to fuel TP53 mutagenesis and ALL relapse
- Author
-
Yao Chen, Fan Yang, Cai-Wen Duan, Samuel W. Brady, Yu Liu, Shuhong Shen, Tianyi Wang, Jeffery M. Klco, Hui Li, Chao Tang, Cornelia Eckert, Lixia Ding, Lijuan Du, Xiaotu Ma, Huiying Sun, Omkar Pathak, Bin-Bing S. Zhou, Jiaoyang Cai, Arend von Stackelberg, Hong-Zhuan Chen, Pandurang Kolekar, Houshun Fang, Jinghui Zhang, Benshang Li, Renate Kirschner-Schwabe, Xiaomeng Li, and Malwine J. Barz
- Subjects
Cancer Research ,Mutation rate ,Thiopurine methyltransferase ,biology ,business.industry ,Cancer ,medicine.disease ,Somatic evolution in cancer ,MSH6 ,Oncology ,MSH2 ,PMS2 ,medicine ,Cancer research ,biology.protein ,DNA mismatch repair ,business - Abstract
Chemotherapy is curative for most children with acute lymphoblastic leukemia (ALL). Here we provide direct evidence that thiopurine chemotherapeutics can also directly induce drug resistance mutations leading to relapse. Using a large relapsed ALL cohort assembled from Chinese, US and German patients, we found that TP53 R248Q mutations were highly enriched at relapse compared to diagnosis. Relapse-specific TP53 R248Q was associated with the acquisition of MMR deficiency mutations in MSH2, MSH6, or PMS2 and a novel relapse-specific mutational signature. Using isogenic MCF10A cells with or without engineered MSH2 knockout, and the Nalm6 ALL cell line which has native MMR deficiency, we found that this novel signature was caused by a synergistic mutagenic interaction between thiopurine treatment and mismatch repair (MMR) deficiency (called the thio-dMMR signature) that contributes to a hypermutator phenotype and acquisition of TP53 R248Q in residual ALL during remission. Treatment-induced TP53-mutant clones then expand due to broad chemoresistance, leading to eventual relapse. Indeed, thiopurines preferentially induced C>T mutations at the center of NCG trinucleotides, which can lead to TP53 R248Q, and the thiopurine mutation rate was accelerated 2- to 10-fold in MMR-deficient ALL and cell lines. Thiopurine treatment induced C>T mutations preferentially on the transcribed strand, rather than the untranscribed strand, of mRNAs, which further increased the likelihood of TP53 R248Q induction. Further, experimental thiopurine treatment was able to directly induce TP53 R248Q variants in MMR-deficient cultured cells, including Nalm6 and MCF10A MSH2-/-, by activating the thio-dMMR mutational signature, while MMR-proficient MCF10A cells did not experience R248Q induction. The sequential acquisition of MMR deficiency mutations, followed by TP53 mutations, during post-diagnosis ALL evolution was supported by clonal evolution analysis of serial patient samples. p53 R248Q promoted resistance to multiple ALL chemotherapeutic agents, and was associated with on-treatment relapse and poor relapse-treatment response. Our findings indicate that the enrichment of TP53 R248Q in relapsed ALL is due to synergistic mutagenesis from thiopurine treatment and MMR deficiency, followed by selection for TP53 R248Q's chemoresistance phenotype. This suggests that cancer drug resistance mutations may not always pre-exist subclonally at diagnosis, but may be therapy-induced in some patients. Additionally, the qualitative and quantitative mutational signature output of a mutagen (e.g., thiopurines) can vary based on the genetic background. Finally, our findings suggest potential therapeutic strategies, including avoiding thiopurine treatment in MMR-deficient relapses, and therapeutic p53 mutant reactivation, to deal with this genetically-unstable, chemoresistant disease. Citation Format: Fan Yang, Samuel W. Brady, Huiying Sun, Chao Tang, Lijuan Du, Malwine Barz, Xiaotu Ma, Yao Chen, Houshun Fang, Xiaomeng Li, Pandurang Kolekar, Omkar Pathak, Jiaoyang Cai, Lixia Ding, Tianyi Wang, Arend von Stackelberg, Shuhong Shen, Caiwen Duan, Cornelia Eckert, Hongzhuan Chen, Yu Liu, Jeffery M. Klco, Hui Li, Benshang Li, Jinghui Zhang, Renate Kirschner-Schwabe, Bin-Bing S. Zhou. Thiopurines and mismatch repair deficiency cooperate to fuel TP53 mutagenesis and ALL relapse [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 633.
- Published
- 2021
30. Rational Response-Adapted Risk Stratification and Treatment for Children with Late Bone Marrow Relapses of B-Cell Precursor Acute Lymphoblastic Leukaemia: A Report from the ALL-REZ BFM Trial Group
- Author
-
Günter Henze, Arend von Stackelberg, Jean-Pierre Borquin, Lucie Sramkova, Renate Kirschner-Schwabe, Renate Panzer-Grümayer, Kathy Astrahantseff, Cornelia Eckert, Christina Peters, Arndt Borkhardt, Gunnar Cario, Gabriele Escherich, Nikola Hagedorn, Stefanie Groeneveld-Krentz, Peter Bader, Christiane Chen-Santel, and Andishe Attarbaschi
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Proportional hazards model ,Disease ,Minimal residual disease ,body regions ,Transplantation ,Dissection ,Haematopoiesis ,medicine.anatomical_structure ,Informed consent ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Bone marrow ,business - Abstract
Background: Minimal residual disease (MRD) supports a more accurate assessment when children and adolescents with late bone marrow relapses of B-cell precursor (BCP) acute lymphoblastic leukaemia (ALL) will benefit from allogeneic haematopoietic stem cell transplantation (alloHSCT). A more detailed dissection of MRD heterogeneity, dynamics and the specific genetic aberrations involved promises to further improve treatment stratification. Methods: Patients treated according to the ALL-REZ BFM 2002 trial/registry protocol with late BCP ALL bone marrow relapses (n=413) were included. AlloHSCT was indicated for patients with MRD≥10-3 after induction treatment (poor response). Findings: Patients with good (MRD
- Published
- 2019
31. PDX models recapitulate the genetic and epigenetic landscape of pediatric T-cell leukemia
- Author
-
Renate Kirschner-Schwabe, Büşra Erarslan-Uysal, Margit Happich, Tobias Rausch, Gunnar Cario, Yassen Assenov, Joachim B. Kunz, Gabriele Escherich, Beat Bornhauser, Andreas E. Kulozik, Sebastian M. Waszak, Cornelia Eckert, Martina U. Muckenthaler, Blerim Marovca, Martin Schrappe, Jan O. Korbel, Julia Seemann, Martin Stanulla, Kseniya Bakharevich, Viktoras Frismantas, Caroline Von Knebel Doeberitz, Martin Zimmermann, Jean-Pierre Bourquin, Paulina Richter-Pechanska, University of Zurich, and Kulozik, Andreas E
- Subjects
0301 basic medicine ,Medicine (General) ,T-cell leukemia ,ATAC-seq ,610 Medicine & health ,Biology ,QH426-470 ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Chromatin, Epigenetics, Genomics & Functional Genomics ,03 medical and health sciences ,Mice ,Immune system ,R5-920 ,Recurrence ,ATAC‐Seq ,medicine ,Genetics ,Animals ,Humans ,Epigenetics ,Longitudinal Studies ,Research Articles ,Epigenomics ,Cancer ,PDX stability ,Promoter ,medicine.disease ,Chromatin ,Leukemia ,030104 developmental biology ,Gene Expression Regulation ,10036 Medical Clinic ,1313 Molecular Medicine ,Cancer research ,Molecular Medicine ,Heterografts ,T‐cell leukemia ,Neoplasm Transplantation ,T‐ALL ,Haematology ,Research Article - Abstract
We compared 24 primary pediatric T‐cell acute lymphoblastic leukemias (T‐ALL) collected at the time of initial diagnosis and relapse from 12 patients and 24 matched patient‐derived xenografts (PDXs). DNA methylation profile was preserved in PDX mice in 97.5% of the promoters (ρ = 0.99). Similarly, the genome‐wide chromatin accessibility (ATAC‐Seq) was preserved remarkably well (ρ = 0.96). Interestingly, both the ATAC regions, which showed a significant decrease in accessibility in PDXs and the regions hypermethylated in PDXs, were associated with immune response, which might reflect the immune deficiency of the mice and potentially the incomplete interaction between murine cytokines and human receptors. The longitudinal approach of this study allowed an observation that samples collected from patients who developed a type 1 relapse (clonal mutations maintained at relapse) preserved their genomic composition; whereas in patients who developed a type 2 relapse (subset of clonal mutations lost at relapse), the preservation of the leukemia's composition was more variable. In sum, this study underlines the remarkable genomic stability, and for the first time documents the preservation of the epigenomic landscape in T‐ALL‐derived PDX models.
- Published
- 2018
32. Structure and mechanisms of NT5C2 mutations driving thiopurine resistance in relapsed lymphoblastic leukemia
- Author
-
Scott Lew, Farhad Forouhar, Renate Kirschner-Schwabe, Adolfo A. Ferrando, Jayaraman Seetharaman, Chelsea L. Dieck, Gannie Tzoneva, Liang Tong, Zachary Carpenter, Marta Sanchez-Martin, and Alberto Ambesi-Impiombato
- Subjects
0301 basic medicine ,Models, Molecular ,Protein Conformation, alpha-Helical ,Cancer Research ,Antimetabolites, Antineoplastic ,medicine.medical_treatment ,Allosteric regulation ,medicine.disease_cause ,Article ,03 medical and health sciences ,Jurkat Cells ,Structure-Activity Relationship ,0302 clinical medicine ,Allosteric Regulation ,Recurrence ,Catalytic Domain ,Hydrolase ,medicine ,Animals ,Humans ,Gene ,5'-Nucleotidase ,Mutation ,Chemotherapy ,Thiopurine methyltransferase ,biology ,Mechanism (biology) ,Chemistry ,Gene Expression Regulation, Leukemic ,Mercaptopurine ,Cell Biology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Mice, Inbred C57BL ,Cytosol ,030104 developmental biology ,HEK293 Cells ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein - Abstract
Summary Activating mutations in the cytosolic 5′-nucleotidase II gene NT5C2 drive resistance to 6-mercaptopurine in acute lymphoblastic leukemia. Here we demonstrate that constitutively active NT5C2 mutations K359Q and L375F reconfigure the catalytic center for substrate access and catalysis in the absence of allosteric activator. In contrast, most relapse-associated mutations, which involve the arm segment and residues along the surface of the inter-monomeric cavity, disrupt a built-in switch-off mechanism responsible for turning off NT5C2. In addition, we show that the C-terminal acidic tail lost in the Q523X mutation functions to restrain NT5C2 activation. These results uncover dynamic mechanisms of enzyme regulation targeted by chemotherapy resistance-driving NT5C2 mutations, with important implications for the development of NT5C2 inhibitor therapies.
- Published
- 2018
33. Author Correction : The landscape of genomic alterations across childhood cancers
- Author
-
Christian P. Kratz, Benedikt Brors, Manfred Gessler, Jan J. Molenaar, Sebastian M. Waszak, Dietmar R. Lohmann, Vasilisa A. Rudneva, Kristian W. Pajtler, Gideon Zipprich, Daniel Baumhoer, Roland Kappler, Michael Heinold, Matthias Schlesner, Birgit Burkhardt, Stefan Rutkowski, Ewa Koscielniak, Sander Lambo, Sebastian Bender, Stephan Wolf, Michaela Nathrath, Angela J. Waanders, Jürgen Eils, Barbara C. Worst, Angelika Eggert, Michael C. Frühwald, Susanne Gröbner, Cornelia Eckert, Barbara Hutter, Hendrik Witt, Yanling Liu, Paul A. Northcott, Maia Segura-Wang, Pablo Landgraf, Sebastian Brabetz, Danny A. Zwijnenburg, Jenny Wegert, Arndt Borkhardt, Marcel Kool, Gudrun Fleischhack, Renate Kirschner-Schwabe, Kortine Kleinheinz, Christof M. Kramm, Daniel Hübschmann, Pascal Johann, Simone Fulda, Dominik Sturm, Gunther Richter, Peter Lichter, Katja von Hoff, Michaela Kuhlen, Gilles Vassal, Jan O. Korbel, Johannes H. Schulte, Rosario M. Piro, Joachim Weischenfeldt, Reiner Siebert, Udo Kontny, Christian Lawerenz, Gnana Prakash Balasubramanian, David T.W. Jones, Charlotte M. Niemeyer, Uta Dirksen, Lukas Chavez, Serap Erkek, Adam C. Resnick, Frank Westermann, Stefan S. Bielack, Stefan M. Pfister, Ursula D. Weber, Xin Zhou, Marc Zapatka, Cornelis M. van Tilburg, Roland Eils, Jan Koster, Stefan Burdach, Simone Hettmer, Thomas Klingebiel, Andreas E. Kulozik, Olaf Witt, Ivo Buchhalter, Pichai Raman, Claudia Blattmann, Jinghui Zhang, and Elke Pfaff
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Multidisciplinary ,biology ,Published Erratum ,MEDLINE ,Medizin ,Translational research ,biology.organism_classification ,language.human_language ,German ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Paediatric cancer ,030220 oncology & carcinogenesis ,Family medicine ,medicine ,language ,book.journal ,Center (algebra and category theory) ,Memphis ,Psychology ,Developmental neurobiology ,book - Abstract
In this Article, author Benedikt Brors was erroneously associated with affiliation number '8' (Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, USA); the author's two other affiliations (affiliations '3' and '7', both at the German Cancer Research Center (DKFZ)) were correct. This has been corrected online.
- Published
- 2018
34. Intragenic amplification of PAX5: a novel subgroup in B-cell precursor acute lymphoblastic leukemia?
- Author
-
Yuka Yamashita, Grazia Fazio, Hélène Cavé, Sabine Strehl, Kentaro Ohki, Judith M. Boer, Renate Kirschner-Schwabe, Agata Pastorczak, Eva Froňková, Joelle Tchinda, Markéta Žaliová, Marleen Bakkus, Monique L. den Boer, Cristina Leschi, Christine J. Harrison, Lucy Chilton, Stefanie Groeneveld-Krentz, Renate Panzer-Grümayer, Anthony V. Moorman, Wojciech Młynarski, Martin Stanulla, Giovanni Cazzaniga, Andishe Attarbaschi, Maria S. Pombo-de-Oliveira, Thayana Conceição Barbosa, Rosemary Sutton, Jan Trka, Gisela Barbany, Karin Nebral, Claire Schwab, Toshihiko Imamura, Roland P. Kuiper, Esmé Waanders, Ingegerd Ivanov Öfverholm, University of Zurich, Clinical Biology, Hematology, Pediatrics, Schwab, C, Nebral, K, Chilton, L, Leschi, C, Waanders, E, Boer, J, Žaliová, M, Sutton, R, Öfverholm, I, Ohki, K, Yamashita, Y, Groeneveld-Krentz, S, Froňková, E, Bakkus, M, Tchinda, J, Barbosa, T, Fazio, G, Mlynarski, W, Pastorczak, A, Cazzaniga, G, Pombo-de-Oliveira, M, Trka, J, Kirschner-Schwabe, R, Imamura, T, Barbany, G, Stanulla, M, Attarbaschi, A, Panzer-Grümayer, R, Kuiper, R, den Boer, M, Cavé, H, Moorman, A, Harrison, C, and Strehl, S
- Subjects
0301 basic medicine ,MED/03 - GENETICA MEDICA ,business.industry ,Lymphoblastic Leukemia ,2720 Hematology ,hemic and immune systems ,610 Medicine & health ,Hematology ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,medicine.anatomical_structure ,immune system diseases ,10036 Medical Clinic ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,Cancer research ,Medicine ,PAX5 ,business ,PAX5, B-cell precursor acute lymphoblastic leukemia, poor outcome ,B cell - Abstract
Key Points Intragenic PAX5 amplification defines a novel, relapse-prone subtype of B-cell precursor acute lymphoblastic leukemia with a poor outcome.
- Published
- 2017
35. NOTCH1 mutation, TP53 alteration and myeloid antigen expression predict outcome heterogeneity in children with first relapse of T-cell acute lymphoblastic leukemia
- Author
-
Stefanie Groeneveld-Krentz, Obul Reddy Bandapalli, Günter Henze, Cornelia Eckert, Martina U. Muckenthaler, Leonid Karawajew, Richard Ratei, Corinne Kox, Katharina Schedel, Renate Kirschner-Schwabe, Jana Hof, Wolf-Dieter Ludwig, Joachim B. Kunz, Andreas E. Kulozik, Peter Rhein, and Arend von Stackelberg
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Proportional hazards model ,Lymphoblastic Leukemia ,T cell ,Hematopoietic stem cell ,Hematology ,Myeloid antigen ,03 medical and health sciences ,First relapse ,0302 clinical medicine ,medicine.anatomical_structure ,Immunophenotyping ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,Immunology ,Medicine ,business ,Online Only Articles ,030215 immunology - Abstract
Relapse of T-cell acute lymphoblastic leukemia (T-ALL) has a dismal prognosis, with only 20% of afflicted children surviving.[1][1] Children with relapsed T-ALL are commonly treated within high-risk arms of second-line treatment protocols that include mandatory hematopoietic stem cell
- Published
- 2017
36. Intragenic amplification of
- Author
-
Claire, Schwab, Karin, Nebral, Lucy, Chilton, Cristina, Leschi, Esmé, Waanders, Judith M, Boer, Markéta, Žaliová, Rosemary, Sutton, Ingegerd Ivanov, Öfverholm, Kentaro, Ohki, Yuka, Yamashita, Stefanie, Groeneveld-Krentz, Eva, Froňková, Marleen, Bakkus, Joelle, Tchinda, Thayana da Conceição, Barbosa, Grazia, Fazio, Wojciech, Mlynarski, Agata, Pastorczak, Giovanni, Cazzaniga, Maria S, Pombo-de-Oliveira, Jan, Trka, Renate, Kirschner-Schwabe, Toshihiko, Imamura, Gisela, Barbany, Martin, Stanulla, Andishe, Attarbaschi, Renate, Panzer-Grümayer, Roland P, Kuiper, Monique L, den Boer, Hélène, Cavé, Anthony V, Moorman, Christine J, Harrison, and Sabine, Strehl
- Subjects
immune system diseases ,hemic and lymphatic diseases ,hemic and immune systems ,Stimulus Report - Abstract
Intragenic PAX5 amplification defines a novel, relapse-prone subtype of B-cell precursor acute lymphoblastic leukemia with a poor outcome.
- Published
- 2017
37. The activating STAT5B N642H mutation is a common abnormality in pediatric T-cell acute lymphoblastic leukemia and confers a higher risk of relapse
- Author
-
Arend von Stackelberg, Renate Kirschner-Schwabe, Martin Schrappe, Adrian M. Stütz, Jan O. Korbel, Tobias Rausch, Rupert Handgretinger, Viktoras Frismantas, Nina Vaezipour, Smadar Avigad, Martina U. Muckenthaler, Obul Reddy Bandapalli, Noa Tal, Nava Gershman, Jana Hof, Andreas E. Kulozik, Martin Zimmermann, Jean-Pierre Bourquin, Stephanie Schuessele, Rolf Koehler, Ifat Geron, Shai Izraeli, Juliane Eilers, Joachim B. Kunz, Martin Stanulla, Beat Bornhauser, University of Zurich, and Korbel, Jan O
- Subjects
T cell ,Lymphoblastic Leukemia ,2720 Hematology ,STAT5B ,610 Medicine & health ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease_cause ,Malignancy ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,Humans ,Medicine ,Relapse risk ,Child ,Online Only Articles ,Mutation ,business.industry ,hemic and immune systems ,Hematology ,Cell cycle ,medicine.disease ,medicine.anatomical_structure ,10036 Medical Clinic ,Child, Preschool ,Cancer research ,Neoplasm Recurrence, Local ,Abnormality ,business - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes that accounts for approximately 15% of pediatric acute lymphoblastic leukemias. A variety of genetic events affecting cellular processes such as the cell cycle, differentiation and survival have been identified in
- Published
- 2014
38. S130 TRANSIENT SUBCLONES CARRYING NT5C2 MUTATIONS DEFINE A HIGH-RISK PATIENT GROUP WITH POOR OUTCOME IN PEDIATRIC RELAPSED B-CELL PRECURSOR ACUTE LYMPHOBLASTIC LEUKEMIA
- Author
-
Stefanie Groeneveld-Krentz, Adolfo A. Ferrando, A. von Stackelberg, Annabell Szymansky, Renate Kirschner-Schwabe, M.J. Pogodzinski, Jana Hof, and C Eckert
- Subjects
Oncology ,medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,Internal medicine ,Lymphoblastic Leukemia ,medicine ,Hematology ,Patient group ,business ,B cell - Published
- 2019
39. Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL
- Author
-
Martin S. Tallman, Elisabeth Paietta, Gannie Tzoneva, Renate Kirschner-Schwabe, Adolfo A. Ferrando, Jacob M. Rowe, Arianne Perez-Garcia, Valeria Tosello, Maddalena Paganin, Giuseppe Basso, Hossein Khiabanian, Zachary Carpenter, Teresa Palomero, Maddalena Allegretta, Jana Hof, Raul Rabadan, and Janis Racevskis
- Subjects
Nucleotidase activity ,medicine.medical_treatment ,Antineoplastic Agents ,Drug resistance ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Malignant transformation ,5'-nucleotidase ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Nucleotidase ,medicine ,Humans ,Thioguanine ,5'-Nucleotidase ,030304 developmental biology ,0303 health sciences ,Chemotherapy ,Base Sequence ,Mercaptopurine ,leukemia ,Cancer ,General Medicine ,Sequence Analysis, DNA ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,3. Good health ,Leukemia ,HEK293 Cells ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Immunology ,Mutation ,Cancer research ,Arabinonucleosides - Abstract
Acute lymphoblastic leukemia (ALL) is an aggressive hematological tumor resulting from the malignant transformation of lymphoid progenitors. Despite intensive chemotherapy, 20% of pediatric patients and over 50% of adult patients with ALL do not achieve a complete remission or relapse after intensified chemotherapy, making disease relapse and resistance to therapy the most substantial challenge in the treatment of this disease. Using whole-exome sequencing, we identify mutations in the cytosolic 5'-nucleotidase II gene (NT5C2), which encodes a 5'-nucleotidase enzyme that is responsible for the inactivation of nucleoside-analog chemotherapy drugs, in 20/103 (19%) relapse T cell ALLs and 1/35 (3%) relapse B-precursor ALLs. NT5C2 mutant proteins show increased nucleotidase activity in vitro and conferred resistance to chemotherapy with 6-mercaptopurine and 6-thioguanine when expressed in ALL lymphoblasts. These results support a prominent role for activating mutations in NT5C2 and increased nucleoside-analog metabolism in disease progression and chemotherapy resistance in ALL.
- Published
- 2013
40. Mutations and Deletions of the TP53 Gene Predict Nonresponse to Treatment and Poor Outcome in First Relapse of Childhood Acute Lymphoblastic Leukemia
- Author
-
Arend von Stackelberg, Wolf-Dieter Ludwig, Stefanie Krentz, Günter Henze, Gabriele Körner, Cornelia Eckert, Shabnam Shalapour, Karl Seeger, Christian Hagemeier, Peter Rhein, Renate Kirschner-Schwabe, Jana Hof, Leonid Karawajew, and Claudia van Schewick
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,medicine.medical_treatment ,Kaplan-Meier Estimate ,Drug resistance ,medicine.disease_cause ,Polymerase Chain Reaction ,Disease-Free Survival ,law.invention ,Risk Factors ,law ,Internal medicine ,medicine ,Humans ,Multicenter Studies as Topic ,Child ,Gene ,Childhood Acute Lymphoblastic Leukemia ,Polymerase chain reaction ,Proportional Hazards Models ,Clinical Trials as Topic ,Chemotherapy ,Mutation ,Proportional hazards model ,business.industry ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Flow Cytometry ,First relapse ,Treatment Outcome ,Drug Resistance, Neoplasm ,Child, Preschool ,Female ,Tumor Suppressor Protein p53 ,business ,Gene Deletion - Abstract
Purpose In the clinical management of children with relapsed acute lymphoblastic leukemia (ALL), treatment resistance remains a major challenge. Alterations of the TP53 gene are frequently associated with resistance to chemotherapy, but their significance in relapsed childhood ALL has remained controversial because of small studies. Patients and Methods Therefore, we systematically studied 265 first-relapse patients enrolled in the German Acute Lymphoblastic Leukemia Relapse Berlin-Frankfurt-Mü nster 2002 (ALL-REZ BFM 2002) trial for sequence and copy number alterations of the TP53 gene by using direct sequencing and multiplex ligation-dependent probe amplification. Results We observed copy number and sequence alterations of TP53 in 12.4% (27 of 218) of patients with B-cell precursor ALL and 6.4% (three of 47) of patients with T-cell ALL relapse. Backtracking to initial ALL in 23 matched samples revealed that 54% of all TP53 alterations were gained at relapse. Within B-cell precursor ALL, TP53 alterations were consistently associated with nonresponse to chemotherapy (P < .001) and poor event-free survival (P < .001) and overall survival rates (P = .002). TP53 alterations also had a significant impact on survival within intermediate-risk (S2) and high-risk (S3/S4) relapse patients (P = .007 and P = .019, respectively). This prognostic significance of TP53 alterations was confirmed in multivariate analysis. Besides their clinical impact, TP53 alterations were associated with a higher fraction of leukemic cells in S/G2-M phase of the cell cycle at relapse diagnosis. Conclusion Alterations of the TP53 gene are of particular importance in the relapse stage of childhood ALL, in which they independently predict high risk of treatment failure in a significant number of patients. Therefore, they will aid in future risk assessment of children with ALL relapse.
- Published
- 2011
41. Very early/early relapses of acute lymphoblastic leukemia show unexpected changes of clonal markers and high heterogeneity in response to initial and relapse treatment
- Author
-
Rolf Koehler, A. v. Stackelberg, Anja Moericke, Günter Henze, Renate Kirschner-Schwabe, Nikola Hagedorn, André Schrauder, C. R. Bartram, Gunnar Cario, Martin Stanulla, C Eckert, Thomas Flohr, and Martin Schrappe
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Population ,Receptors, Antigen, T-Cell ,Antigen ,Recurrence ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Neoplasm ,Prospective Studies ,Child ,Prospective cohort study ,education ,Childhood Acute Lymphoblastic Leukemia ,Gene Rearrangement ,education.field_of_study ,Genes, Immunoglobulin ,biology ,business.industry ,Hematology ,Gene rearrangement ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Minimal residual disease ,Child, Preschool ,Immunology ,biology.protein ,Female ,Antibody ,business - Abstract
Minimal residual disease (MRD) quantified after induction treatment of childhood acute lymphoblastic leukemia (ALL) predicts risk of relapse. It has been assumed that early relapses derive from a residual population of leukemic cells, which is still present after induction and that relapsed disease will consequently be more resistant to treatment. To test these hypotheses, we performed a prospective study on patients treated according to the frontline-trial ALL-BFM 2000, which used MRD response for risk-group stratification. Patients (n=45) showed a median time to relapse of 1.5 years. In 89% of patients at least one T-cell-receptor/immunoglobulin gene rearrangement chosen for initial MRD quantification remained stable; however, at least one of the preferred markers for MRD stratification at relapse was different to diagnosis in 50% of patients. A similar proportion of very early, early and late relapses appeared to gain a marker at relapse although backtracking-analysis revealed that in 77% of cases, the gained markers were present as small sub-clones at initial diagnosis. Comparing initial and relapse MRD response to induction, 38% of patients showed a similar, 38% a better and 25% a poorer response after relapse. These data demonstrate an unexpectedly high clonal heterogeneity among very early/early relapses and challenge some current assumptions about relapsed ALL.
- Published
- 2011
42. Longitudinal Multilevel Omic Analysis of Pediatric T-ALL Reveals Distinct Mechanisms for Disease Progression in Type 1 and in Type 2 Relapses
- Author
-
Renate Kirschner-Schwabe, Beat Bornhauser, Jean-Pierre Bourquin, Paulina Richter-Pechanska, Joachim B. Kunz, Büşra Erarslan-Uysal, Martin Zimmermann, Viktoras Frismantas, Cornelia Eckert, Kseniya Bakharevich, Tobias Rausch, Martin Schrappe, Caroline Von Knebel Doeberitz, Korbel Jo, Martina U. Muckenthaler, Stephan Fuhrmann, Stefan M. Pfister, Martin Stanulla, Andreas E. Kulozik, G Cario, Gabriele Escherich, and Maria Pamela Dobay
- Subjects
Oncology ,medicine.medical_specialty ,Mutation ,DNA repair ,Immunology ,Clone (cell biology) ,O-6-methylguanine-DNA methyltransferase ,Cell Biology ,Hematology ,Disease ,Biology ,medicine.disease_cause ,Biochemistry ,Somatic evolution in cancer ,Internal medicine ,medicine ,Epigenetics ,Gene - Abstract
We aimed at understanding the relapse-driving processes in pediatric T-ALL and performed an integrated longitudinal multi-level omics analysis of 13 T-ALL patients at initial diagnosis (INI) and relapse (REL). We compared the mutation (SNV/InDels) and copy number alteration (CNA) patterns as well as gene expression, methylation levels and chromatin accessibility by ATAC-Seq. Aberrant expression of T-ALL transcription factors (TAL1, TAL2,LMO2, TLX1, TLX3, NKX2.4 and NKX2.5) was preserved from initial presentation to relapse in all patients. These leukemia-driving events defined the expression patterns, methylation profiles and the chromatin accessibility landscapes. A global differential analysis of the RNA-Seq data (DESeq2, padj We then focused our analysis on the 2 types of relapse in pediatric T-ALL, which we have previously defined on the basis of subclonal mutation profiles (Kunz et al., 2015). These types of relapse are characterized by either clonal evolution of cells derived from the major clone at initial presentation (type 1) or emergence and evolution of a minor initial clone showing a molecular profile that is distinct from the predominant initial clone (type 2). When considering type 1 and type 2 relapses separately we identified a strong trend for type 2 relapses to acquire more mutations (p=0.0879, ttest) than type 1 relapses. Further to the known activating mutations in NT5C2 acquired at relapse by 8/13 patients no other mutations or CNAs were recurrently acquired in the relapses of this group of patients. However, mutations in proto-oncogenes or genes involved in DNA surveillance were acquired by 7/8 type 2 relapse patients in our series. Changes of CNAs also occurred more frequently in type 2 than in type 1 relapses (pval= 0.0267, ttest). This increased complexity on the genetic level was also apparent on the epigenetic level, with an increase of changes in the methylation pattern (mean difference in β value between INI and REL: type 1 - 0.00034; type2 - 0.002 (pval< 0.0001, chi2)), chromatin accessibility (number of differentially accessible ATAC-peaks: type 1 - 4 (0.006%) ; type 2 - 1018 (1.3%); (pval< 0.0001, chi2)) and on the expression level (number of differentially expressed genes: type 1 - 11; type 2 - 111, pval When considering differences between leukemias at the time of initial diagnosis, which later develop either type 1 or type 2 relapses we found 1.016 genes to be differentially expressed (524: up; 492: down in type 1; DE-Seq2: padj In sum, the multilevel omic comparison of pediatric T-ALL that develop either a type 1 or a type 2 relapse show remarkably more complex changes of the genetic and epigenetic profiles during the transition from initial to relapsing disease. Notably, pediatric T-ALLs, who later develop a type 1 relapse display an epigenetic and transcriptomic landscape predicting an upregulation of DNA repair functions, which we suggest to potentially play a role in developing resistance to DNA damaging agents in this type of relapse. Disclosures Muckenthaler: Novartis: Research Funding. Bourquin:Amgen: Other: Travel Support. Kulozik:bluebird bio: Consultancy, Honoraria.
- Published
- 2018
43. Pediatric T-ALLs Developing into a Type 2 Relapse Originate from Cells That Carry the Potential of Variable Maturation into Subclones with Distinct Chromatin Landscapes
- Author
-
Tsvetomir Loukanov, Beat Bornhauser, Jan O. Korbel, Andreas E. Kulozik, Cornelia Eckert, Martina U. Muckenthaler, Matthias Gorenflo, Martin Zimmermann, Renate Kirschner-Schwabe, Joachim B. Kunz, Martin Schrappe, Gunnar Cario, Büşra Erarslan-Uysal, Tobias Rausch, Gabriele Escherich, Martin Stanulla, Kseniya Bakharevich, Viktoras Frismantas, Sebastian M. Waszak, Jean-Pierre Bourquin, and Paulina Richter-Pechanska
- Subjects
Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Chromatin ,Fusion gene ,Leukemia ,Early maturation ,medicine ,Progenitor cell ,CD8 ,Progenitor ,Epigenomics - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy that is classified according to surface marker expression. In order to reveal the cells of origin in pediatric T-ALL and to understand mechanisms of relapse we used ATAC-Seq (Assay for Transposase-Accessible Chromatin Sequencing) to compare chromatin accessibility landscapes of healthy T-cell precursors to those of T-ALL cells obtained at initial diagnosis (INI) and relapse (REL). We have FACS sorted 7 differentiation stages of normal T-cell precursors contained in the thymus of infants undergoing cardiac surgery (DN2, DN3, ISP, DPCD3-, DPCD3+, CD4+ and CD8+) and subjected these to ATAC-Seq. Unsupervised learning by principal component analysis (PCA) clustered sorted populations according to the maturation stage, demonstrating that regulatory chromatin signatures of thymocytes are highly stage-specific and re-shaped during T-cell differentiation. We next compared normal T-cell precursors at different stages of maturation to pediatric T-ALLs and found fundamental differences with 30% of open chromatin regions to be more and 28% being less accessible in T-ALL (DESeq2, padj We then subjected the ATAC-seq data of all matched leukemia samples obtained at initial disease and at relapse to PCA. INI and REL samples derived from the same patient always clustered in close proximity and were separated according to the T-ALL driving fusion genes. A global analysis of differential accessibility revealed only 0.26% of ATAC-regions to be less- or more-accessible at relapse when compared to the matched initial samples (DESeq2, padj Moreover, we trained the deconvolution algorithm CIBERSORT to recognize particular T-cell differentiation stages using ATAC-profiles of the 7 FACS-sorted healthy T-cell populations. We used regulatory chromatin landscape of non-sorted (total) thymus to assess the accuracy of deconvolution. Comparison of predicted fractions in total thymus to FACS measurements revealed highly accurate identification of the maturation stages (r2 = 0.95). CIBERSORT analysis confirmed that the profiles were largely preserved between INI and REL of each sample pair. Notably, however, while in T-ALLs that later developed into a type 1 relapse only one type of early T-cell progenitor dominated the deconvolution profile, T-ALLs that developed into a type 2 relapse showed heterogeneous profiles with contributions of progenitors at different maturation stages. In sum, these epigenomic analyses revealed that the chromatin landscape of normal T-cell precursors evolves in the course of thymic maturation and that the early maturation stages are the likely origin of T-ALL cells. Remarkably, pediatric T-ALLs that later develop a type 2 relapse consist of subclones with a variable profile of chromatin accessibility that define different stages of maturation. These data indicate that T-ALLs with the propensity to develop a type 2 relapse differ from type 1 in that they originate from early precursors that carry the potential of further development into different stages of maturation before the leukemia becomes apparent with a highly subclonal pattern. Disclosures Muckenthaler: Novartis: Research Funding. Bourquin:Amgen: Other: Travel Support. Kulozik:bluebird bio: Consultancy, Honoraria.
- Published
- 2018
44. Abstract PR02: Mechanisms of NT5C2 activating mutations driving thiopurine resistance in relapsed lymphoblastic leukemia
- Author
-
Farhad Forouhar, Adolfo A. Ferrando, Jayaraman Seetharaman, Gannie Tzoneva, Zachary Carpenter, Alberto Ambesi-Impiombato, Marta Sanchez-Martin, Scott Lew, Renate Kirschner-Schwabe, Liang Tong, and Chelsea L. Dieck
- Subjects
Cancer Research ,Conformational change ,Mutation ,Thiopurine methyltransferase ,biology ,Nucleotidase activity ,Chemistry ,Mutant ,Allosteric regulation ,Cancer ,medicine.disease_cause ,medicine.disease ,Oncology ,medicine ,biology.protein ,Cancer research ,Function (biology) - Abstract
Activating mutations in cytosolic 5'-nucleotidase II (NT5C2) drive resistance to 6-mercaptopurine (6-MP) in relapsed acute lymphoblastic leukemia (ALL). These gain-of function alleles encode proteins with increased nucleotidase activity, yet their specific molecular mechanisms are poorly understood. Allosteric activation of NT5C2 is primarily mediated by a marked conformational change in the helix A region, which reconfigures the catalytic center for substrate access and catalysis. Here we demonstrate that constitutively active NT5C2 mutant proteins (K359Q and L375F) adopt active helix A region structures in the absence of allosteric activator. Moreover, crystallographic, structural modeling, genetic, and functional antibody analyses identify a switch-off mechanism mediated by the dynamic mobilization of Asp407 in the tip region of the arm segment which, guided by positively charged residues located in the surface of a basic inter-monomeric cavity exposed upon allosteric activation, turns off NT5C2 by interacting with and disrupting the helical conformation of helix A. Loss of this regulatory mechanism in NT5C2 mutants involving the tip region of the arm segment and the inter-monomeric cavity drives increased nucleotidase activity and allosteric response. Finally, increased allosteric activation by the Q523X mutation results from loss of the NT5C2 C-terminal acidic tail segment, which promotes a closed inactive state in basal conditions. These results uncover dynamic mechanisms responsible for NT5C2 regulation targeted by chemotherapy resistance-driving NT5C2 mutations, with important implications for the development of NT5C2 inhibitor therapies. This abstract is also being presented as Poster 05. Citation Format: Chelsea L. Dieck, Gannie Tzoneva, Farhad Forouhar, Zachary Carpenter, Marta Sanchez-Martin, Alberto Ambesi-Impiombato, Renate Kirschner-Schwabe, Scott Lew, Jayaraman Seetharaman, Liang Tong, Adolfo Ferrando. Mechanisms of NT5C2 activating mutations driving thiopurine resistance in relapsed lymphoblastic leukemia [abstract]. In: Proceedings of the Second AACR Conference on Hematologic Malignancies: Translating Discoveries to Novel Therapies; May 6-9, 2017; Boston, MA. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(24_Suppl):Abstract nr PR02.
- Published
- 2017
45. Gene expression shift towards normal B cells, decreased proliferative capacity and distinct surface receptors characterize leukemic blasts persisting during induction therapy in childhood acute lymphoblastic leukemia
- Author
-
Richard Ratei, Karl Seeger, Christian Hagemeier, Leonid Karawajew, Peter Rhein, Anja Moericke, W.-D. Ludwig, Renate Kirschner-Schwabe, Rainer Spang, Stefanie Scheid, and Martin Schrappe
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Acute lymphocytic leukemia ,Internal medicine ,medicine ,Humans ,Child ,Childhood Acute Lymphoblastic Leukemia ,Cell Proliferation ,Receptors, Interferon ,B-Lymphocytes ,Acute leukemia ,CD11b Antigen ,Hematology ,business.industry ,Gene Expression Profiling ,Cell Cycle ,Infant ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Cell cycle ,medicine.disease ,Minimal residual disease ,Gene expression profiling ,Methotrexate ,Cytokine ,Oncology ,Child, Preschool ,Immunology ,Prednisone ,Female ,Blast Crisis ,business - Abstract
In childhood acute lymphoblastic leukemia (ALL), persistence of leukemic blasts during therapy is of crucial prognostic significance. In the present study, we address molecular and cell biologic features of blasts persisting after 1 week of induction glucocorticoid therapy. Genome-wide gene expression analysis of leukemic samples from precursor B-cell ALL patients (n=18) identified a set of genes differentially expressed in blasts at diagnosis day 0 (d0) and persisting on day 8 (d8). Expression changes indicate a shift towards mature B cells, inhibition of cell cycling and increased expression of adhesion (CD11b/ITGAM) and cytokine (CD119/IFNGR1) receptors. A direct comparison with normal B cells, which are largely therapy resistant, confirmed the differentiation shift at the mRNA (n=10) and protein (n=109) levels. Flow cytometric analysis in independent cohorts of patients confirmed both a decreased proliferative activity (n=13) and the upregulation of CD11b and CD119 (n=29) in d8 blasts. The differentiation shift and low proliferative activity in d8 blasts may account for the persistence of blasts during therapy and affect their sensitivity to further therapeutic treatment. CD11b and CD119 are potential specific markers for d8 blast persistence and detection of minimal residual disease, which warrant further investigation.
- Published
- 2007
46. Prenatal manifestation of pancytopenia in Pearson marrow-pancreas syndrome caused by a mitochondrial DNA deletion
- Author
-
Karl Seeger, Lena Wronski, Shabnam Shalapour, Katharina Blumchen, Javier Prada, Markus Schuelke, Martin Brauer, Almut Giese, Pablo Hernáiz Driever, Renate Kirschner-Schwabe, and Günter Henze
- Subjects
Mitochondrial DNA ,Pancytopenia ,Infant ,Pancreatic Diseases ,Syndrome ,Biology ,medicine.disease ,DNA, Mitochondrial ,Fetal Diseases ,medicine.anatomical_structure ,Pregnancy ,Prenatal Diagnosis ,Pearson marrow-pancreas syndrome ,Genetics ,medicine ,Cancer research ,Humans ,Female ,Chromosome Deletion ,Pancreas ,Bone Marrow Diseases ,Genetics (clinical) - Published
- 2007
47. Pediatric T-cell lymphoblastic leukemia evolves into relapse by clonal selection, acquisition of mutations and promoter hypomethylation
- Author
-
Joachim Weischenfeldt, Juliane Eilers, Arend von Stackelberg, Joachim B. Kunz, Renate Kirschner-Schwabe, Rolf Koehler, Smadar Avigad, Martin Schrappe, Tobias Rausch, Rupert Handgretinger, Sarah Elitzur, Vladimir Benes, Cornelia Eckert, Martina U. Muckenthaler, Yassen Assenov, Jana Hof, Andreas E. Kulozik, Obul Reddy Bandapalli, Adrian M. Stütz, Jan O. Korbel, Stephanie Schuessele, Paulina Pechanska, and Martin Stanulla
- Subjects
Male ,Adolescent ,medicine.medical_treatment ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease_cause ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit ,hemic and lymphatic diseases ,medicine ,Humans ,Multiplex ligation-dependent probe amplification ,Epigenetics ,Child ,Promoter Regions, Genetic ,Exome sequencing ,Mutation ,Chemotherapy ,Editorials ,DNA, Neoplasm ,Hematology ,DNA Methylation ,medicine.disease ,Leukemia ,Child, Preschool ,DNA methylation ,SMARCA4 ,Cancer research ,Female - Abstract
Relapsed precursor T-cell acute lymphoblastic leukemia is characterized by resistance against chemotherapy and is frequently fatal. We aimed at understanding the molecular mechanisms resulting in relapse of T-cell acute lymphoblastic leukemia and analyzed 13 patients at first diagnosis, remission and relapse by whole exome sequencing, targeted ultra-deep sequencing, multiplex ligation dependent probe amplification and DNA methylation array. Compared to primary T-cell acute lymphoblastic leukemia, in relapse the number of single nucleotide variants and small insertions and deletions approximately doubled from 11.5 to 26. Targeted ultra-deep sequencing sensitively detected subclones that were selected for in relapse. The mutational pattern defined two types of relapses. While both are characterized by selection of subclones and acquisition of novel mutations, 'type 1' relapse derives from the primary leukemia whereas 'type 2' relapse originates from a common pre-leukemic ancestor. Relapse-specific changes included activation of the nucleotidase NT5C2 resulting in resistance to chemotherapy and mutations of epigenetic modulators, exemplified by SUZ12, WHSC1 and SMARCA4. While mutations present in primary leukemia and in relapse were enriched for known drivers of leukemia, relapse-specific changes revealed an association with general cancer-promoting mechanisms. This study thus identifies mechanisms that drive progression of pediatric T-cell acute lymphoblastic leukemia to relapse and may explain the characteristic treatment resistance of this condition.
- Published
- 2015
- Full Text
- View/download PDF
48. Expression of Late Cell Cycle Genes and an Increased Proliferative Capacity Characterize Very Early Relapse of Childhood Acute Lymphoblastic Leukemia
- Author
-
Dennis Kostka, Karl Seeger, Rainer Spang, Christian Hagemeier, Andreas E. Kulozik, Ute Ungethüm, Cornelia Eckert, Leonid Karawajew, Claudio Lottaz, Jörn Tödling, Arend von Stackelberg, Peter Rhein, Günter Henze, Wolf-Dieter Ludwig, and Renate Kirschner-Schwabe
- Subjects
G2 Phase ,Oncology ,Cancer Research ,medicine.medical_specialty ,Cell Cycle Proteins ,Immunophenotyping ,Predictive Value of Tests ,Recurrence ,Acute lymphocytic leukemia ,Internal medicine ,Gene expression ,medicine ,Humans ,Prospective Studies ,Child ,Childhood Acute Lymphoblastic Leukemia ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Gene Expression Profiling ,Cell Cycle ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Cell cycle ,Prognosis ,medicine.disease ,Minimal residual disease ,Cell Cycle Gene ,Up-Regulation ,Gene expression profiling ,Immunology ,business ,Cell Division - Abstract
Purpose: In childhood acute lymphoblastic leukemia (ALL), ∼25% of patients suffer from relapse. In recurrent disease, despite intensified therapy, overall cure rates of 40% remain unsatisfactory and survival rates are particularly poor in certain subgroups. The probability of long-term survival after relapse is predicted from well-established prognostic factors (i.e., time and site of relapse, immunophenotype, and minimal residual disease). However, the underlying biological determinants of these prognostic factors remain poorly understood. Experimental Design: Aiming at identifying molecular pathways associated with these clinically well-defined prognostic factors, we did gene expression profiling on 60 prospectively collected samples of first relapse patients enrolled on the relapse trial ALL-REZ BFM 2002 of the Berlin-Frankfurt-Münster study group. Results: We show here that patients with very early relapse of ALL are characterized by a distinctive gene expression pattern. We identified a set of 83 genes differentially expressed in very early relapsed ALL compared with late relapsed disease. The vast majority of genes were up-regulated and many were late cell cycle genes with a function in mitosis. In addition, samples from patients with very early relapse showed a significant increase in the percentage of S and G2-M phase cells and this correlated well with the expression level of cell cycle genes. Conclusions: Very early relapse of ALL is characterized by an increased proliferative capacity of leukemic blasts and up-regulated mitotic genes. The latter suggests that novel drugs, targeting late cell cycle proteins, might be beneficial for these patients that typically face a dismal prognosis.
- Published
- 2006
49. Identification of a genetically defined ultra-high-risk group in relapsed pediatric T-lymphoblastic leukemia
- Author
-
Martin Stanulla, C Eckert, Renate Kirschner-Schwabe, Paulina Richter-Pechanska, Joachim B. Kunz, Andreas E. Kulozik, Gunnar Cario, Martin Zimmermann, G Scapinello, Judit C. Sági, Martin Schrappe, Obul Reddy Bandapalli, Martina U. Muckenthaler, Jana Hof, Jan O. Korbel, Vladimir Benes, Tobias Rausch, and Elena Orlova
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Oncology ,medicine.medical_specialty ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease_cause ,Disease-Free Survival ,03 medical and health sciences ,Risk Factors ,Internal medicine ,Gene duplication ,medicine ,Humans ,Child ,Interleukin-7 receptor ,Hematology ,Prognosis ,medicine.disease ,MSH6 ,Exact test ,Leukemia ,030104 developmental biology ,Child, Preschool ,Immunology ,Original Article ,KRAS - Abstract
In the search for genes that define critical steps of relapse in pediatric T-cell acute lymphoblastic leukemia (T-ALL) and can serve as prognostic markers, we performed targeted sequencing of 313 leukemia-related genes in 214 patients: 67 samples collected at the time of relapse and 147 at initial diagnosis. As relapse-specific genetic events, we identified activating mutations in NT5C2 (P=0.0001, Fisher’s exact test), inactivation of TP53 (P=0.0007, Fisher’s exact test) and duplication of chr17:q11.2-24.3 (P=0.0068, Fisher’s exact test) in 32/67 of T-ALL relapse samples. Alterations of TP53 were frequently homozygous events, which significantly correlated with higher rates of copy number alterations in other genes compared with wild-type TP53 (P=0.0004, Mann–Whitney’s test). We subsequently focused on mutations with prognostic impact and identified genes governing DNA integrity (TP53, n=8; USP7, n=4; MSH6, n=4), having key roles in the RAS signaling pathway (KRAS, NRAS, n=8), as well as IL7R (n=4) and CNOT3 (n=4) to be exclusively mutated in fatal relapses. These markers recognize 24/49 patients with a second event. In 17 of these patients with mostly refractory relapse and dire need for efficient treatment, we identified candidate targets for personalized therapy with p53 reactivating compounds, MEK inhibitors or JAK/STAT-inhibitors that may be incorporated in future treatment strategies.
- Published
- 2017
50. Ras pathway mutations are prevalent in relapsed childhood acute lymphoblastic leukemia and confer sensitivity to MEK inhibition
- Author
-
Christina Halsey, Arend von Stackelberg, Stefanie Groeneveld-Krentz, Jana Hof, Lynne Minto, Isabella Swidenbank, Helen J. Blair, Elizabeth Matheson, Christine J. Harrison, Julie Irving, Marian Case, James M. Allan, Frida Ponthan, Josef Vormoor, Cornelia Eckert, and Renate Kirschner-Schwabe
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Immunology ,Mice, Transgenic ,Mice, SCID ,medicine.disease_cause ,Biochemistry ,Mice ,Gene Frequency ,Mice, Inbred NOD ,Recurrence ,Acute lymphocytic leukemia ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Child ,Protein Kinase Inhibitors ,Mutation ,Clinical Trials as Topic ,Lymphoid Neoplasia ,business.industry ,Wild type ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,MAP Kinase Kinase Kinases ,Xenograft Model Antitumor Assays ,PTPN11 ,Genes, ras ,Ras Signaling Pathway ,Drug Resistance, Neoplasm ,Cancer research ,Selumetinib ,Benzimidazoles ,KRAS ,business ,Signal Transduction - Abstract
For most children who relapse with acute lymphoblastic leukemia (ALL), the prognosis is poor, and there is a need for novel therapies to improve outcome. We screened samples from children with B-lineage ALL entered into the ALL-REZ BFM 2002 clinical trial (www.clinicaltrials.gov, #NCT00114348) for somatic mutations activating the Ras pathway (KRAS, NRAS, FLT3, and PTPN11) and showed mutation to be highly prevalent (76 from 206). Clinically, they were associated with high-risk features including early relapse, central nervous system (CNS) involvement, and specifically for NRAS/KRAS mutations, chemoresistance. KRAS mutations were associated with a reduced overall survival. Mutation screening of the matched diagnostic samples found many to be wild type (WT); however, by using more sensitive allelic-specific assays, low-level mutated subpopulations were found in many cases, suggesting that they survived up-front therapy and subsequently emerged at relapse. Preclinical evaluation of the mitogen-activated protein kinase kinase 1/2 inhibitor selumetinib (AZD6244, ARRY-142886) showed significant differential sensitivity in Ras pathway-mutated ALL compared with WT cells both in vitro and in an orthotopic xenograft model engrafted with primary ALL; in the latter, reduced RAS-mutated CNS leukemia. Given these data, clinical evaluation of selumetinib may be warranted for Ras pathway-mutated relapsed ALL.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.