1
|
Scott S, Dillon R, Thiede C, Sadiq S, Cartwright A, Clouston HJ, Travis D, Mokretar K, Potter N, Chantry A, Whitby L. Assessment of acute myeloid leukemia molecular measurable residual disease testing in an interlaboratory study. Blood Adv 2023; 7:3686-3694. [PMID: 36939402 PMCID: PMC10368676 DOI: 10.1182/bloodadvances.2022009379] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/21/2023] Open
Abstract
The European LeukaemiaNet (ELN) measurable residual disease (MRD) working group has published consensus guidelines to standardize molecular genetic MRD testing of the t(8;21)(q22;q22.1) RUNX1::RUNX1T1, inv(16)(p13.1q22) CBFB::MYH11, t(15;17)(q24.1;q21.2) PML::RARA, and NPM1 type A markers. A study featuring 29 international laboratories was performed to assess interlaboratory variation in testing and the subsequent interpretation of results, both crucial to patient safety. Most participants in this study were able to detect, accurately quantify, and correctly interpret MRD testing results, with a level of proficiency expected from a clinical trial or standard-of-care setting. However, a few testing and interpretive errors were identified that, in a patient setting, would have led to misclassification of patient outcomes and inappropriate treatment pathways being followed. Of note, a high proportion of participants reported false-positive results in the NPM1 marker-negative sample. False-positive results may have clinical consequences, committing patients to unneeded additional chemotherapy and/or transplant with the attendant risk of morbidity and mortality, which therefore highlights the need for ongoing external quality assessment/proficiency testing in this area. Most errors identified in the study were related to the interpretation of results. It was noted that the ELN guidance lacks clarity for certain clinical scenarios and highlights the requirement for urgent revision of the guidelines to elucidate these issues and related educational efforts around the revisions to ensure effective dissemination.
Collapse
Affiliation(s)
- Stuart Scott
- Laboratory Medicine, UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Richard Dillon
- Department of Haematology, Guy’s International Centre of Excellence in Myeloid Disorders, Guy’s and St. Thomas NHS Foundation Trust, London, United Kingdom
- Department of Medical & Molecular Genetics, King’s College, London, United Kingdom
| | - Christian Thiede
- Department of Medicine, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- AgenDix, Applied Molecular Diagnostics GmbH, Dresden, Germany
| | - Sadia Sadiq
- Laboratory Medicine, UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Ashley Cartwright
- Laboratory Medicine, UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Hazel J. Clouston
- Laboratory Medicine, UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Debbie Travis
- Laboratory Medicine, UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Katya Mokretar
- Cancer genetics, Guy’s Hospital, South East Genomics Laboratory Hub, Synnovis, London, United Kingdom
| | - Nicola Potter
- Department of Medical & Molecular Genetics, King’s College, London, United Kingdom
| | - Andrew Chantry
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Liam Whitby
- Laboratory Medicine, UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
2
|
Hashimoto Y, Masunaga N, Kagara N, Abe K, Yoshinami T, Tsukabe M, Sota Y, Miyake T, Tanei T, Shimoda M, Shimazu K. Detection of Ultra-Rare ESR1 Mutations in Primary Breast Cancer Using LNA-Clamp ddPCR. Cancers (Basel) 2023; 15:cancers15092632. [PMID: 37174098 PMCID: PMC10177270 DOI: 10.3390/cancers15092632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
ESR1 mutations in breast cancer are one of the mechanisms of resistance to aromatase inhibitors. These mutations are common in metastatic breast cancer; however, these are rare in primary breast cancer. However, these data have been analyzed mainly in formalin-fixed, paraffin-embedded tissue; thus, rare mutations that may be present in primary breast cancer may be overlooked. In this study, we developed a highly sensitive mutation detection method called locked nucleic acid (LNA)-clamp droplet digital PCR (ddPCR) and validated it. The mutation detection sensitivity was substantiated to 0.003%. Then, we used this method to analyze ESR1 mutations in fresh-frozen (FF) tissues of primary breast cancer. cDNA extracted from the FF tissues of 212 patients with primary breast cancers were measured. Twenty-eight ESR1 mutations were found in twenty-seven (12.7%) patients. Sixteen (7.5%) patients had Y537S mutations and twelve (5.7%) had D538G mutations. Two mutations with a variant allele frequency (VAF) of ≥0.1% and twenty-six mutations with a VAF of <0.1% were found. By using this LNA-clamp ddPCR, this study demonstrated the presence of minor clones with a VAF of <0.1% in primary breast cancer.
Collapse
Affiliation(s)
- Yoko Hashimoto
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Naofumi Kagara
- Department of Breast Surgery, Osaka General Medical Center, 3-1-56, Bandai-Higashi, Sumiyoshi-ku, Osaka 558-8558, Osaka, Japan
| | - Kaori Abe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Masami Tsukabe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Tomohiro Miyake
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
3
|
Kunadt D, Stasik S, Metzeler KH, Röllig C, Schliemann C, Greif PA, Spiekermann K, Rothenberg-Thurley M, Krug U, Braess J, Krämer A, Hochhaus A, Scholl S, Hilgendorf I, Brümmendorf TH, Jost E, Steffen B, Bug G, Einsele H, Görlich D, Sauerland C, Schäfer-Eckart K, Krause SW, Hänel M, Hanoun M, Kaufmann M, Wörmann B, Kramer M, Sockel K, Egger-Heidrich K, Herold T, Ehninger G, Burchert A, Platzbecker U, Berdel WE, Müller-Tidow C, Hiddemann W, Serve H, Stelljes M, Baldus CD, Neubauer A, Schetelig J, Thiede C, Bornhäuser M, Middeke JM, Stölzel F. Impact of IDH1 and IDH2 mutational subgroups in AML patients after allogeneic stem cell transplantation. J Hematol Oncol 2022; 15:126. [PMID: 36064577 PMCID: PMC9442956 DOI: 10.1186/s13045-022-01339-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
Background The role of allogeneic hematopoietic cell transplantation (alloHCT) in acute myeloid leukemia (AML) with mutated IDH1/2 has not been defined. Therefore, we analyzed a large cohort of 3234 AML patients in first complete remission (CR1) undergoing alloHCT or conventional chemo-consolidation and investigated outcome in respect to IDH1/2 mutational subgroups (IDH1 R132C, R132H and IDH2 R140Q, R172K). Methods Genomic DNA was extracted from bone marrow or peripheral blood samples at diagnosis and analyzed for IDH mutations with denaturing high-performance liquid chromatography, Sanger sequencing and targeted myeloid panel next-generation sequencing, respectively. Statistical as-treated analyses were performed using R and standard statistical methods (Kruskal–Wallis test for continuous variables, Chi-square test for categorical variables, Cox regression for univariate and multivariable models), incorporating alloHCT as a time-dependent covariate. Results Among 3234 patients achieving CR1, 7.8% harbored IDH1 mutations (36% R132C and 47% R132H) and 10.9% carried IDH2 mutations (77% R140Q and 19% R172K). 852 patients underwent alloHCT in CR1. Within the alloHCT group, 6.2% had an IDH1 mutation (43.4% R132C and 41.4% R132H) and 10% were characterized by an IDH2 mutation (71.8% R140Q and 24.7% R172K). Variants IDH1 R132C and IDH2 R172K showed a significant benefit from alloHCT for OS (p = .017 and p = .049) and RFS (HR = 0.42, p = .048 and p = .009) compared with chemotherapy only. AlloHCT in IDH2 R140Q mutated AML resulted in longer RFS (HR = 0.4, p = .002). Conclusion In this large as-treated analysis, we showed that alloHCT is able to overcome the negative prognostic impact of certain IDH mutational subclasses in first-line consolidation treatment and could pending prognostic validation, provide prognostic value for AML risk stratification and therapeutic decision making. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01339-8.
Collapse
Affiliation(s)
- Desiree Kunadt
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany.
| | - Sebastian Stasik
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Klaus H Metzeler
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany.,Klinik und Poliklinik für Hämatologie, Zelltherapie und Hämostaseologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Christoph Röllig
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | | | - Philipp A Greif
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Karsten Spiekermann
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Maja Rothenberg-Thurley
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Utz Krug
- Medizinische Klinik III, Klinikum Leverkusen, Leverkusen, Germany
| | - Jan Braess
- Krankenhaus Barmherzige Brüder Regensburg, Regensburg, Germany
| | - Alwin Krämer
- Medizinische Klinik Und Poliklinik, Abteilung Innere Medizin V, Universität Heidelberg, Heidelberg, Germany
| | - Andreas Hochhaus
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Sebastian Scholl
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Inken Hilgendorf
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | | | - Edgar Jost
- Medizinische Klinik IV, Uniklinik RWTH Aachen, Aachen, Germany
| | - Björn Steffen
- Medizinische Klinik 2, Hämatologie/Onkologie, Goethe-Universität, Frankfurt am Main, Germany
| | - Gesine Bug
- Medizinische Klinik 2, Hämatologie/Onkologie, Goethe-Universität, Frankfurt am Main, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Dennis Görlich
- Institut für Biometrie und Klinische Forschung, Universität Münster, Münster, Germany
| | - Cristina Sauerland
- Institut für Biometrie und Klinische Forschung, Universität Münster, Münster, Germany
| | - Kerstin Schäfer-Eckart
- Klinik für Innere Medizin 5, Klinikum Nürnberg, Paracelsus Medizinische Privatuniversität, Nuremberg, Germany
| | - Stefan W Krause
- Medizinische Klinik 5, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mathias Hänel
- Medizinische Klinik III, Klinikum Chemnitz, Chemnitz, Germany
| | - Maher Hanoun
- Klinik für Hämatologie, Universitätsklinikum Essen, Essen, Germany
| | - Martin Kaufmann
- Abteilung für Hämatologie, Onkologie und Palliativmedizin, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Bernhard Wörmann
- Abteilung für Hämatologie, Onkologie und Palliativmedizin, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Michael Kramer
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Katja Sockel
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | | | - Tobias Herold
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Gerhard Ehninger
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Andreas Burchert
- Klinik für Innere Medizin, Schwerpunkt Hämatologie, Onkologie und Immunologie, Philipps Universität Marburg, Marburg, Germany
| | - Uwe Platzbecker
- Klinik und Poliklinik für Hämatologie, Zelltherapie und Hämostaseologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Wolfgang E Berdel
- Medizinische Klinik A, Universitätsklinikum Münster, Münster, Germany
| | - Carsten Müller-Tidow
- Medizinische Klinik Und Poliklinik, Abteilung Innere Medizin V, Universität Heidelberg, Heidelberg, Germany
| | - Wolfgang Hiddemann
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Hubert Serve
- Medizinische Klinik 2, Hämatologie/Onkologie, Goethe-Universität, Frankfurt am Main, Germany
| | - Matthias Stelljes
- Medizinische Klinik A, Universitätsklinikum Münster, Münster, Germany
| | - Claudia D Baldus
- Klinik für Innere Medizin II, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Andreas Neubauer
- Klinik für Innere Medizin, Schwerpunkt Hämatologie, Onkologie und Immunologie, Philipps Universität Marburg, Marburg, Germany
| | - Johannes Schetelig
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany.,DKMS Clinical Trials Unit, Dresden, Germany
| | - Christian Thiede
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Martin Bornhäuser
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany.,National Center for Tumor Diseases, Dresden (NCT/UCC), Dresden, Germany.,German Consortium for Translational Cancer Research (DKTK), DKFZ, Heidelberg, Germany
| | - Jan M Middeke
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Friedrich Stölzel
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | | |
Collapse
|
4
|
Differential impact of IDH1/2 mutational subclasses on outcome in adult AML: Results from a large multicenter study. Blood Adv 2021; 6:1394-1405. [PMID: 34794176 PMCID: PMC8905706 DOI: 10.1182/bloodadvances.2021004934] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 10/14/2021] [Indexed: 11/20/2022] Open
Abstract
Patients with IDH1-R132C have a lower complete remission rate and a trend toward reduced OS. Patients with IDH2-R172K in the European LeukemiaNet intermediate/adverse-risk group have significantly better relapse-free survival and OS.
Mutations of the isocitrate dehydrogenase-1 (IDH1) and IDH2 genes are among the most frequent alterations in acute myeloid leukemia (AML) and can be found in ∼20% of patients at diagnosis. Among 4930 patients (median age, 56 years; interquartile range, 45-66) with newly diagnosed, intensively treated AML, we identified IDH1 mutations in 423 (8.6%) and IDH2 mutations in 575 (11.7%). Overall, there were no differences in response rates or survival for patients with mutations in IDH1 or IDH2 compared with patients without mutated IDH1/2. However, distinct clinical and comutational phenotypes of the most common subtypes of IDH1/2 mutations could be associated with differences in outcome. IDH1-R132C was associated with increased age, lower white blood cell (WBC) count, less frequent comutation of NPM1 and FLT3 internal tandem mutation (ITD) as well as with lower rate of complete remission and a trend toward reduced overall survival (OS) compared with other IDH1 mutation variants and wild-type (WT) IDH1/2. In our analysis, IDH2-R172K was associated with significantly lower WBC count, more karyotype abnormalities, and less frequent comutations of NPM1 and/or FLT3-ITD. Among patients within the European LeukemiaNet 2017 intermediate- and adverse-risk groups, relapse-free survival and OS were significantly better for those with IDH2-R172K compared with WT IDH, providing evidence that AML with IDH2-R172K could be a distinct entity with a specific comutation pattern and favorable outcome. In summary, the presented data from a large cohort of patients with IDH1/2 mutated AML indicate novel and clinically relevant findings for the most common IDH mutation subtypes.
Collapse
|
5
|
Kapp-Schwoerer S, Weber D, Corbacioglu A, Gaidzik VI, Paschka P, Krönke J, Theis F, Rücker FG, Teleanu MV, Panina E, Jahn N, Herzig J, Kubanek L, Schrade A, Göhring G, Fiedler W, Kindler T, Schroeder T, Mayer KT, Lübbert M, Wattad M, Götze KS, Horst HA, Koller E, Wulf G, Schleicher J, Bentz M, Krauter J, Bullinger L, Krzykalla J, Benner A, Schlenk RF, Thol F, Heuser M, Ganser A, Döhner H, Döhner K. Impact of gemtuzumab ozogamicin on MRD and relapse risk in patients with NPM1-mutated AML: results from the AMLSG 09-09 trial. Blood 2020; 136:3041-3050. [PMID: 33367545 DOI: 10.1182/blood.2020005998] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/19/2020] [Indexed: 01/07/2023] Open
Abstract
Monitoring of measurable residual disease (MRD) provides prognostic information in patients with Nucleophosmin1-mutated (NPM1mut) acute myeloid leukemia (AML) and represents a powerful tool to evaluate treatment effects within clinical trials. We determined NPM1mut transcript levels (TLs) by quantitative reverse-transcription polymerase chain reaction and evaluated the prognostic impact of NPM1mut MRD and the effect of gemtuzumab ozogamicin (GO) on NPM1mut TLs and the cumulative incidence of relapse (CIR) in patients with NPM1mut AML enrolled in the randomized phase 3 AMLSG 09-09 trial. A total of 3733 bone marrow (BM) samples and 3793 peripheral blood (PB) samples from 469 patients were analyzed. NPM1mut TL log10 reduction ≥ 3 and achievement of MRD negativity in BM and PB were significantly associated with a lower CIR rate, after 2 treatment cycles and at end of treatment (EOT). In multivariate analyses, MRD positivity was consistently revealed to be a poor prognostic factor in BM and PB. With regard to treatment effect, the median NPM1mut TLs were significantly lower in the GO-Arm across all treatment cycles, resulting in a significantly greater proportion of patients achieving MRD negativity at EOT (56% vs 41%; P = .01). The better reduction in NPM1mut TLs after 2 treatment cycles in MRD positive patients by the addition of GO led to a significantly lower CIR rate (4-year CIR, 29.3% vs 45.7%, P = .009). In conclusion, the addition of GO to intensive chemotherapy in NPM1mut AML resulted in a significantly better reduction in NPM1mut TLs across all treatment cycles, leading to a significantly lower relapse rate.
Collapse
Affiliation(s)
| | - Daniela Weber
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Andrea Corbacioglu
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Verena I Gaidzik
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Peter Paschka
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Jan Krönke
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Frauke Theis
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | | | - Ekaterina Panina
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Nikolaus Jahn
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Julia Herzig
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Lena Kubanek
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Anika Schrade
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Gudrun Göhring
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Walter Fiedler
- Hubertus Wald University Cancer Center, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Kindler
- Department of Hematology, Medical Oncology, and Pneumology, University Cancer Center Mainz, Mainz, Germany
| | - Thomas Schroeder
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Karin T Mayer
- Department of Hematology and Oncology, University Hospital Bonn, Bonn, Germany
| | - Michael Lübbert
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mohammed Wattad
- Department of Hematology, Oncology and Stem Cell Transplantation, Kliniken Essen Süd, Essen, Germany
| | - Katharina S Götze
- III. Department of Medicine, Hematology, and Medical Oncology, Technical University of Munich, Munich, Germany
| | - Heinz A Horst
- Department of Internal Medicine II, University Hospital Schleswig Holstein, Campus Kiel, Kiel, Germany
| | - Elisabeth Koller
- III. Department of Medicine, Hematology, and Medical Oncology, Hanuschkrankenhaus Wien, Vienna, Austria
| | - Gerald Wulf
- Department of Hematology and Oncology, Georg-August-University Göttingen, Göttingen, Germany
| | - Jan Schleicher
- Department of Hematology and Oncology, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Martin Bentz
- Department of Internal Medicine III, Municipal Hospital of Karlsruhe, Karlsruhe, Germany
| | - Jürgen Krauter
- Department of Internal Medicine III, Municipal Hospital of Braunschweig, Braunschweig, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumorimmunology, Charité University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Julia Krzykalla
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Richard F Schlenk
- Nationales Centrum für Tumorerkrankungen Trial Center, National Center of Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany; and
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
6
|
Cocciardi S, Dolnik A, Kapp-Schwoerer S, Rücker FG, Lux S, Blätte TJ, Skambraks S, Krönke J, Heidel FH, Schnöder TM, Corbacioglu A, Gaidzik VI, Paschka P, Teleanu V, Göhring G, Thol F, Heuser M, Ganser A, Weber D, Sträng E, Kestler HA, Döhner H, Bullinger L, Döhner K. Clonal evolution patterns in acute myeloid leukemia with NPM1 mutation. Nat Commun 2019; 10:2031. [PMID: 31048683 PMCID: PMC6497712 DOI: 10.1038/s41467-019-09745-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 03/28/2019] [Indexed: 12/15/2022] Open
Abstract
Mutations in the nucleophosmin 1 (NPM1) gene are considered founder mutations in the pathogenesis of acute myeloid leukemia (AML). To characterize the genetic composition of NPM1 mutated (NPM1mut) AML, we assess mutation status of five recurrently mutated oncogenes in 129 paired NPM1mut samples obtained at diagnosis and relapse. We find a substantial shift in the genetic pattern from diagnosis to relapse including NPM1mut loss (n = 11). To better understand these NPM1mut loss cases, we perform whole exome sequencing (WES) and RNA-Seq. At the time of relapse, NPM1mut loss patients (pts) feature distinct mutational patterns that share almost no somatic mutation with the corresponding diagnosis sample and impact different signaling pathways. In contrast, profiles of pts with persistent NPM1mut are reflected by a high overlap of mutations between diagnosis and relapse. Our findings confirm that relapse often originates from persistent leukemic clones, though NPM1mut loss cases suggest a second "de novo" or treatment-associated AML (tAML) as alternative cause of relapse.
Collapse
Affiliation(s)
- Sibylle Cocciardi
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Anna Dolnik
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Silke Kapp-Schwoerer
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Susanne Lux
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Tamara J Blätte
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Sabrina Skambraks
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Jan Krönke
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Florian H Heidel
- Department of Internal Medicine II, Hematology and Oncology, Friedrich-Schiller-University Medical Center, Jena, 07743, Germany.,Leibniz-Institute on Aging, Fritz-Lipmann-Institute, Jena, 07745, Germany
| | - Tina M Schnöder
- Department of Internal Medicine II, Hematology and Oncology, Friedrich-Schiller-University Medical Center, Jena, 07743, Germany.,Leibniz-Institute on Aging, Fritz-Lipmann-Institute, Jena, 07745, Germany
| | - Andrea Corbacioglu
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Verena I Gaidzik
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Peter Paschka
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Veronica Teleanu
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Gudrun Göhring
- Institute of Cell & Molecular Pathology, Hannover Medical School, Hannover, 30625, Germany
| | - Felicitas Thol
- Department of Haematology, Haemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, 30625, Germany
| | - Michael Heuser
- Department of Haematology, Haemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, 30625, Germany
| | - Arnold Ganser
- Department of Haematology, Haemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, 30625, Germany
| | - Daniela Weber
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Eric Sträng
- Institute of Medical Systems Biology, Ulm University, Ulm, 30625, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, 30625, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany
| | - Lars Bullinger
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany. .,Department of Hematology, Oncology and Tumorimmunology, Charité University Medicine, Berlin, 13353, Germany.
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, 89081, Germany.
| |
Collapse
|
7
|
[Diagnosis and treatment of acute myeloid leukemia : The updated 2018 Onkopedia Guideline]. Internist (Berl) 2019; 60:257-272. [PMID: 30767029 DOI: 10.1007/s00108-019-0562-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In April 2018, an updated version of the previously published guidelines on acute myeloid leukemia (AML) from 2010 and 2017 was released. A revision was necessary because of two positive aspects: First, new data and insights on risk stratification and monitoring, and second, the clinical development and approval of new agents. The modified genetic risk classification allows a more precise distinction of different diagnostic groups and consequently a better matched post-remission treatment. The availability of new targeted drugs such as inhibitors turns genetic analyses from a mere prognostic tool into an instrument for treatment decisions. Several recently approved agents expand the treatment options for AML and raise hope for an improved prognosis and cure in the future.
Collapse
|
8
|
Gebhard C, Glatz D, Schwarzfischer L, Wimmer J, Stasik S, Nuetzel M, Heudobler D, Andreesen R, Ehninger G, Thiede C, Rehli M. Profiling of aberrant DNA methylation in acute myeloid leukemia reveals subclasses of CG-rich regions with epigenetic or genetic association. Leukemia 2018; 33:26-36. [PMID: 29925905 DOI: 10.1038/s41375-018-0165-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/19/2018] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
Malignant transformation is frequently associated with disease-specific epigenetic alterations, but the underlying mechanisms and pathophysiological consequences remain poorly understood. Here, we used global comparative DNA methylation profiling at CG-rich regions of 27 acute myeloid leukemia (AML) samples to select a subset of aberrantly methylated CG-rich regions (~400 regions, ~15,000 CpGs) for quantitative DNA methylation profiling in a large cohort of AML patients (n = 196) using MALDI-TOF analysis of bisulfite-treated DNA. Meta-analysis separated a subgroup of CG-rich regions showing highly correlated DNA methylation changes that were marked by histone H3 lysine 27 trimethylation in normal hematopoietic progenitor cells. While the group of non-polycomb group (PcG) target regions displayed methylation patterns that correlated well with molecular and cytogenetic markers, PcG target regions displayed a much weaker association with genetic features. However, the degree of methylation gain across the latter panel showed significant correlation with active DNMT3A levels and with overall survival. Our study suggests that both epigenetic as well as genetic aberrations underlay AML-related changes in DNA methylation at CG-rich regions and that the former may provide a marker to improve classification and prognostication of adult AML patients.
Collapse
Affiliation(s)
- Claudia Gebhard
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany.,RCI Regensburg Centre for Interventional Immunology, University Hospital Regensburg, 93042, Regensburg, Germany
| | - Dagmar Glatz
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany.,RCI Regensburg Centre for Interventional Immunology, University Hospital Regensburg, 93042, Regensburg, Germany
| | - Lucia Schwarzfischer
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany
| | - Julia Wimmer
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany
| | - Sebastian Stasik
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, 01307, Dresden, Germany
| | - Margit Nuetzel
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany
| | - Reinhard Andreesen
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany.,RCI Regensburg Centre for Interventional Immunology, University Hospital Regensburg, 93042, Regensburg, Germany
| | - Gerhard Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, 01307, Dresden, Germany
| | - Christian Thiede
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, 01307, Dresden, Germany
| | - Michael Rehli
- Department of Internal Medicine III, University Hospital Regensburg, 93042, Regensburg, Germany. .,RCI Regensburg Centre for Interventional Immunology, University Hospital Regensburg, 93042, Regensburg, Germany.
| |
Collapse
|
9
|
Pettersson L, Levéen P, Axler O, Dvorakova D, Juliusson G, Ehinger M. Improved minimal residual disease detection by targeted quantitative polymerase chain reaction inNucleophosmin 1type a mutated acute myeloid leukemia. Genes Chromosomes Cancer 2016; 55:750-66. [DOI: 10.1002/gcc.22375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/25/2022] Open
Affiliation(s)
| | - Per Levéen
- Department of Pathology; University and Regional Laboratories, Skåne University Hospital; Lund Sweden
| | - Olof Axler
- Department of Pathology; University and Regional Laboratories, Skåne University Hospital; Lund Sweden
| | - Dana Dvorakova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy, Masaryk University and University Hospital Brno; Brno Czech Republic
| | - Gunnar Juliusson
- Department of Hematology; Skåne University Hospital; Lund Sweden
| | - Mats Ehinger
- Department of Pathology; University and Regional Laboratories, Skåne University Hospital; Lund Sweden
| |
Collapse
|
10
|
Olsson L, Zettermark S, Biloglav A, Castor A, Behrendtz M, Forestier E, Paulsson K, Johansson B. The genetic landscape of paediatric de novo acute myeloid leukaemia as defined by single nucleotide polymorphism array and exon sequencing of 100 candidate genes. Br J Haematol 2016; 174:292-301. [PMID: 27022003 DOI: 10.1111/bjh.14056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/22/2016] [Indexed: 01/07/2023]
Abstract
Cytogenetic analyses of a consecutive series of 67 paediatric (median age 8 years; range 0-17) de novo acute myeloid leukaemia (AML) patients revealed aberrations in 55 (82%) cases. The most common subgroups were KMT2A rearrangement (29%), normal karyotype (15%), RUNX1-RUNX1T1 (10%), deletions of 5q, 7q and/or 17p (9%), myeloid leukaemia associated with Down syndrome (7%), PML-RARA (7%) and CBFB-MYH11 (5%). Single nucleotide polymorphism array (SNP-A) analysis and exon sequencing of 100 genes, performed in 52 and 40 cases, respectively (39 overlapping), revealed ≥1 aberration in 89%; when adding cytogenetic data, this frequency increased to 98%. Uniparental isodisomies (UPIDs) were detected in 13% and copy number aberrations (CNAs) in 63% (median 2/case); three UPIDs and 22 CNAs were recurrent. Twenty-two genes were targeted by focal CNAs, including AEBP2 and PHF6 deletions and genes involved in AML-associated gene fusions. Deep sequencing identified mutations in 65% of cases (median 1/case). In total, 60 mutations were found in 30 genes, primarily those encoding signalling proteins (47%), transcription factors (25%), or epigenetic modifiers (13%). Twelve genes (BCOR, CEBPA, FLT3, GATA1, KIT, KRAS, NOTCH1, NPM1, NRAS, PTPN11, SMC3 and TP53) were recurrently mutated. We conclude that SNP-A and deep sequencing analyses complement the cytogenetic diagnosis of paediatric AML.
Collapse
Affiliation(s)
- Linda Olsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden.,Department of Clinical Genetics, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden
| | - Sofia Zettermark
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Andrea Biloglav
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anders Castor
- Department of Paediatrics, Skåne University Hospital, Lund, Sweden
| | - Mikael Behrendtz
- Department of Paediatrics, Linköping University Hospital, Linköping, Sweden
| | - Erik Forestier
- Department of Medical Bioscience, University of Umeå, Umeå, Sweden
| | - Kajsa Paulsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Bertil Johansson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden.,Department of Clinical Genetics, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden
| |
Collapse
|
11
|
Kawaguchi-Ihara N, Itoh M, Murohashi I, Tohda S. Establishment of a quenching probe method for detection of NPM1 mutations in acute myeloid leukemia cells. Oncol Lett 2016; 11:2429-2432. [PMID: 27073492 DOI: 10.3892/ol.2016.4225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 01/26/2016] [Indexed: 12/31/2022] Open
Abstract
Nucleophosmin (NPM1) mutations, generally consisting of a four base-pair insertion, are present in ~60% of all cytogenetically normal acute myeloid leukemia (AML) cases. The mutation is clinically significant as an important prognostic factor. Direct sequencing is the current standard method of mutation detection, however, it is quite costly and time consuming. The present study aimed to establish a highly sensitive quenching probe (QP) method to detect NPM1 mutations efficiently. Melting curve analysis was performed using a QP, following polymerase chain reaction for amplification of the involved region of the gene. The curve derived from the fluorescent intensity with respect to the temperature of OCI/AML3, a heterozygous NPM1 mutant AML cell line, was W-shaped with melting peaks at 61°C and 68°C. That of M-07e, the homozygous wild type cell line, was V-shaped with a melting peak at 68°C. Thus, the curve derived from the mutant allele was easily discriminated from that of the wild-type allele. The mutant allele was detected in concentrations as low as 3% as determined by a subsequent sensitivity study. With a short testing time and a high sensitivity, this assay was applicable for NPM1-mutated AML patient samples and is appropriate for screening NPM1 mutations. It does require further examination as to whether it would be useful as a detection method for other mutant alleles since NPM1 mutations may consist of 61 known types of mutant sequences. To the best of our knowledge, this is the first report describing the QP method for the detection of NPM1 mutations.
Collapse
Affiliation(s)
- Noriko Kawaguchi-Ihara
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan; Department of Health Sciences, Saitama Prefectural University, Koshigaya, Saitama 343-8540, Japan
| | - Mai Itoh
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Ikuo Murohashi
- Department of Health Sciences, Saitama Prefectural University, Koshigaya, Saitama 343-8540, Japan
| | - Shuji Tohda
- Department of Laboratory Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| |
Collapse
|
12
|
Ziai JM, Siddon AJ. Pathology Consultation on Gene Mutations in Acute Myeloid Leukemia. Am J Clin Pathol 2015; 144:539-54. [PMID: 26386075 DOI: 10.1309/ajcp77zfpuqgygwy] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES Acute myeloid leukemia (AML) is a rapidly fatal disease without the use of aggressive chemotherapy regimens. Cytogenetic and molecular studies are commonly used to classify types of AML based on prognosis, as well as to determine therapeutic regimens. METHODS Although there are several AML classifications determined by particular translocations, cytogenetically normal AML represents a molecularly, as well as clinically, heterogeneous group of diseases. Laboratory evaluation of AML will become increasingly important as new mutations with both prognostic and therapeutic implications are being recognized. Moreover, because many patients with AML are being treated more effectively, these mutations may become increasingly useful as markers of minimal residual disease, which can be interpreted in an individualized approach. RESULTS Current laboratory studies of gene mutations in AML include analysis of NPM1, FLT3, CEBPA, and KIT. In addition to these genes, many other genes are emerging as potentially useful in determining patients' prognosis, therapy, and disease course. CONCLUSIONS This article briefly reviews the current most clinically relevant gene mutations and their clinical and immunophenotypic features, prognostic information, and methods used for detection.
Collapse
Affiliation(s)
| | - Alexa J. Siddon
- Departments of Pathology, Yale School of Medicine, New Haven, CT
- Laboratory Medicine, Yale School of Medicine, New Haven, CT
- VA Connecticut Healthcare, West Haven, CT
| | | |
Collapse
|
13
|
Artificial Nucleic Acid Probes and Their Applications in Clinical Microbiology. METHODS IN MICROBIOLOGY 2015. [DOI: 10.1016/bs.mim.2015.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
14
|
The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML. Blood 2013; 122:83-92. [DOI: 10.1182/blood-2012-10-461749] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Key Points
NPM1 RT-PCR levels >1% are associated with poor overall and disease-free survival in AML patients treated with chemotherapy. NPM1 MRD levels >10% are associated with poor overall and disease-free survival in AML patients after allogeneic transplantation.
Collapse
|
15
|
von Bonin M, Wermke M, Cosgun KN, Thiede C, Bornhauser M, Wagemaker G, Waskow C. In vivo expansion of co-transplanted T cells impacts on tumor re-initiating activity of human acute myeloid leukemia in NSG mice. PLoS One 2013; 8:e60680. [PMID: 23585844 PMCID: PMC3621959 DOI: 10.1371/journal.pone.0060680] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/01/2013] [Indexed: 01/19/2023] Open
Abstract
Human cells from acute myeloid leukemia (AML) patients are frequently transplanted into immune-compromised mouse strains to provide an in vivo environment for studies on the biology of the disease. Since frequencies of leukemia re-initiating cells are low and a unique cell surface phenotype that includes all tumor re-initiating activity remains unknown, the underlying mechanisms leading to limitations in the xenotransplantation assay need to be understood and overcome to obtain robust engraftment of AML-containing samples. We report here that in the NSG xenotransplantation assay, the large majority of mononucleated cells from patients with AML fail to establish a reproducible myeloid engraftment despite high donor chimerism. Instead, donor-derived cells mainly consist of polyclonal disease-unrelated expanded co-transplanted human T lymphocytes that induce xenogeneic graft versus host disease and mask the engraftment of human AML in mice. Engraftment of mainly myeloid cell types can be enforced by the prevention of T cell expansion through the depletion of lymphocytes from the graft prior transplantation.
Collapse
Affiliation(s)
- Malte von Bonin
- Regeneration in Hematopoiesis, Center for Regenerative Therapies Dresden – CRTD, DFG Research Center and Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Wermke
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Kadriye Nehir Cosgun
- Regeneration in Hematopoiesis, Center for Regenerative Therapies Dresden – CRTD, DFG Research Center and Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
| | - Christian Thiede
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Bornhauser
- Medizinische Klinik und Poliklinik 1, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Gerard Wagemaker
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Claudia Waskow
- Regeneration in Hematopoiesis, Center for Regenerative Therapies Dresden – CRTD, DFG Research Center and Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
16
|
New Quantitative Method to Identify NPM1 Mutations in Acute Myeloid Leukaemia. LEUKEMIA RESEARCH AND TREATMENT 2013; 2013:756703. [PMID: 23691328 PMCID: PMC3649360 DOI: 10.1155/2013/756703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/18/2013] [Indexed: 12/31/2022]
Abstract
Somatic mutations in the NPM1 gene, which encodes for nucleophosmin, have been reported to be the most frequent genetic abnormalities found in acute myeloid leukaemia (AML). Their identification and quantification remain crucial for the patients' residual disease monitoring. We investigated a new method that could represent a novel reliable alternative to sequencing for its identification. This method was based on high-resolution melting analysis in order to detect mutated patients and on an allele-specific oligonucleotide real-time quantitative polymerase chain reaction (ASO-RQ-PCR) for the identification and quantification of the transcripts carrying NPM1 mutations (NPM1m). Few patients carrying known NPM1m enabled us to set up a table with the different primers' ΔCT values, identifying a profile for each mutation type. We then analysed a series of 337 AML patients' samples for NPM1 mutational status characterization and confirmed the ASO-RQ-PCR results by direct sequencing. We identified some mutations in 86 samples, and the results were fully correlated in 100% of the 36 sequenced samples. We also detected other rare NPM1m in two samples, that we confirmed by direct sequencing. This highly specific method provides a novel quick, useful, and costless tool, easy to use in routine practice.
Collapse
|
17
|
Asari M, Oka K, Omura T, Maseda C, Tasaki Y, Shiono H, Matsubara K, Matsuda M, Shimizu K. Universal fluorescent labeling of amplification products using locked nucleic acids. Electrophoresis 2013; 34:448-55. [PMID: 23161600 DOI: 10.1002/elps.201200215] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 09/16/2012] [Accepted: 09/25/2012] [Indexed: 12/15/2022]
Abstract
Amplification/hybridization-based genetic analyses using primers containing locked nucleic acids (LNAs) present many benefits. Here, we developed a novel design for universal fluorescent PCR using LNAs. Universal fluorescent PCR generates intermediate nonlabeled fragments and final fluorescent fragments in a two-step amplification process that uses locus-specific primers with universal tails and universal fluorescent primers. In this study, a few standard nucleotides were replaced with LNAs only in the fluorescent universal primers. The sequence of the fluorescent universal primer significantly affected the amplification efficiency. For primers with three LNAs, the fluorescent primers with stable M13(-47) sequences provided the most efficient signal (approximately tenfold higher than the primers with M13(-21) sequences at lower Tm values). Moreover, AT-rich LNA substitutions in the fluorescent primers produced much lower amplification efficiencies than GC-rich substitutions. GC-rich LNAs produced greater differences in Tm values among primers, and resulted in the preferential production of fluorescently labeled amplicons. The specificity and sensitivity of LNA-containing fluorescent primers were assessed by genotyping eight STRs in Japanese individuals, and full STR profiles could be generated using as little as 0.25 ng of genomic DNA. The method permitted clear discrimination of alleles and represents sensitive STR genotyping at a reduced cost.
Collapse
Affiliation(s)
- Masaru Asari
- Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
The increasing sensitivity of PCR has meant that in the last two decades PCR has emerged as a major tool in diet studies, enabling us to refine our understanding of trophic links and to elucidate the diets of predators whose prey is as yet uncharacterized. The achievements and methods of PCR-based diet studies have been reviewed several times, but here we review an important development in the field: the use of PCR enrichment techniques to promote the amplification of prey DNA over that of the predator. We first discuss the success of using group-specific primers either in parallel single reactions or in multiplex reactions. We then concentrate on the more recent use of PCR enrichment techniques such as restriction enzyme digests, peptide nucleic acid clamping, DNA blocking and laser capture microdissection. We also survey the vast literature on enrichment techniques in clinical biology, to ascertain the pitfalls of enrichment techniques and what refinements have yielded some highly sensitive methods. We find that while there are several new approaches to enrichment, peptide nucleic acid clamping and DNA blocking are generally sufficient techniques for the characterization of diets of predators and highlight the most important considerations of the approach.
Collapse
Affiliation(s)
- R O'Rorke
- Leigh Marine Laboratory, University of Auckland, Warkworth, Northland 0941, New Zealand.
| | | | | |
Collapse
|
19
|
Matsuda K, Sugano M, Honda T. PCR for monitoring of minimal residual disease in hematologic malignancy. Clin Chim Acta 2011; 413:74-80. [PMID: 22057038 DOI: 10.1016/j.cca.2011.10.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/18/2011] [Accepted: 10/19/2011] [Indexed: 12/23/2022]
Abstract
Monitoring minimal residual disease (MRD) is useful to evaluate therapeutic response and risk of relapse in patients with hematologic malignancy. Currently available quantitative MRD assays are fluorescence in situ hybridization of chromosomal aberrations; multiparameter flow cytometry of leukemia-associated immunophenotypes; and quantitative polymerase chain reaction (qPCR) analysis of fusion genes, immunoglobulin/T-cell receptor gene rearrangements, genetic alterations, or over-expressed genes. Among the PCR-based markers, genetic alterations are found in acute myelogenous leukemia patients with cytogenetically normal karyotype and can be considered as applicable targets for monitoring of MRD. Screening, confirmation and quantification procedures are important to develop the patient- or tumor-specific MRD assays using the PCR-based markers. Wild-type blocking PCR or coamplification at lower denaturing temperature-PCR is suited for screening of low-abundant genetic alterations, and allele-specific qPCR using primers including mismatched base and locked nucleic acids can quantify not only insertion and duplication of several nucleotides but also single nucleotide mutation in the presence of an excess amount of wild-type nucleotides. In addition to the well-established MRD markers, such as immunoglobulin/T-cell receptor gene rearrangements and fusion genes, utilizing potential MRD markers such as genetic alterations may expand the spectrum of patients in whom MRD can be monitored.
Collapse
Affiliation(s)
- Kazuyuki Matsuda
- Department of Laboratory Medicine, Shinshu University Hospital, Asahi, Matsumoto, Japan.
| | | | | |
Collapse
|
20
|
Krönke J, Schlenk RF, Jensen KO, Tschürtz F, Corbacioglu A, Gaidzik VI, Paschka P, Onken S, Eiwen K, Habdank M, Späth D, Lübbert M, Wattad M, Kindler T, Salih HR, Held G, Nachbaur D, von Lilienfeld-Toal M, Germing U, Haase D, Mergenthaler HG, Krauter J, Ganser A, Göhring G, Schlegelberger B, Döhner H, Döhner K. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a study from the German-Austrian acute myeloid leukemia study group. J Clin Oncol 2011; 29:2709-16. [PMID: 21555683 DOI: 10.1200/jco.2011.35.0371] [Citation(s) in RCA: 298] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To evaluate the prognostic value of minimal residual disease (MRD) in patients with acute myeloid leukemia (AML) with NPM1 mutation (NPM1(mut)). PATIENTS AND METHOD RNA-based real-time quantitative polymerase chain reaction (RQ-PCR) specific for the detection of six different NPM1(mut) types was applied to 1,682 samples (bone marrow, n = 1,272; blood, n = 410) serially obtained from 245 intensively treated younger adult patients who were 16 to 60 years old. RESULTS NPM1(mut) transcript levels as a continuous variable were significantly associated with prognosis after each treatment cycle. Achievement of RQ-PCR negativity after double induction therapy identified patients with a low cumulative incidence of relapse (CIR; 6.5% after 4 years) compared with RQ-PCR-positive patients (53.0%; P < .001); this translated into significant differences in overall survival (90% v 51%, respectively; P = .001). After completion of therapy, CIR was 15.7% in RQ-PCR-negative patients compared with 66.5% in RQ-PCR-positive patients (P < .001). Multivariable analyses after double induction and after completion of consolidation therapy revealed higher NPM1(mut) transcript levels as a significant factor for a higher risk of relapse and death. Serial post-treatment assessment of MRD allowed early detection of relapse in patients exceeding more than 200 NPM1(mut)/10(4) ABL copies. CONCLUSION We defined clinically relevant time points for NPM1(mut) MRD assessment that allow for the identification of patients with AML who are at high risk of relapse. Monitoring of NPM1(mut) transcript levels should be incorporated in future clinical trials to guide therapeutic decisions.
Collapse
Affiliation(s)
- Jan Krönke
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Acute myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity? Blood 2010; 117:1109-20. [PMID: 21030560 DOI: 10.1182/blood-2010-08-299990] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
After the discovery of NPM1-mutated acute myeloid leukemia (AML) in 2005 and its subsequent inclusion as a provisional entity in the 2008 World Health Organization classification of myeloid neoplasms, several controversial issues remained to be clarified. It was unclear whether the NPM1 mutation was a primary genetic lesion and whether additional chromosomal aberrations and multilineage dysplasia had any impact on the biologic and prognostic features of NPM1-mutated AML. Moreover, it was uncertain how to classify AML patients who were double-mutated for NPM1 and CEBPA. Recent studies have shown that: (1) the NPM1 mutant perturbs hemopoiesis in experimental models; (2) leukemic stem cells from NPM1-mutated AML patients carry the mutation; and (3) the NPM1 mutation is usually mutually exclusive of biallelic CEPBA mutations. Moreover, the biologic and clinical features of NPM1-mutated AML do not seem to be significantly influenced by concomitant chromosomal aberrations or multilineage dysplasia. Altogether, these pieces of evidence point to NPM1-mutated AML as a founder genetic event that defines a distinct leukemia entity accounting for approximately one-third of all AML.
Collapse
|
22
|
Hafez M, Ye F, Jackson K, Yang Z, Karp JE, Labourier E, Gocke CD. Performance and clinical evaluation of a sensitive multiplex assay for the rapid detection of common NPM1 mutations. J Mol Diagn 2010; 12:629-35. [PMID: 20616361 DOI: 10.2353/jmoldx.2010.090219] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Determination of NPM1 mutation status has become essential for the molecular classification of acute myeloid leukemias (AML). Methods with high clinical sensitivity and specificity adapted to the molecular laboratory workflow are required for the diagnosis, prognosis, and monitoring of AML with normal karyotype. We report here the development and evaluation of a novel, streamlined, RNA-based assay for the rapid multiplex detection of common NPM1 mutations in a 96-well assay format. Using synthetic transcripts and total RNA from leukemic cell lines, we show that the assay can specifically detect NPM1 wild-type and mutants A, B, D, or J transcripts in the same reaction. Dilution experiments indicate an assay dynamic range >4 log units with an analytical sensitivity of approximately 0.01%. Evaluation of 69 clinical specimens at initial diagnosis resulted in 100% agreement with reference methods. Of patients with AML with normal karyotype, 53% carried one of four different mutations. The assay was also combined with other laboratory-developed tests to simultaneously detect NPM1 mutant transcripts and fusion transcripts resulting from t(8;21) or inv(16) in a single reaction well. Overall, these results show that the assay is a versatile and specific tool for the screening of NPM1 mutations in patients with AML. Its high analytical sensitivity further suggests potential utility for the monitoring of residual disease in AML with normal karyotype.
Collapse
Affiliation(s)
- Michael Hafez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Falini B. Acute myeloid leukemia with mutated nucleophosmin (NPM1): molecular, pathological, and clinical features. Cancer Treat Res 2010; 145:149-168. [PMID: 20306250 DOI: 10.1007/978-0-387-69259-3_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The NPM1 gene encodes for nucleophosmin, a nucleolus-located shuttling protein that is involved in multiple cell functions, including regulation of ribosome biogenesis, control of centrosome duplication and preservation of ARF tumor suppressor integrity. The NPM1 gene is specifically mutated in about 30% acute myeloid leukemia (AML) but not in other human neoplasms. Mutations cause crucial changes at the C-terminus of the NPM1 protein that are responsible for the aberrant nuclear export and accumulation of NPM1 mutants in the cytoplasm of leukemic cells. Diagnosis of AML with mutated NPM1 can be done using molecular techniques, immunohistochemistry (looking at cytoplasmic dislocation of nucleophosmin that is predictive of NPM1 mutations) and Western blotting with antibodies specifically directed against NPM1 mutants. Because of its distinctive molecular, pathological, immunophenotypic and prognostic features, AML with mutated NPM1 (synonym: NPMc+ AML) has been included, as a new provisional entity, in the 2008 World Health Organization (WHO) classification of myeloid neoplasms.
Collapse
|
24
|
Fuster Ó, Barragán E, Bolufer P, Cervera J, Larráyoz MJ, Jiménez-Velasco A, Martínez-López J, Valencia A, Moscardó F, Sanz MÁ. Rapid detection of KIT mutations in core-binding factor acute myeloid leukemia using high-resolution melting analysis. J Mol Diagn 2009; 11:458-63. [PMID: 19644024 PMCID: PMC2729844 DOI: 10.2353/jmoldx.2009.090043] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2009] [Indexed: 12/11/2022] Open
Abstract
The most frequent KIT mutations reported in core-binding factor acute myeloid leukemia are point mutations and insertions/deletions in exons 17 and 8. The vast majority of KIT mutation detection procedures are time-consuming, costly, or with a high lower limit of detection. High-resolution melting (HRM) is a gene scanning method that combines simplicity and rapid identification of genetic variants. We describe an HRM method for the simultaneous screening of exons 8 and 17 KIT mutations and report the results obtained in 69 core-binding factor acute myeloid leukemia patients. Mutation detection was compared with sequencing as the gold standard. The HRM method used high-resolution melting master reagents (Roche) and the LightCycler 480 (Roche) platform. HRM was reproducible, showed a lower limit of detection of 1%, and discriminated all patients with mutated KIT from controls without false positive or false negative results. Additionally, most of the mutations were differentiated from the other mutations. KIT mutations were present in 15.9% of patients, showing a higher incidence in inv(16) (25.8%) than in t(8;21) (7.9%). The presence of a KIT mutation was associated with a high white blood cell count, and adult patients with an exon 17 mutation had a higher incidence of relapse. These findings verify that HRM is a reliable, rapid, and sensitive method for KIT mutation screening. Furthermore, our study corroborates the unfavorable prognosis associated with exon 17 KIT mutations.
Collapse
Affiliation(s)
- Óscar Fuster
- Laboratory of Molecular Biology, Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | - Eva Barragán
- Laboratory of Molecular Biology, Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | - Pascual Bolufer
- Laboratory of Molecular Biology, Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | - José Cervera
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | | - Ana Valencia
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain
| | - Federico Moscardó
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain
| | - Miguel Ángel Sanz
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain
| |
Collapse
|
25
|
DR negativity is a distinctive feature of M1/M2 AML cases with NPM1 mutation. Leuk Res 2008; 32:1141-3. [DOI: 10.1016/j.leukres.2007.11.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 11/12/2007] [Accepted: 11/13/2007] [Indexed: 11/20/2022]
|
26
|
Laughlin TS, Becker MW, Liesveld JL, Mulford DA, Abboud CN, Brown P, Rothberg PG. Rapid method for detection of mutations in the nucleophosmin gene in acute myeloid leukemia. J Mol Diagn 2008; 10:338-45. [PMID: 18556765 DOI: 10.2353/jmoldx.2008.070175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mutations in exon 12 of the nucleophosmin gene (NPM1) that cause the encoded protein to abnormally relocate to the cytoplasm are found at diagnosis in about 50% of karyotypically normal acute myeloid leukemias and are associated with a more favorable outcome. We have devised a PCR-based assay for NPM1 exon 12 mutations using differential melting of an oligo probe labeled with a fluorescent dye. The nucleobase quenching (NBQ) phenomenon was used to detect probe hybridization, and an oligonucleotide containing locked nucleic acid (LNA) nucleotides was used as a PCR clamp to suppress amplification of the normal sequence and enhance the analytical sensitivity of the assay. After the NBQ assay, the specimens with a mutation were removed from the capillary and sequenced to identify the mutation. The use of the LNA clamp facilitates interpretation of the mutant sequence because of the lower intensity of the overlapping normal sequence. Analysis of a series of 70 patient specimens revealed 17 positive for an NPM1 mutation and 53 negatives. All of the NBQ results (positives and negatives) were confirmed with other methods. The analytical sensitivity of the NBQ assay is variable depending on the concentration of the PCR clamp and other parameters. Using a 100 nmol/L concentration of the LNA clamp, NPM1 mutations were detectable in a 10-fold excess of wild-type DNA. This assay may be valuable for screening disease specimens.
Collapse
Affiliation(s)
- Todd S Laughlin
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Wertheim G, Bagg A. Nucleophosmin (NPM1) mutations in acute myeloid leukemia: an ongoing (cytoplasmic) tale of dueling mutations and duality of molecular genetic testing methodologies. J Mol Diagn 2008; 10:198-202. [PMID: 18403611 DOI: 10.2353/jmoldx.2008.080019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Gerald Wertheim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-4283, USA
| | | |
Collapse
|
28
|
Ottone T, Ammatuna E, Lavorgna S, Noguera NI, Buccisano F, Venditti A, Giannì L, Postorino M, Federici G, Amadori S, Lo-Coco F. An allele-specific rt-PCR assay to detect type A mutation of the nucleophosmin-1 gene in acute myeloid leukemia. J Mol Diagn 2008; 10:212-6. [PMID: 18403613 DOI: 10.2353/jmoldx.2008.070166] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nucleophosmin-1 (NPM1) mutations represent the most frequent gene alteration in acute myeloid leukemia (AML). The most common NPM1 mutation type, accounting for 75 to 80% of cases, is referred to as mutation A (NPM1-mutA). These NPM1 alterations have been shown to possess prognostic significance because they appear to identify patients who will benefit from chemotherapy. Given the high prevalence and stability of these mutations over the course of disease, NPM1 mutations may serve as ideal targets for minimal residual disease (MRD) assessment in AML. Current detection methods are costly, require sophisticated equipment, and are often not sufficiently sensitive. We report here an allele-specific (ASO)-RT-PCR assay that enables rapid and sensitive detection of NPM1-mutA. A semi-nested ASO-PCR method was also designed to increase the sensitivity of our assay for the monitoring of MRD. We analyzed bone marrow cells collected from 52 patients with AML at presentation. NPM1-mutA was detected in leukemic cells from 21 patients. Assay specificity was confirmed by capillary electrophoresis and DNA sequencing. ASO-RT-PCR and semi-nested ASO-PCR assays could detect NPM1-mutA with sensitivities of 10(-2) and 10(-5), respectively. Results obtained here verify that our ASO-RT-PCR assay is a specific and sensitive method for the routine screening of NPM1-mutA, as well as for MRD monitoring of AML patients with this alteration. This method is convenient and easily applicable in countries with limited resources and no access to real-time quantitative PCR-based technologies.
Collapse
Affiliation(s)
- Tiziana Ottone
- Dipartimento di Biopatologia e Diagnostica per Immagini, University Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mrózek K, Döhner H, Bloomfield CD. Influence of new molecular prognostic markers in patients with karyotypically normal acute myeloid leukemia: recent advances. Curr Opin Hematol 2007; 14:106-14. [PMID: 17255787 DOI: 10.1097/moh.0b013e32801684c7] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Molecular study of cytogenetically normal acute myeloid leukemia is among the most active areas of leukemia research. Despite having the same normal karyotype, adults with de-novo cytogenetically normal acute myeloid leukemia who constitute the largest cytogenetic group of acute myeloid leukemia, are very diverse with respect to acquired gene mutations and gene expression changes. These genetic alterations affect clinical outcome and may assist in selection of proper treatment. Herein we critically summarize recent clinically relevant molecular genetic studies of cytogenetically normal acute myeloid leukemia. RECENT FINDINGS NPM1 gene mutations causing aberrant cytoplasmic localization of nucleophosmin have been demonstrated to be the most frequent submicroscopic alterations in cytogenetically normal acute myeloid leukemia and to confer improved prognosis, especially in patients without a concomitant FLT3 gene internal tandem duplication. Overexpressed BAALC, ERG and MN1 genes and expression of breast cancer resistance protein have been shown to confer poor prognosis. A gene-expression signature previously suggested to separate cytogenetically normal acute myeloid leukemia patients into prognostic subgroups has been validated on a different microarray platform, although gene-expression signature-based classifiers predicting outcome for individual patients with greater accuracy are still needed. SUMMARY The discovery of new prognostic markers has increased our understanding of leukemogenesis and may lead to improved prognostication and generation of novel risk-adapted therapies.
Collapse
Affiliation(s)
- Krzysztof Mrózek
- Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive, Cancer Center, The Ohio State University, Columbus, Ohio 43210-1228, USA.
| | | | | |
Collapse
|
30
|
Scholl S, Mügge LO, Landt O, Loncarevic IF, Kunert C, Clement JH, Höffken K. Rapid screening and sensitive detection of NPM1 (nucleophosmin) exon 12 mutations in acute myeloid leukaemia. Leuk Res 2007; 31:1205-11. [PMID: 17306368 DOI: 10.1016/j.leukres.2006.12.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2006] [Revised: 12/15/2006] [Accepted: 12/17/2006] [Indexed: 10/23/2022]
Abstract
Nucleophosmin mutations of exon 12 (NPM1 mutations) represent the most frequent molecular aberration that can be found in patients with acute myeloid leukaemia (AML) and can be detected in about 35% of AML patients. NPM1 mutations are characterised by four basepair insertions within the region corresponding to the C-terminus of the protein leading to its translocation out of the nucleus. Until now, more than 40 different subsets of mutations have been identified and about 90% of NPM1 mutations are represented by subtype A and B (78% versus 12%, respectively). So far, standard screening of NPM1 mutations using conventional polymerase chain reaction (PCR) followed by capillary electrophoresis is rather time consuming. We established a new method for rapid screening of NPM1 mutations using the fluorescence resonance energy transfer (FRET) principle. Furthermore, based on individual NPM1 mutations type A and B, we designed mutation specific primers to perform a highly sensitive PCR assay that can be applied for the detection of minimal residual disease (MRD). In summary, we demonstrate new methodological approaches for rapid screening of NPM1 mutations as well as for MRD analyses based on the most frequent NPM1 mutations.
Collapse
Affiliation(s)
- S Scholl
- Department of Internal Medicine II (Oncology and Haematology), Friedrich Schiller University, Jena, Germany.
| | | | | | | | | | | | | |
Collapse
|
31
|
Thiede C, Creutzig E, Reinhardt D, Ehninger G, Creutzig U. Different types of NPM1 mutations in children and adults: evidence for an effect of patient age on the prevalence of the TCTG-tandem duplication in NPM1-exon 12. Leukemia 2006; 21:366-7. [PMID: 17151698 DOI: 10.1038/sj.leu.2404519] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|