51
|
Abstract
PURPOSE OF REVIEW Clonal hematopoiesis of indeterminate potential (CHIP) increases with age and occurs when a single mutant hematopoietic stem cell (HSC) contributes to a significant clonal proportion of mature blood lineages. Somatic mutations in the TP53 gene, which encodes the tumor suppressor protein p53, rank in the top five among genes that were mutated in CHIP. This review focuses on mechanisms by which mutant p53 promotes CHIP progression and drives the pathogenesis of hematological malignancies, including myelodysplastic syndromes, and acute myeloid leukemia. RECENT FINDINGS TP53 was frequently mutated in individuals with CHIP. Although clinical studies suggest that expansion of HSCs with TP53 mutations predisposes the elderly to hematological neoplasms, there is a significant gap in knowledge regarding the mechanisms by which TP53 mutations promote HSC expansion. Recent findings suggest that several cellular stressors, including hematopoietic transplantation, genotoxic stress, and inflammation, promote the expansion of HSCs with TP53 mutations. Further, TP53 mutations identified in CHIP cooperate with genetic and/or epigenetic changes in leukemogenesis. SUMMARY TP53 mutations identified in CHIP are associated with increased risks of de novo and therapy-related hematological neoplasms. Thus, targeting mutant p53 and related pathways holds great potential in preventing CHIP progression and treating hematological malignancies.
Collapse
Affiliation(s)
- Sisi Chen
- Department of Biochemistry and Molecular Biology
| | - Yan Liu
- Department of Biochemistry and Molecular Biology
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
52
|
Hunter AM, Sallman DA. Current status and new treatment approaches in TP53 mutated AML. Best Pract Res Clin Haematol 2019; 32:134-144. [PMID: 31203995 DOI: 10.1016/j.beha.2019.05.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
Mutations in the essential tumor suppressor gene, TP53, are observed in only 5-10% of acute myeloid leukemia (AML) cases, but are highly associated with therapy-related AML and cases with complex karyotype. The mutational status of TP53 is a critical prognostic indicator, with dismal outcomes consistently observed across studies. Response rates to traditional cytotoxic chemotherapy are poor and long-term survival after allogeneic hematopoietic stem cell transplant is rare. Therapy with hypomethylating agents has resulted in a modest improvement in outcomes over intensive chemotherapy, but durable responses are seldom observed. In view of the intrinsic resistance to standard chemotherapies conferred by mutations in TP53, novel treatment approaches are required. In this review, we examine the current treatment landscape in TP53 mutated AML and discuss emerging therapeutic approaches currently under clinical investigation.
Collapse
Affiliation(s)
- Anthony M Hunter
- Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - David A Sallman
- Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
53
|
Hepatic leukemia factor is a novel leukemic stem cell regulator in DNMT3A, NPM1, and FLT3-ITD triple-mutated AML. Blood 2019; 134:263-276. [PMID: 31076446 DOI: 10.1182/blood.2018862383] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 05/02/2019] [Indexed: 12/20/2022] Open
Abstract
FLT3, DNMT3A, and NPM1 are the most frequently mutated genes in cytogenetically normal acute myeloid leukemia (AML), but little is known about how these mutations synergize upon cooccurrence. Here we show that triple-mutated AML is characterized by high leukemia stem cell (LSC) frequency, an aberrant leukemia-specific GPR56 highCD34low immunophenotype, and synergistic upregulation of Hepatic Leukemia Factor (HLF). Cell sorting based on the LSC marker GPR56 allowed isolation of triple-mutated from DNMT3A/NPM1 double-mutated subclones. Moreover, in DNMT3A R882-mutated patients, CpG hypomethylation at the HLF transcription start site correlated with high HLF mRNA expression, which was itself associated with poor survival. Loss of HLF via CRISPR/Cas9 significantly reduced the CD34+GPR56+ LSC compartment of primary human triple-mutated AML cells in serial xenotransplantation assays. HLF knockout cells were more actively cycling when freshly harvested from mice, but rapidly exhausted when reintroduced in culture. RNA sequencing of primary human triple-mutated AML cells after shRNA-mediated HLF knockdown revealed the NOTCH target Hairy and Enhancer of Split 1 (HES1) and the cyclin-dependent kinase inhibitor CDKN1C/p57 as novel targets of HLF, potentially mediating these effects. Overall, our data establish HLF as a novel LSC regulator in this genetically defined high-risk AML subgroup.
Collapse
|
54
|
Prochazka KT, Pregartner G, Rücker FG, Heitzer E, Pabst G, Wölfler A, Zebisch A, Berghold A, Döhner K, Sill H. Clinical implications of subclonal TP53 mutations in acute myeloid leukemia. Haematologica 2019; 104:516-523. [PMID: 30309854 PMCID: PMC6395341 DOI: 10.3324/haematol.2018.205013] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023] Open
Abstract
The role of subclonal TP53 mutations, defined by a variant allele frequency of <20%, has not been addressed in acute myeloid leukemia yet. We, therefore, analyzed their prognostic value in a cohort of 1,537 patients with newly diagnosed disease, prospectively treated within three trials of the "German-Austrian Acute Myeloid Leukemia Study Group". Mutational analysis was performed by targeted deep sequencing and patients with TP53 mutations were categorized by their variant allele frequency into groups with frequencies >40%, 20%-40% and <20%. A total of 108 TP53 mutations were found in 98 patients (6.4%). Among these, 61 patients had variant allele frequencies >40%, 19 had variant allele frequencies between 20%-40% and 18 had frequencies <20%. Compared to specimens with clonal TP53 mutations, those with subclonal ones showed significantly fewer complex karyotypes and chromosomal losses. In either TP53-mutated group, patients experienced significantly fewer complete responses (P<0.001) and had worse overall and event-free survival rates (P<0.0001). In Cox regression analyses adjusting for age, white blood cell count, cytogenetic risk and type of acute myeloid leukemia, the adverse prognostic effect of TP53 mutations remained significant for all TP53-mutated subgroups. These data suggest that subclonal TP53 mutations are a novel prognostic parameter in acute myeloid leukemia and emphasize the usefulness of next-generation sequencing technologies for risk stratification in this disorder. The study was registered at ClinicalTrials.gov with number NCT00146120.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Alleles
- Chromosome Aberrations
- Clonal Evolution/genetics
- Female
- Gene Frequency
- Genes, p53
- Humans
- Kaplan-Meier Estimate
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Mutation
- Prognosis
- Young Adult
Collapse
Affiliation(s)
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Austria
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital of Ulm, Germany
| | - Ellen Heitzer
- Institute of Human Genetics, Medical University of Graz, Austria
| | - Gabriel Pabst
- Division of Hematology, Medical University of Graz, Austria
| | - Albert Wölfler
- Division of Hematology, Medical University of Graz, Austria
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, Austria
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Austria
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Germany
| | - Heinz Sill
- Division of Hematology, Medical University of Graz, Austria
| |
Collapse
|
55
|
Moison C, Lavallée VP, Thiollier C, Lehnertz B, Boivin I, Mayotte N, Gareau Y, Fréchette M, Blouin-Chagnon V, Corneau S, Lavallée S, Lemieux S, Marinier A, Hébert J, Sauvageau G. Complex karyotype AML displays G2/M signature and hypersensitivity to PLK1 inhibition. Blood Adv 2019; 3:552-563. [PMID: 30782614 PMCID: PMC6391664 DOI: 10.1182/bloodadvances.2018028480] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/11/2019] [Indexed: 01/07/2023] Open
Abstract
Patients diagnosed with acute myeloid leukemia with complex karyotype (CK AML) have an adverse prognosis using current therapies, especially when accompanied by TP53 alterations. We hereby report the RNA-sequencing analysis of the 68 CK AML samples included in the Leucegene 415 patient cohort. We confirm the frequent occurrence of TP53 alterations in this subgroup and further characterize the allele expression profile and transcript alterations of this gene. We also document that the RAS pathway (N/KRAS, NF1, PTPN11, BRAF) is frequently altered in this disease. Targeted chemical interrogation of genetically characterized primary CK AML samples identifies polo-like kinase 1 (PLK1) inhibitors as the most selective agents for this disease subgroup. TP53 status did not alter sensitivity to PLK1 inhibitors. Interestingly, CK AML specimens display a G2/M transcriptomic signature that includes higher expression levels of PLK1 and correlates with PLK1 inhibition sensitivity. Together, our results highlight vulnerability in CK AML. In line with these in vitro data, volasertib shows a strong anti-AML activity in xenotransplantation mouse models of human adverse AML. Considering that PLK1 inhibitors are currently being investigated clinically in AML and myelodysplastic syndromes, our results provide a new rationale for PLK1-directed therapy in patients with adverse cytogenetic AML.
Collapse
Affiliation(s)
- Céline Moison
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Vincent-Philippe Lavallée
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
| | - Clarisse Thiollier
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Bernhard Lehnertz
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Isabel Boivin
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Nadine Mayotte
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Yves Gareau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Mélanie Fréchette
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Valérie Blouin-Chagnon
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sophie Corneau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Lavallée
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada; and
| | - Sébastien Lemieux
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Department of Computer Science and Operations Research and
| | - Anne Marinier
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Josée Hébert
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada; and
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Guy Sauvageau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada; and
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
56
|
Chung J, Sallman DA, Padron E. TP53 and therapy-related myeloid neoplasms. Best Pract Res Clin Haematol 2019; 32:98-103. [PMID: 30927980 DOI: 10.1016/j.beha.2019.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 02/18/2019] [Indexed: 01/15/2023]
Abstract
Therapy-related myeloid neoplasms (t-MNs) are the most serious late complications in patients treated with traditional cytotoxic chemotherapy and/or radiation. T-MNs are aggressive and chemorefractory hematologic malignancies, with a median survival of less than 6 months. TP53 mutations are highly enriched in t-MN patients, though the mechanism for this selective enrichment has only come to light over the past several years. In this review, we discuss the history and function of p53, and the role of TP53 mutations in the origin and progression of t-MNs. Emerging data has begun to elucidate who may be at highest risk of developing t-MNs, which ideally will enable us to develop preventative strategies for this devastating disease. As t-MNs may not be avoidable, novel therapies are urgently needed for this patient group and are underway as exemplified by recent investigation in restoring wild-type p53 function as well as directly targeting TP53 mutant variants. With better prevention and treatment, outcomes will hopefully begin to improve in the near future.
Collapse
Affiliation(s)
- Jae Chung
- Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - David A Sallman
- Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Eric Padron
- Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
57
|
Gu R, Yang X, Wei H. Molecular landscape and targeted therapy of acute myeloid leukemia. Biomark Res 2018; 6:32. [PMID: 30455953 PMCID: PMC6225571 DOI: 10.1186/s40364-018-0146-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/04/2018] [Indexed: 12/22/2022] Open
Abstract
For decades, genetic aberrations including chromosome and molecular abnormalities are important diagnostic and prognostic factors in acute myeloid leukemia (AML). ATRA and imatinib have been successfully used in AML and chronic myelogenous leukemia, which proved that targeted therapy by identifying molecular lesions could improve leukemia outcomes. Recent advances in next generation sequencing have revealed molecular landscape of AML, presenting us with many molecular abnormalities. The individual prognostic information derived from a specific mutation could be modified by other molecular lesions. Therefore, the genomic complexity in AML poses a huge challenge to successful translation into more accurate risk stratification and targeted therapy. Herein, a summary of these mutations and targeted therapies are described. We focus on the prognostic information of recent identified molecular lesions and emerging targeted therapy.
Collapse
Affiliation(s)
- Runxia Gu
- Leukemia Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 People’s Republic of China
| | - Xue Yang
- Leukemia Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 People’s Republic of China
| | - Hui Wei
- Leukemia Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 People’s Republic of China
| |
Collapse
|
58
|
Kayser S, Levis MJ. Clinical implications of molecular markers in acute myeloid leukemia. Eur J Haematol 2018; 102:20-35. [PMID: 30203623 DOI: 10.1111/ejh.13172] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
The recently updated World Health Organization (WHO) Classification of myeloid neoplasms and leukemia reflects the fact that research in the underlying pathogenic mechanisms of acute myeloid leukemia (AML) has led to remarkable advances in our understanding of the disease. Gene mutations now allow us to explore the enormous diversity among cytogenetically defined subsets of AML, particularly the large subset of cytogenetically normal AML. Despite the progress in unraveling the tumor genome, only a small number of recurrent mutations have been incorporated into risk-stratification schemes and have been proven to be clinically relevant, targetable lesions. We here discuss the utility of molecular markers in AML in prognostication and treatment decision making, specifically highlighting the aberrations included in the current WHO classification.
Collapse
Affiliation(s)
- Sabine Kayser
- Department of Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark J Levis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
59
|
Marquis M, Beaubois C, Lavallée VP, Abrahamowicz M, Danieli C, Lemieux S, Ahmad I, Wei A, Ting SB, Fleming S, Schwarer A, Grimwade D, Grey W, Hills RK, Vyas P, Russell N, Sauvageau G, Hébert J. High expression of HMGA2 independently predicts poor clinical outcomes in acute myeloid leukemia. Blood Cancer J 2018; 8:68. [PMID: 30061630 PMCID: PMC6066481 DOI: 10.1038/s41408-018-0103-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/23/2018] [Accepted: 06/01/2018] [Indexed: 11/25/2022] Open
Abstract
In acute myeloid leukemia (AML), risk stratification based on cytogenetics and mutation profiling is essential but remains insufficient to select the optimal therapy. Accurate biomarkers are needed to improve prognostic assessment. We analyzed RNA sequencing and survival data of 430 AML patients and identified HMGA2 as a novel prognostic marker. We validated a quantitative PCR test to study the association of HMGA2 expression with clinical outcomes in 358 AML samples. In this training cohort, HMGA2 was highly expressed in 22.3% of AML, mostly in patients with intermediate or adverse cytogenetics. High expression levels of HMGA2 (H + ) were associated with a lower frequency of complete remission (58.8% vs 83.4%, P < 0.001), worse 3-year overall survival (OS, 13.2% vs 43.5%, P < 0.001) and relapse-free survival (RFS, 10.8% vs 44.2%, P < 0.001). A positive HMGA2 test also identified a subgroup of patients unresponsive to standard treatments. Multivariable analyses showed that H + was independently associated with significantly worse OS and RFS, including in the intermediate cytogenetic risk category. These associations were confirmed in a validation cohort of 260 patient samples from the UK NCRI AML17 trial. The HMGA2 test could be implemented in clinical trials developing novel therapeutic strategies for high-risk AML.
Collapse
Affiliation(s)
- Miriam Marquis
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Cyrielle Beaubois
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Vincent-Philippe Lavallée
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada
| | - Michal Abrahamowicz
- Epidemiology and Biostatistics Department, McGill University, Montréal, Canada
| | - Coraline Danieli
- Epidemiology and Biostatistics Department, McGill University, Montréal, Canada
| | - Sébastien Lemieux
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Department of Computer Science and Operations Research, Université de Montréal, Montréal, Canada
| | - Imran Ahmad
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Andrew Wei
- Department of Haematology, Alfred Hospital, Melbourne, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Stephen B Ting
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Department of Haematology, Eastern Health, Box Hill Hospital, Melbourne, Australia
| | - Shaun Fleming
- Department of Haematology, Alfred Hospital, Melbourne, Australia
| | - Anthony Schwarer
- Department of Haematology, Eastern Health, Box Hill Hospital, Melbourne, Australia
| | - David Grimwade
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College London, London, UK
- UK National Cancer Research Institute (NCRI) Haematological Oncology Clinical Studies Group, Cardiff, UK
| | - William Grey
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Robert K Hills
- UK National Cancer Research Institute (NCRI) Haematological Oncology Clinical Studies Group, Cardiff, UK
- Centre for Trials Research, Cardiff University School of Medicine, Cardiff, UK
| | - Paresh Vyas
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine and Department of Haematology, University of Oxford and Oxford University Hospitals NHS Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Nigel Russell
- UK National Cancer Research Institute (NCRI) Haematological Oncology Clinical Studies Group, Cardiff, UK
- Centre for Clinical Haematology, Nottingham University Hospital (City Hospital Campus), Nottingham, UK
| | - Guy Sauvageau
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Josée Hébert
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada.
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada.
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
60
|
Marquis M, Beaubois C, Lavallée VP, Abrahamowicz M, Danieli C, Lemieux S, Ahmad I, Wei A, Ting SB, Fleming S, Schwarer A, Grimwade D, Grey W, Hills RK, Vyas P, Russell N, Sauvageau G, Hébert J. High expression of HMGA2 independently predicts poor clinical outcomes in acute myeloid leukemia. Blood Cancer J 2018. [PMID: 30061630 DOI: 10.1038/s41408‐018‐0103‐6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In acute myeloid leukemia (AML), risk stratification based on cytogenetics and mutation profiling is essential but remains insufficient to select the optimal therapy. Accurate biomarkers are needed to improve prognostic assessment. We analyzed RNA sequencing and survival data of 430 AML patients and identified HMGA2 as a novel prognostic marker. We validated a quantitative PCR test to study the association of HMGA2 expression with clinical outcomes in 358 AML samples. In this training cohort, HMGA2 was highly expressed in 22.3% of AML, mostly in patients with intermediate or adverse cytogenetics. High expression levels of HMGA2 (H + ) were associated with a lower frequency of complete remission (58.8% vs 83.4%, P < 0.001), worse 3-year overall survival (OS, 13.2% vs 43.5%, P < 0.001) and relapse-free survival (RFS, 10.8% vs 44.2%, P < 0.001). A positive HMGA2 test also identified a subgroup of patients unresponsive to standard treatments. Multivariable analyses showed that H + was independently associated with significantly worse OS and RFS, including in the intermediate cytogenetic risk category. These associations were confirmed in a validation cohort of 260 patient samples from the UK NCRI AML17 trial. The HMGA2 test could be implemented in clinical trials developing novel therapeutic strategies for high-risk AML.
Collapse
Affiliation(s)
- Miriam Marquis
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada.,The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Cyrielle Beaubois
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada.,The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Vincent-Philippe Lavallée
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada.,Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada
| | - Michal Abrahamowicz
- Epidemiology and Biostatistics Department, McGill University, Montréal, Canada
| | - Coraline Danieli
- Epidemiology and Biostatistics Department, McGill University, Montréal, Canada
| | - Sébastien Lemieux
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada.,Department of Computer Science and Operations Research, Université de Montréal, Montréal, Canada
| | - Imran Ahmad
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Andrew Wei
- Department of Haematology, Alfred Hospital, Melbourne, Australia.,Australian Centre for Blood Diseases, Monash University, Melbourne, Australia.,Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Stephen B Ting
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia.,Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia.,Department of Haematology, Eastern Health, Box Hill Hospital, Melbourne, Australia
| | - Shaun Fleming
- Department of Haematology, Alfred Hospital, Melbourne, Australia
| | - Anthony Schwarer
- Department of Haematology, Eastern Health, Box Hill Hospital, Melbourne, Australia
| | - David Grimwade
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College London, London, UK.,UK National Cancer Research Institute (NCRI) Haematological Oncology Clinical Studies Group, Cardiff, UK
| | - William Grey
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Robert K Hills
- UK National Cancer Research Institute (NCRI) Haematological Oncology Clinical Studies Group, Cardiff, UK.,Centre for Trials Research, Cardiff University School of Medicine, Cardiff, UK
| | - Paresh Vyas
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine and Department of Haematology, University of Oxford and Oxford University Hospitals NHS Trust, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Nigel Russell
- UK National Cancer Research Institute (NCRI) Haematological Oncology Clinical Studies Group, Cardiff, UK.,Centre for Clinical Haematology, Nottingham University Hospital (City Hospital Campus), Nottingham, UK
| | - Guy Sauvageau
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada.,The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada.,Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Josée Hébert
- The Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, Montréal, Canada. .,The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada. .,Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Canada. .,Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
61
|
Abstract
PURPOSE OF REVIEW Assessment of measurable residual disease (MRD) after treatment can identify patients with acute myeloid leukemia (AML) that are at high risk of poor outcomes. However, there is no consensus yet regarding a standardized approach to measuring MRD that is most clinically meaningful. We review multiparameter flow cytometry (MFC) and reverse transcriptase polymerase chain reaction (RT-PCR), and discuss a framework for assessing remission MRD using next-generation sequencing (NGS). RECENT FINDINGS MFC and RT-PCR may not fully capitalize on the major advances that have been made in characterizing the genetic landscape of AML, which has offered insight into the biological and clinical implications of clonal genetic architecture. NGS has increasingly been shown to provide a qualitative and quantitative assessment of MRD with significant prognostic implications. The assessment of clonal architecture by NGS may complement or extend existing approaches for MRD monitoring. Long-term serial monitoring of diagnostic, remission, and relapse samples with clinical correlation will need to be performed in order to determine the impact of various MRD patterns using this technique.
Collapse
|
62
|
Prognostic factors influencing survival after allogeneic transplantation for AML/MDS patients with TP53 mutations. Blood 2018; 131:2989-2992. [PMID: 29769261 DOI: 10.1182/blood-2018-02-832360] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
63
|
Abstract
Real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR)-based detection of abnormal fusion transcripts is an important strategy for the diagnosis and monitoring of patients with acute myeloid leukemia (AML) with t(8;21)(q22;q22); RUNX1-RUNX1T1, inv(16)(p13.1;q22); CBFB-MYH11 or t(15;17)(q22;q12); PML-RARA. In RT-qPCR assays, patient-derived cDNA is subjected to amplification using PCR primers directed against the fusion transcript of interest as well as a reference gene for normalization. Quantification is typically performed by constructing standard curves for each PCR run using a series of plasmid standards of known concentration that harbor the same fusion transcript or the same reference gene of interest. Fusion transcripts and reference gene copy numbers are then calculated in patient samples using these standard curves. The process of constructing standard curves is laborious and consumes additional reagents. In this chapter, we give the method details for a multiplex RT-qPCR strategy to detect and quantify the acute myeloid leukemia (AML)-associated fusion transcripts PML-RARA in patients with t(15;17) without the need for standard curves. This general method can also be applied to other AML-associated fusion transcripts such as CBFB-MYH11 and RUNX1-RUNX1T1.
Collapse
|
64
|
Cheng Z, Hu K, Tian L, Dai Y, Pang Y, Cui W, Zhao H, Qin T, Han Y, Hu N, Chen L, Wang C, Zhang Y, Wu D, Ke X, Shi J, Fu L. Clinical and biological implications of mutational spectrum in acute myeloid leukemia of FAB subtypes M4 and M5. Cancer Gene Ther 2018; 25:77-83. [PMID: 29491461 DOI: 10.1038/s41417-018-0013-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/01/2017] [Accepted: 12/15/2017] [Indexed: 02/05/2023]
Abstract
The mutational spectrum and molecular characteristics of acute myelomonocytic lineage leukemia, namely acute myeloid leukemia (AML) French-American-British (FAB) subtypes M4 and M5, are largely unknown. In order to explore the mutational spectrum and prognostic factors of FAB-M4 and -M5, next-generation sequencing (NGS) was performed to screen for mutated genes and fusion genes relevant to the pathogenesis of AML. Of the 63 patients enrolled in the study, 60% had more than three mutated genes. NPM1 had the highest mutation frequency, followed by DNMT3A, FLT3, NRAS, RUNX1, and TET2. Univariate analysis suggested that age ≥60 years was an independent factor for both poor event-free survival (EFS) and overall survival (OS, P = 0.009, 0.002, respectively), MYH11-CBFβ was associated with better EFS and OS (P = 0.029, 0.016, respectively). However, multivariate analysis was not able to identify any independent risk factor for survival in the cohort of FAB-M4 and -M5 patients, including peripheral white blood cell count, bone marrow blast percentage, MYH11-CBFβ, FLT3-ITD, mutations in NPM1 and DNMT3A, and allogeneic hematopoietic stem cell transplantation (allo-HSCT). Our study provided new insight into the mutational spectrum and molecular characteristics of FAB-M4 and -M5. The clinical implications of the genetic signature of FAB-M4 and -M5 need to be further elucidated by larger studies.
Collapse
Affiliation(s)
- Zhiheng Cheng
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Kai Hu
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Lei Tian
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Yifeng Dai
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, China
| | - Yifan Pang
- Department of Medicine, William Beaumont Hospital, Royal Oak, MI, 48073, USA
| | - Wei Cui
- Department of Clinical Laboratory, Beijing Haidian Hospital, Beijing Haidian Section of Peking University Third Hospital, Beijing, 100080, China
| | - Hongmian Zhao
- Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Tong Qin
- Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Yu Han
- Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Ning Hu
- Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Li Chen
- Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Chao Wang
- Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Yijie Zhang
- Department of Respiratory, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Depei Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xiaoyan Ke
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Jinlong Shi
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, 475000, China.
- Department of Biomedical Engineering, Chinese PLA General Hospital, Beijing, 100853, China.
- Department of Medical Big Data, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Lin Fu
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China.
| |
Collapse
|
65
|
Distinct gene alterations with a high percentage of myeloperoxidase-positive leukemic blasts in de novo acute myeloid leukemia. Leuk Res 2018; 65:34-41. [PMID: 29306105 DOI: 10.1016/j.leukres.2017.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 11/01/2017] [Accepted: 12/30/2017] [Indexed: 10/18/2022]
Abstract
The myeloperoxidase (MPO)-positivity of blasts in bone marrow smears is an important marker for not only the diagnosis, but also the prognosis of acute myeloid leukemia (AML). To investigate the relationship between genetic alterations and MPO-positivity, we performed targeted sequencing for 51 genes and 10 chimeric gene transcripts in 164 newly diagnosed de novo AML patients; 107 and 57 patients were classified as AML with >50% MPO-positive blasts (MPO-high group) and ≤50% MPO-positive blasts, (MPO-low group), respectively. The univariate analysis revealed that RUNX1-RUNX1T1 (P < 0.001), the KIT mutation (P < 0.001), and CEBPA double mutation (P = 0.001) were more likely to be found in the MPO-high group, while the DNMT3A mutation (P = 0.001), FLT3 tyrosine kinase domain mutation (P = 0.004), and TP53 mutation (P = 0.020) were more likely to be present in the MPO-low group. Mutations in genes related to DNA hypermethylation signatures (IDH1, IDH2, TET2, and WT1 genes) were more frequent in the MPO-high group (P = 0.001) when patients with fusion genes of core-binding factors were excluded from the analysis. Our results suggest that MPO-positivity of blasts was related with the distinct gene mutation patterns among de novo AML patients.
Collapse
|
66
|
Conventional and Molecular Cytogenomic Basis of Hematologic Malignancies. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
67
|
Wang B, Liu Y, Hou G, Wang L, Lv N, Xu Y, Xu Y, Wang X, Xuan Z, Jing Y, Li H, Jin X, Deng A, Wang L, Gao X, Dou L, Liang J, Chen C, Li Y, Yu L. Mutational spectrum and risk stratification of intermediate-risk acute myeloid leukemia patients based on next-generation sequencing. Oncotarget 2017; 7:32065-78. [PMID: 27062340 PMCID: PMC5077997 DOI: 10.18632/oncotarget.7028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 01/13/2016] [Indexed: 11/25/2022] Open
Abstract
Intermediate-risk acute myeloid leukemia (IR-AML), which accounts for a substantial number of AML cases, is highly heterogeneous. Although several mutations have been identified, the heterogeneity of AML is uncertain because novel mutations have yet to be discovered. Here we applied next generation sequencing (NGS) platform to screen mutational hotspots in 410 genes relevant to hematological malignancy. IR-AML samples (N=95) were sequenced by Illumina Hiseq and mutations in 101 genes were identified. Only seven genes (CEBPA, NPM1, DNMT3A, FLT3-ITD, NRAS, IDH2 and WT1) were mutated in more than 10% of patients. Genetic interaction analysis identified several cooperative and exclusive patterns of overlapping mutations. Mutational analysis indicated some correlation between genotype and phenotype. FLT3-ITD mutations were identified as independent factors of poor prognosis, while CEBPA mutations were independent favorable factors. Co-occurrence of FLT3-ITD, NPM1 and DNMT3A mutations was identified with associated with specific clinical AML features and poor outcomes. Furthermore, by integrating multiple mutations in the survival analysis, 95 IR-AML patients could be stratified into three distinct risk groups allowing reductions in IR-AML by one-third. Our study offers deep insights into the molecular pathogenesis and biology of AML and indicated that the prognosis of IR-AML could be further stratified by different mutation combinations which may direct future treatment intervention.
Collapse
Affiliation(s)
- Bianhong Wang
- Medical Center, Tsinghua University, Beijing 100084, China
| | - Yangyang Liu
- Research and Development Department, Annoroad Gene Technology Co. Ltd, Beijing 100176, China
| | - Guangyuan Hou
- Research and Development Department, Annoroad Gene Technology Co. Ltd, Beijing 100176, China
| | - Lili Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Na Lv
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yuanyuan Xu
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yihan Xu
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiuli Wang
- Research and Development Department, Annoroad Gene Technology Co. Ltd, Beijing 100176, China
| | - Zhaoling Xuan
- Research and Development Department, Annoroad Gene Technology Co. Ltd, Beijing 100176, China
| | - Yu Jing
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Honghua Li
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiangshu Jin
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Ailing Deng
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Li Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaoning Gao
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Liping Dou
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Junbin Liang
- Research and Development Department, Annoroad Gene Technology Co. Ltd, Beijing 100176, China
| | - Chongjian Chen
- Research and Development Department, Annoroad Gene Technology Co. Ltd, Beijing 100176, China
| | - Yonghui Li
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - Li Yu
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
68
|
Aziz H, Ping CY, Alias H, Ab Mutalib NS, Jamal R. Gene Mutations as Emerging Biomarkers and Therapeutic Targets for Relapsed Acute Myeloid Leukemia. Front Pharmacol 2017; 8:897. [PMID: 29270125 PMCID: PMC5725465 DOI: 10.3389/fphar.2017.00897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/24/2017] [Indexed: 12/19/2022] Open
Abstract
It is believed that there are key differences in the genomic profile between adult and childhood acute myeloid leukemia (AML). Relapse is the significant contributor of mortality in patients with AML and remains as the leading cause of cancer death among children, posing great challenges in the treatment of AML. The knowledge about the genomic lesions in childhood AML is still premature as most genomic events defined in children were derived from adult cohorts. However, the emerging technologies of next generation sequencing have narrowed the gap of knowledge in the biology of AML by the detection of gene mutations for each sub-type which have led to the improvement in terms of prognostication as well as the use of targeted therapies. In this review, we describe the recent understanding of the genomic landscape including the prevalence of mutation, prognostic impact, and targeted therapies that will provide an insight into the pathogenesis of AML relapse in both adult and childhood cases.
Collapse
Affiliation(s)
- Habsah Aziz
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Chow Y Ping
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Hamidah Alias
- Department of Paediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Rahman Jamal
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
69
|
Mevatee P, Tantiworawit A, Traisathit P, Puaninta C, Mevatee U, Angsuchawan S, Bumroongkit K. FLT3-ITD, NPM1, and DNMT3A Gene Mutations and Risk Factors in Normal Karyotype Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients in Upper Northern Thailand. Asian Pac J Cancer Prev 2017; 18:3031-3039. [PMID: 29172276 PMCID: PMC5773788 DOI: 10.22034/apjcp.2017.18.11.3031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Objective: Approximately 40-45% of AML and MDS patients have a cytogenetically normal karyotype (CN-AML and CN-MDS). The frequency and types of gene mutations in these cases may differ among various populations. The objective of this study was to identify frequencies and types of FLT3-ITD, NPM1, and DNMT3A mutations, and associations of them with clinical data and risk factors in CN-AML and CN-MDS cases in upper Northern Thailand. Methods: Bone marrow samples of 40 CN-AML and 60 CN-MDS patients were analyzed for gene mutations by direct sequencing. In addition, data for potential risk factors were obtained for comparison. Results: Frequencies of FLT3-ITD, NPM1, and DNMT3A mutations were 25.0%, 17.5%, and 10.0%, respectively in CN-AML, but all zero in CN-MDS cases. NPM1 mutations were found at a median age older than the wild type (58 vs 47 years) while DNMT3A mutations were associated with an increase in the white blood cell count. In all patients, factors for the mutations of these three genes included age ≤ 60 years, and a history of hypertension. Conclusion: When considering mutations in only normal karyotype patients, the frequency of FLT3-ITD, NPM1, DNMT3A mutations in CN-AML patients in upper Northern Thailand were found to occur at lower rates than in Western patients and to differ from other Asian populations including parts of Thailand. No mutations were observed in CN-MDS cases. Some types of gene mutations differed from previous studies, possibly attributable to differences in geography, lifestyle and genetic backgrounds. Links with age ≤ 60 years and history of hypertension were found. Investigation of these three genes in an intermediate risk group with a normal karyotype is useful for a better understanding of molecular leukemogenetic steps in CN-AML and CN-MDS patients and may be beneficial for planning treatment and prevention in the population of upper Northern Thailand.
Collapse
Affiliation(s)
- Piyanan Mevatee
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Thailand.
| | | | | | | | | | | | | |
Collapse
|
70
|
|
71
|
McCurdy SR, Levis MJ. Emerging molecular predictive and prognostic factors in acute myeloid leukemia. Leuk Lymphoma 2017; 59:2021-2039. [DOI: 10.1080/10428194.2017.1393669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Shannon R. McCurdy
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark J. Levis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
72
|
Sweet K, Lancet J. State of the Art Update and Next Questions: Acute Myeloid Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:703-709. [PMID: 29110833 DOI: 10.1016/j.clml.2017.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 01/02/2023]
Abstract
As our general understanding regarding the complex nature of acute myeloid leukemia (AML) is expanding, so is our ability to translate this biological data into clinically relevant information. The use of whole genome and whole exome sequencing has begun to shed light on the importance of a variety of somatic mutations that are frequently identified in AML. In turn, this has allowed the field to incorporate mutational data into prognostic classifications which can guide treatment decisions. Furthermore, minimal residual disease (MRD) monitoring in AML is more commonplace as the prognostic relevance of MRD at various time points during treat is becoming clear. Many novel treatments have recently been approved, or are expected to gain approval in the near future, and this is opening the door to a more personalized approach to the management of AML.
Collapse
|
73
|
Simon L, Lavallée VP, Bordeleau ME, Krosl J, Baccelli I, Boucher G, Lehnertz B, Chagraoui J, MacRae T, Ruel R, Chantigny Y, Lemieux S, Marinier A, Hébert J, Sauvageau G. Chemogenomic Landscape of RUNX1-mutated AML Reveals Importance of RUNX1 Allele Dosage in Genetics and Glucocorticoid Sensitivity. Clin Cancer Res 2017; 23:6969-6981. [DOI: 10.1158/1078-0432.ccr-17-1259] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/07/2017] [Accepted: 08/24/2017] [Indexed: 11/16/2022]
|
74
|
Abstract
Therapy-related myeloid neoplasms (t-MN) arise as a late effect of chemotherapy and/or radiation administered for a primary condition, typically a malignant disease, solid organ transplant or autoimmune disease. Survival is measured in months, not years, making t-MN one of the most aggressive and lethal cancers. In this Review, we discuss recent developments that reframe our understanding of the genetic and environmental aetiology of t-MN. Emerging data are illuminating who is at highest risk of developing t-MN, why t-MN are chemoresistant and how we may use this information to treat and ultimately prevent this lethal disease.
Collapse
MESH Headings
- Antineoplastic Agents, Alkylating/adverse effects
- Bone Marrow Cells
- Chromosome Aberrations
- Chromosomes, Human, Pair 5
- Chromosomes, Human, Pair 7
- Clone Cells/physiology
- Gene-Environment Interaction
- Genetic Predisposition to Disease
- Hematopoiesis
- Humans
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/therapy
- Mutation
- Myelodysplastic Syndromes/etiology
- Myelodysplastic Syndromes/therapy
- Neoplasms, Second Primary/etiology
- Neoplasms, Second Primary/therapy
- Prognosis
- Radiation Exposure/adverse effects
- Risk Factors
Collapse
Affiliation(s)
- Megan E McNerney
- Department of Pathology and the Department of Pediatrics, The University of Chicago, Chicago, Illinois 60637, USA
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois 60637, USA
| | - Lucy A Godley
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois 60637, USA
| | - Michelle M Le Beau
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois 60637, USA
| |
Collapse
|
75
|
Chung C, Ma H. Driving Toward Precision Medicine for Acute Leukemias: Are We There Yet? Pharmacotherapy 2017; 37:1052-1072. [DOI: 10.1002/phar.1977] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Hilary Ma
- MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
76
|
You E, Cho YU, Jang S, Seo EJ, Lee JH, Lee JH, Lee KH, Koh KN, Im HJ, Seo JJ, Park YM, Lee JK, Park CJ. Frequency and Clinicopathologic Features of RUNX1 Mutations in Patients With Acute Myeloid Leukemia Not Otherwise Specified. Am J Clin Pathol 2017; 148:64-72. [PMID: 28927163 DOI: 10.1093/ajcp/aqx046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To evaluate the frequency and clinicopathologic characteristics of RUNX1 mutations, focusing on patients with acute myeloid leukemia not otherwise specified (AML NOS). METHODS Diagnostic samples from 219 patients with AML NOS were analyzed for RUNX1 mutations using standard polymerase chain reaction and direct sequencing. RESULTS Thirty-one RUNX1 mutations were detected in 33 (15.1%) patients. Mutations clustered in the Runt homology (61.3%) and transactivation domains (25.8%). Frameshift mutations were most common (51.6%), followed by missense (41.9%) and nonsense (6.5%) mutations. Patients with RUNX1 mutations had a lower platelet count (P = .013) and shorter relapse-free survival (P = .045) than those without. The presence of RUNX1 and NPM1 or CEBPA mutations was mutually exclusive. A literature review, including our study, showed that patients with RUNX1 mutations were associated with intermediate risk; coexisting mutations such as FLT3-ITD, ASXL1, TET2, and DNMT3A; and a relatively cytogenetic heterogeneity. CONCLUSIONS Our findings strengthen previous data concerning RUNX1 mutations in AML and support the notion that RUNX1 mutational status should be integrated into a diagnostic workup of AML, particularly for AML NOS or an intermediate-risk group.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Young-Mi Park
- Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Jong-Keuk Lee
- Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | | |
Collapse
|
77
|
High-throughput sequencing for noninvasive disease detection in hematologic malignancies. Blood 2017; 130:440-452. [PMID: 28600337 DOI: 10.1182/blood-2017-03-735639] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Noninvasive monitoring of minimal residual disease (MRD) has led to significant advances in personalized management of patients with hematologic malignancies. Improved therapeutic options and prolonged survival have further increased the need for sensitive tumor assessment that can inform treatment decisions and patient outcomes. At diagnosis or relapse of most hematologic neoplasms, malignant cells are often easily accessible in the blood as circulating tumor cells (CTCs), making them ideal targets to noninvasively profile the molecular features of each patient. In other cancer types, CTCs are generally rare and noninvasive molecular detection relies on circulating tumor DNA (ctDNA) shed from tumor deposits into circulation. The ability to precisely detect and quantify CTCs and ctDNA could minimize invasive procedures and improve prediction of clinical outcomes. Technical advances in MRD detection methods in recent years have led to reduced costs and increased sensitivity, specificity, and applicability. Among currently available tests, high-throughput sequencing (HTS)-based approaches are increasingly attractive for noninvasive molecular testing. HTS-based methods can simultaneously identify multiple genetic markers with high sensitivity and specificity without individual optimization. In this review, we present an overview of techniques used for noninvasive molecular disease detection in selected myeloid and lymphoid neoplasms, with a focus on the current and future role of HTS-based assays.
Collapse
|
78
|
Lal R, Lind K, Heitzer E, Ulz P, Aubell K, Kashofer K, Middeke JM, Thiede C, Schulz E, Rosenberger A, Hofer S, Feilhauer B, Rinner B, Svendova V, Schimek MG, Rücker FG, Hoefler G, Döhner K, Zebisch A, Wölfler A, Sill H. Somatic TP53 mutations characterize preleukemic stem cells in acute myeloid leukemia. Blood 2017; 129:2587-2591. [PMID: 28258055 DOI: 10.1182/blood-2016-11-751008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | | | | | | | | | - Karl Kashofer
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Jan M Middeke
- Department of Internal Medicine I, University Hospital Dresden, Dresden, Germany
| | - Christian Thiede
- Department of Internal Medicine I, University Hospital Dresden, Dresden, Germany
| | | | | | | | | | | | - Vendula Svendova
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria; and
| | - Michael G Schimek
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria; and
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | | | | | | |
Collapse
|
79
|
Mori J, Yanada M, Uchida N, Fukuda T, Sakura T, Hidaka M, Watakabe-Inamoto K, Kanamori H, Ogawa H, Ichinohe T, Tanaka J, Atsuta Y, Yano S. Outcomes of Allogeneic Hematopoietic Cell Transplantation in Acute Myeloid Leukemia Patients with Abnormalities of the Short Arm of Chromosome 17. Biol Blood Marrow Transplant 2017; 23:1398-1404. [PMID: 28455005 DOI: 10.1016/j.bbmt.2017.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/20/2017] [Indexed: 01/12/2023]
Abstract
We retrospectively analyzed a Japanese nationwide database to elucidate the impact of abnormalities in the short arm of chromosome 17 (abnl[17p]) on the outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with acute myeloid leukemia. Of 10,923 patients, 262 (2.4%) had abnl(17p), 235 of whom were classified into the poor cytogenetic risk group according to the National Comprehensive Cancer Network criteria. The median follow-up period was 1425 days. In abnl(17p) versus non-abnl(17p) patients of poor cytogenetic risk group, overall survival (OS), disease-free survival, cumulative incidence of disease relapse, and nonrelapse mortality rates at 5 years after allo-HSCT were 9.2% versus 27.4%, 7.8% versus 25.0%, 66.6% versus 49.4%, and 25.6% versus 25.6%, respectively. In contrast to the other types of abnl(17p), isochromosome 17q rarely encompassed the poor cytogenetic risk traits and did not adversely affect OS. Among the abnl(17p) patients, male sex, nonremission disease status at transplantation, and poor cytogenetic risk group were significantly associated with shorter OS. In conclusion, the presence of an abnl(17p) negatively affects allo-HSCT outcomes, which are influenced by the type of abnormality. Prompt initiation of allo-HSCT during complete remission may improve outcomes.
Collapse
Affiliation(s)
- Jinichi Mori
- Institute of Medical Science, the University of Tokyo, Tokyo, Japan; Department of Hematology, Jyoban Hospital, Tokiwakai group, Fukushima, Japan.
| | - Masamitsu Yanada
- Department of Hematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Naoyuki Uchida
- Department of Hematology, Toranomon Hospital, Toranomon, Tokyo, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Toru Sakura
- Leukemia Research Center, Saiseikai Maebashi Hospital, Gunma, Japan
| | - Michihiro Hidaka
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Kyoko Watakabe-Inamoto
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Heiwa Kanamori
- Department of Hematology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Hiroyasu Ogawa
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Hyogo, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Junji Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Aichi, Japan; Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
80
|
Sandhöfer N, Metzeler KH, Kakadia PM, Pasalic Z, Hiddemann W, Neusser M, Steinlein O, Fiegl M, Subklewe M, Spiekermann K, Bohlander SK, Schneider S, Braess J. A fluorescence in situ hybridization-based screen allows rapid detection of adverse cytogenetic alterations in patients with acute myeloid leukemia. Genes Chromosomes Cancer 2017; 56:632-638. [PMID: 28420034 DOI: 10.1002/gcc.22466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 04/11/2017] [Accepted: 04/11/2017] [Indexed: 11/12/2022] Open
Abstract
In adult acute myeloid leukemia (AML), the karyotype of the leukemic cell is among the strongest prognostic factors. The Medical Research Council (MRC) and the European LeukemiaNet (ELN) classifications distinguish between favorable, intermediate and adverse cytogenetic risk patients who differ in their treatment response and overall survival. Conventional cytogenetic analyses are a mandatory component of AML diagnostics but they are time-consuming; therefore, therapeutic decisions in elderly patients are often delayed. We investigated whether a screening approach using a panel of seven fluorescence in situ hybridization (FISH) probes would allow rapid identification of adverse chromosomal changes. In a cohort of 334 AML patients, our targeted FISH screening approach identified 80% of adverse risk AML patients with a specificity of 99%. Incorporating FISH screening into diagnostic workup has the potential to accelerate risk stratification and treatment selection, particularly in older patients. This approach may allow therapeutic decisions more quickly, which benefits both patients and physicians and might save costs.
Collapse
Affiliation(s)
- Nadine Sandhöfer
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Klaus H Metzeler
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Purvi M Kakadia
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
| | - Zlatana Pasalic
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Wolfgang Hiddemann
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Michaela Neusser
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Ortrud Steinlein
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Michael Fiegl
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Marion Subklewe
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Karsten Spiekermann
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan K Bohlander
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
| | - Stephanie Schneider
- Department of Internal Medicine III, Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, Munich, Germany
| | - Jan Braess
- Department of Oncology and Hematology, Hospital Barmherzige Brüder, Regensburg, Germany
| |
Collapse
|
81
|
Nishida Y, Maeda A, Kim MJ, Cao L, Kubota Y, Ishizawa J, AlRawi A, Kato Y, Iwama A, Fujisawa M, Matsue K, Weetall M, Dumble M, Andreeff M, Davis TW, Branstrom A, Kimura S, Kojima K. The novel BMI-1 inhibitor PTC596 downregulates MCL-1 and induces p53-independent mitochondrial apoptosis in acute myeloid leukemia progenitor cells. Blood Cancer J 2017; 7:e527. [PMID: 28211885 PMCID: PMC5386342 DOI: 10.1038/bcj.2017.8] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022] Open
Abstract
Disease recurrence is the major problem in the treatment of acute myeloid leukemia (AML). Relapse is driven by leukemia stem cells, a chemoresistant subpopulation capable of re-establishing disease. Patients with p53 mutant AML are at an extremely high risk of relapse. B-cell-specific Moloney murine leukemia virus integration site 1 (BMI-1) is required for the self-renewal and maintenance of AML stem cells. Here we studied the effects of a novel small molecule inhibitor of BMI-1, PTC596, in AML cells. Treatment with PTC596 reduced MCL-1 expression and triggered several molecular events consistent with induction of mitochondrial apoptosis: loss of mitochondrial membrane potential, BAX conformational change, caspase-3 cleavage and phosphatidylserine externalization. PTC596 induced apoptosis in a p53-independent manner. PTC596 induced apoptosis along with the reduction of MCL-1 and phosphorylated AKT in patient-derived CD34+CD38low/− stem/progenitor cells. Mouse xenograft models demonstrated in vivo anti-leukemia activity of PTC596, which inhibited leukemia cell growth in vivo while sparing normal hematopoietic cells. Our results indicate that PTC596 deserves further evaluation in clinical trials for refractory or relapsed AML patients, especially for those with unfavorable complex karyotype or therapy-related AML that are frequently associated with p53 mutations.
Collapse
Affiliation(s)
- Y Nishida
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - A Maeda
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - M J Kim
- PTC Therapeutics, South Plainfield, NJ, USA
| | - L Cao
- PTC Therapeutics, South Plainfield, NJ, USA
| | - Y Kubota
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - J Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A AlRawi
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Y Kato
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - A Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - M Fujisawa
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - K Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - M Weetall
- PTC Therapeutics, South Plainfield, NJ, USA
| | - M Dumble
- Bristol-Myers Squibb, Princeton, NJ, USA
| | - M Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T W Davis
- PMV Pharmaceuticals Inc., Cranbury, NJ, USA
| | | | - S Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - K Kojima
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| |
Collapse
|
82
|
Molecular Mutations and Their Cooccurrences in Cytogenetically Normal Acute Myeloid Leukemia. Stem Cells Int 2017; 2017:6962379. [PMID: 28197208 PMCID: PMC5288537 DOI: 10.1155/2017/6962379] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/25/2016] [Indexed: 01/08/2023] Open
Abstract
Adult acute myeloid leukemia (AML) clinically is a disparate disease that requires intensive treatments ranging from chemotherapy alone to allogeneic hematopoietic cell transplantation (allo-HCT). Historically, cytogenetic analysis has been a useful prognostic tool to classify patients into favorable, intermediate, and unfavorable prognostic risk groups. However, the intermediate-risk group, consisting predominantly of cytogenetically normal AML (CN-AML), itself exhibits diverse clinical outcomes and requires further characterization to allow for more optimal treatment decision-making. The recent advances in clinical genomics have led to the recategorization of CN-AML into favorable or unfavorable subgroups. The relapsing nature of AML is thought to be due to clonal heterogeneity that includes founder or driver mutations present in the leukemic stem cell population. In this article, we summarize the clinical outcomes of relevant molecular mutations and their cooccurrences in CN-AML, including NPM1, FLT3ITD, DNMT3A, NRAS, TET2, RUNX1, MLLPTD, ASXL1, BCOR, PHF6, CEBPAbiallelic, IDH1, IDH2R140, and IDH2R170, with an emphasis on their relevance to the leukemic stem cell compartment. We have reviewed the available literature and TCGA AML databases (2013) to highlight the potential role of stem cell regulating factor mutations on outcome within newly defined AML molecular subgroups.
Collapse
|
83
|
Poiré X, Labopin M, Maertens J, Yakoub-Agha I, Blaise D, Ifrah N, Socié G, Gedde-Dhal T, Schaap N, Cornelissen JJ, Vigouroux S, Sanz J, Michaux L, Esteve J, Mohty M, Nagler A. Allogeneic stem cell transplantation in adult patients with acute myeloid leukaemia and 17p abnormalities in first complete remission: a study from the Acute Leukemia Working Party (ALWP) of the European Society for Blood and Marrow Transplantation (EBMT). J Hematol Oncol 2017; 10:20. [PMID: 28100265 PMCID: PMC5241968 DOI: 10.1186/s13045-017-0393-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 01/03/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Acute myeloid leukaemia (AML) with 17p abnormalities (abn(17p)) carries a very poor prognosis due to high refractoriness to conventional chemotherapy, and allogeneic stem cell transplantation (allo-SCT) appears as the only potential curative option. METHODS To address outcomes after allo-SCT in patients with abn(17p), we retrospectively analysed de novo or secondary AML undergoing SCT between 2000 and 2013 from the EBMT registry. RESULTS One hundred thirty-nine patients with confirmed abn(17p) have been selected. At the time of transplant, one hundred twenty-five were in first remission (CR1). Median age was 54 years old. Abn(17p) was associated with a monosomal karyotype in 83% of patients, complex karyotype in 91%, monosomy 5 or 5q deletion (-5/5q-) in 55%, monosomy 7 (-7) in 39% and both -5/5q and -7 in 27%. Seventy-three patients (59%) had a reduced-intensity conditioning regimen. The 2-year overall survival (OS) and leukaemia-free survival (LFS) were 28 and 24%, respectively. The 2-year non-relapse mortality (NRM) was 15%, and 2-year relapse incidence (RI) was 61%. The cumulative incidence of grade II to IV acute graft-versus-host disease (GvHD) was 24% and that of chronic GvHD was 21%. In multivariate analysis, the presence of a -5/5q- in addition to abn(17p) was significantly and independently associated with worse OS, LFS and higher RI. Age and donor types did not correlate with outcome. Conditioning intensity was not statistically associated with OS, LFS and NRM when adjusted for patients' age. CONCLUSIONS In contrast to the dismal prognosis reported for AML patients harbouring abn(17p) undergoing conventional chemotherapy, allogeneic SCT provides responses in about 25% of those patients transplanted in CR1.
Collapse
Affiliation(s)
- Xavier Poiré
- Section of Hematology, Department of Medicine, Cliniques Universitaires Saint-Luc, 10, avenue Hippocrate, 1200, Brussels, Belgium.
| | - Myriam Labopin
- Acute Leukemia Working Party of the EBMT office, Paris, France.,Service d'Hématologie clinique, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, INSERM UMRs U938, Paris, France
| | - Johan Maertens
- Department of Hematology, University Hospital Gasthuisberg, Leuven, Belgium
| | | | - Didier Blaise
- Programme de Transplantation et Thérapie Cellulaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, Marseille, France
| | | | - Gérard Socié
- Department of Hematology, Hôpital Saint-Louis, Paris, France
| | | | - Nicolaas Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan J Cornelissen
- Daniel den Hoed Cancer Centre, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Jaime Sanz
- Servicio de Hematologia, Hospital Universitario La Fe, Valencia, Spain
| | - Lucienne Michaux
- Center for Human Genetics, KU Leuven and University Hospitals, Leuven, Belgium
| | - Jordi Esteve
- Hematology department, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - Mohamad Mohty
- Acute Leukemia Working Party of the EBMT office, Paris, France.,Service d'Hématologie clinique, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, INSERM UMRs U938, Paris, France
| | - Arnon Nagler
- Acute Leukemia Working Party of the EBMT office, Paris, France.,Chaim Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
84
|
Clinical Relevance of RUNX1 and CBFB Alterations in Acute Myeloid Leukemia and Other Hematological Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 962:175-199. [PMID: 28299658 DOI: 10.1007/978-981-10-3233-2_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The translocation t(8;21), leading to a fusion between the RUNX1 gene and the RUNX1T1 locus, was the first chromosomal translocation identified in cancer. Since the first description of this balanced rearrangement in a patient with acute myeloid leukemia (AML) in 1973, RUNX1 translocations and point mutations have been found in various myeloid and lymphoid neoplasms. In this chapter, we summarize the currently available data on the clinical relevance of core binding factor gene alterations in hematological disorders. In the first section, we discuss the prognostic implications of the core binding factor translocations RUNX1-RUNX1T1 and CBFB-MYH11 in AML patients. We provide an overview of the cooperating genetic events in patients with CBF-rearranged AML and their clinical implications, and review current treatment approaches for CBF AML and the utility of minimal residual disease monitoring. In the next sections, we summarize the available data on rare RUNX1 rearrangements in various hematologic neoplasms and the role of RUNX1 translocations in therapy-related myeloid neoplasia. The final three sections of the chapter cover the spectrum and clinical significance of RUNX1 point mutations in AML and myelodysplastic syndromes, in familial platelet disorder with associated myeloid malignancy, and in acute lymphoblastic leukemia.
Collapse
|
85
|
Abstract
Cytogenetic analysis has an essential role in diagnosis, classification, and prognosis of myelodysplastic syndromes (MDS). Some cytogenetic abnormalities are sufficiently characteristic of MDS to be considered MDS defining in the appropriate clinical context. MDS with isolated del(5q) is the only molecularly defined MDS subtype. The genes responsible for many aspects of 5q- syndrome, the distinct clinical phenotype associated with this condition, have now been identified. Cytogenetics forms the cornerstone of the most widely adopted prognostic scoring systems in MDS, the international prognostic scoring system (IPSS) and the revised international prognostic scoring system (IPPS-R). Cytogenetic parameters also have utility in chronic myelomonocytic leukemia (CMML) and have been incorporated into specific prognostic scoring systems for this condition. More recently, it has been appreciated that submicroscopic copy number changes and gene mutations play a significant part in MDS pathogenesis. Integration of molecular genetics and cytogenetics holds much promise for improving clinical care and outcomes for patients with MDS.
Collapse
Affiliation(s)
- Meaghan Wall
- Victorian Cancer Cytogenetics Service, St. Vincent's Hospital, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia.
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
86
|
Zhang L, McGraw KL, Sallman DA, List AF. The role of p53 in myelodysplastic syndromes and acute myeloid leukemia: molecular aspects and clinical implications. Leuk Lymphoma 2016; 58:1777-1790. [PMID: 27967292 DOI: 10.1080/10428194.2016.1266625] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
TP53 gene mutations occurring in patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are associated with high-risk karyotypes including 17p abnormalities, monosomal and complex cytogenetics. TP53 mutations in these disorders portend rapid disease progression and resistance to conventional therapeutics. Notably, the size of the TP53 mutant clone as measured by mutation allele burden is directly linked to overall survival (OS) confirming the importance of p53 as a negative prognostic variable. In nucleolar stress-induced ribosomopathies, such as del(5q) MDS, disassociation of MDM2 and p53 results in p53 accumulation in erythroid precursors manifested as erythroid hypoplasia. P53 antagonism by lenalidomide or other therapeutics such as antisense oligonucleotides, repopulates erythroid precursors and enhances effective erythropoiesis. These findings demonstrate that p53 is an intriguing therapeutic target that is currently under investigation in MDS and AML. This study reviews molecular advances in understanding the role of p53 in MDS and AML, and explores potential therapeutic strategies in this era of personalized medicine.
Collapse
Affiliation(s)
- Ling Zhang
- a Department of Hematopathology and Laboratory Medicine , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Kathy L McGraw
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - David A Sallman
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Alan F List
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| |
Collapse
|
87
|
Castelli G, Pelosi E, Testa U. Targeted therapies in the treatment of adult acute myeloid leukemias: current status and future perspectives. Int J Hematol Oncol 2016; 5:143-164. [PMID: 30302215 PMCID: PMC6172000 DOI: 10.2217/ijh-2016-0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 11/29/2016] [Indexed: 12/19/2022] Open
Abstract
The rapid advancement of next-generation sequencing techniques and the identification of molecular driver events responsible for leukemia development are opening the door to new pharmacologic-targeted agents to tailor treatment of acute myeloid leukemia (AML) in individual patients. However, the use of targeted therapies in AML has met with only modest success. Molecular studies have identified AML subsets characterized by driver mutational events, such as NPM1, FLT3-ITD and IDH1-2 mutations, and have provided preclinical evidence that the targeting of these mutant molecules could represent a valuable therapeutic strategy. Recent studies have provided the first pieces of evidence that FLT3 targeting in FLT3-mutant AMLs, IDH1/2 inhibition in IDH-mutant AMLs and targeting membrane molecules preferentially expressed on leukemic progenitor/stem cells, such as CD33 and CD123, represent a clinically valuable strategy.
Collapse
Affiliation(s)
- Germana Castelli
- Department of Hematology, Oncology & Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology & Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Ugo Testa
- Department of Hematology, Oncology & Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| |
Collapse
|
88
|
He XF, Wang QR, Cen JN, Qiu HY, Sun AN, Chen SN, Wu DP. [EVI1 expression, clinical and cytogenetical characteristics in 447 patients with acute myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 37:936-941. [PMID: 27995876 PMCID: PMC7348514 DOI: 10.3760/cma.j.issn.0253-2727.2016.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Indexed: 02/05/2023]
Abstract
Objective: To investigate EVI1 expression and its associated clinical and cytogenetic characteristics in 447 acute myeloid leukemia (AML) patients. Methods: EVI1 expressions were measured in 447 AML cases from Jan. 2007 to Apr. 2015 to couple with clinical, cytogenetic and mutations' characteristics to summarize the features of AMLs with high EVI1 expression. Results: 17.9% of AML were high EVI1 expression (EVI1 +), and the remainder low EVI1 expression (EVI1-). No significant differences between the two groups in terms of age, sex, hemoglobin level, white blood cell count and platelet count were observed. More M0, M5 and M6 subtypes were observed in EVI1+ group (P= 0.027, 0.004 and 0.011, respectively). Cytogenetic abnormalities of 11q15, 11q23/MLL, 3q26, -7/7q- and t (9;11) were observed more frequently in EVI1 + group (P<0.001, <0.001, <0.001, <0.001, =0.014, respectively). Normal karyotype, inv (16), t (8;21) were observed more frequent in EVI1- group (P=0.001, 0.009, 0.002, respectively). EVI1 + was more observed in high risk cytogenetics. Mutation of NPM1 was more observed in EVI1- group (P <0.001). Remission rate in EVI1 + group was significantly lower than EVI1- group (P<0.001). Leukemia-free survival was improved in EVI1 + AML patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). Conclusions: High EVI1 expression was more observed in FAB subgroup M5, harbored more cytogenetic abnormalities of 11p15, 11q23/MLL, 3q26 rearrangement, -7/7q- and t (9;11). Remission rate of high EVI1 expression AML was lower, which could be improved by allo-HSCT.
Collapse
Affiliation(s)
- X F He
- Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou 215006, China
| | | | | | | | | | | | | |
Collapse
|
89
|
Zheng Z, Li X, Zhu Y, Gu W, Xie X, Jiang J. Prognostic nomogram for previously untreated adult patients with acute myeloid leukemia. Oncotarget 2016; 7:71526-71535. [PMID: 27689396 PMCID: PMC5342098 DOI: 10.18632/oncotarget.12245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/19/2016] [Indexed: 12/26/2022] Open
Abstract
This study was designed to perform an acceptable prognostic nomogram for acute myeloid leukemia. The clinical data from 311 patients from our institution and 165 patients generated with Cancer Genome Atlas Research Network were reviewed. A prognostic nomogram was designed according to the Cox's proportional hazard model to predict overall survival (OS). To compare the capacity of the nomogram with that of the current prognostic system, the concordance index (C-index) was used to validate the accuracy as well as the calibration curve. The nomogram included 6 valuable variables: age, risk stratifications based on cytogenetic abnormalities, status of FLT3-ITD mutation, status of NPM1 mutation, expression of CD34, and expression of HLA-DR. The C-indexes were 0.71 and 0.68 in the primary and validation cohort respectively, which were superior to the predictive capacity of the current prognostic systems in both cohorts. The nomogram allowed both patients with acute myeloid leukemia and physicians to make prediction of OS individually prior to treatment.
Collapse
Affiliation(s)
- Zhuojun Zheng
- Department of Hematology, The Third Affiliated Hospital of Soochow University, China.,Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, China.,Cancer Immunotherapy Engineering Research Center of Jiangsu Province, China.,Institute of Cell Therapy, Soochow University, China
| | - Xiaodong Li
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, China.,Cancer Immunotherapy Engineering Research Center of Jiangsu Province, China.,Institute of Cell Therapy, Soochow University, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, China
| | - Yuandong Zhu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, China
| | - Weiying Gu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, China
| | - Xiaobao Xie
- Department of Hematology, The Third Affiliated Hospital of Soochow University, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, China.,Cancer Immunotherapy Engineering Research Center of Jiangsu Province, China.,Institute of Cell Therapy, Soochow University, China
| |
Collapse
|
90
|
Imataki O, Uemura M. Chromosomal abnormality of acute promyelocytic leukemia other than PML- RARA: a case report of acute promyelocytic leukemia with del(5q). BMC Clin Pathol 2016; 16:16. [PMID: 27708545 PMCID: PMC5050668 DOI: 10.1186/s12907-016-0038-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The recent study described a better outcome in acute promyelocytic leukemia patients treated with all-trans retinoic acid and arsenic oxide compared to those treated with all-trans retinoic acid combined with conventional chemotherapy. The pivotal study indicated that favorable-risk acute promyelocytic leukemia patients can be cured without any cytotoxic chemotherapy. Even high-risk patients are treatable with cytotoxic agents. Acute promyelocytic leukemia does not develop only by the dedifferentiation caused by PML-RARA. A determined oncogene other than PML-RARA which promotes cell proliferation would be required. CASE PRESENTATION We recently treated a 30-year-old Japanese female who achieved molecular remission with only the administration of all-trans retinoic acid. The patient's leukemic clones concomitantly had a del(5q) aberrant chromosome with t(15;17) (q22;q12). The patient's bone marrow cells indicated clonal evolution of the tumor cells expressing CD13dim, CD33+, CD117+, and lacking HLA-DR, CD34 and CD11b. A fluorescence in situ hybridization analysis detected PML-RARA fusion genes in the patient's bone marrow specimens, leading to the diagnosis of acute promyelocytic leukemia. CONCLUSION A del(5q) is one of the characteristic chromosomal abnormalities observed in myelodysplastic syndrome. On the other hand, up to 40 % of acute promyelocytic leukemia cases are known to harbor the addition of a clonal cytogenetic abnormality. However, such a case acute promyelocytic leukemia with del(5q) would be rare, rather than myelodysplastic syndrome, consequently obtaining t(15;17). Which cytogenetic abnormalities, acute promyelocytic leukemia or myelodysplastic syndrome, came first is informative to make a clinical decision for the initial therapy. In this case, we speculated the PML-RARA translocation is an original pathogenesis and thereafter additional cytogenetic abnormalities (del(5q) and -6) common in myelodysplastic syndrome. All-trans retinoic acid lead the patient into molecular remission. We propose that an assessment of additional cytogenetic abnormality in acute promyelocytic leukemia would contribute to the clinical decisions regarding whether to treat disease with all-trans retinoic acid and cytotoxic agents. It would be of interest to know the extent of cytogenetic abnormality in the patients regarding to mixed leukemia. One or more additional cytogenetic abnormalities other than PML-RARA could account for the biological malignant grade and prognostic index.
Collapse
Affiliation(s)
- Osamu Imataki
- Division of Hematology and Stem Cell Transplantation, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793 Japan
| | - Makiko Uemura
- Division of Hematology and Stem Cell Transplantation, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793 Japan
| |
Collapse
|
91
|
Ossenkoppele GJ, Janssen JJWM, van de Loosdrecht AA. Risk factors for relapse after allogeneic transplantation in acute myeloid leukemia. Haematologica 2016; 101:20-5. [PMID: 26721801 DOI: 10.3324/haematol.2015.139105] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acute myeloid leukemia is a clonal neoplasm derived from myeloid progenitor cells with a varying outcome. The initial goal of treatment is the achievement of complete remission, defined for over 40 years by morphology. However, without additional post-remission treatment the majority of patients relapse. In many cases of acute myeloid leukemia, allogeneic stem cell transplantation offers the best prospects of cure. In 2013, 5608 stem cell transplantations in acute myeloid leukemia were performed in Europe (5228 allogeneic and 380 autologous stem cell transplantations). Most stem cell transplantations are performed in first complete remission. However, despite a considerable reduction in the chance of relapse, in most studies, overall survival benefit of allogeneic stem cell transplantation is modest due to substantial non-relapse mortality. Here we discuss the many factors related to the risk of relapse after allogeneic stem cell transplantation.
Collapse
|
92
|
Stengel A, Kern W, Haferlach T, Meggendorfer M, Fasan A, Haferlach C. The impact of TP53 mutations and TP53 deletions on survival varies between AML, ALL, MDS and CLL: an analysis of 3307 cases. Leukemia 2016; 31:705-711. [PMID: 27680515 DOI: 10.1038/leu.2016.263] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/13/2016] [Accepted: 07/19/2016] [Indexed: 12/16/2022]
Abstract
Alterations in TP53 have been described in many cancer types including hematological neoplasms. We aimed at comparing TP53 mutations (mut) and deletions (del) in a large cohort of patients with hematological malignancies (n=3307), including AML (n=858), MDS (n=943), ALL (n=358), CLL (n=1148). Overall, alterations in TP53 were detected in 332/3307 cases (10%). The highest frequency was observed in ALL (total: 19%; mut+del: 6%; mut only: 8%; del only: 5%) and AML (total: 13%; mut+del: 5%; mut only: 7%; del only: 1%), whereas TP53 alterations occurred less frequently in CLL (total: 8%) and MDS (total: 7%). TP53 mutations were significantly more frequent in patients ⩾60 vs <60 years in AML (9% vs 2%, P<0.001) and ALL (12% vs 6%, P<0.001). TP53mut+del had a significant negative impact on overall survival in all entities, whereas differences were observed regarding TP53mut only or TP53del only: TP53mut only impacted survival in AML (36 vs 9 months, P<0.001) and MDS (65 vs 19 months, P<0.001), TP53del only in CLL (not reached vs 64 months, P=0.008) and MDS (65 vs 24 months, P=0.011). As substantial differences between the entities are observed regarding correlation to age and survival, we suggest evaluation of both TP53 deletion and mutation status.
Collapse
Affiliation(s)
- A Stengel
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - W Kern
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - T Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| | | | - A Fasan
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - C Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| |
Collapse
|
93
|
Creutzig U, Zimmermann M, Reinhardt D, Rasche M, von Neuhoff C, Alpermann T, Dworzak M, Perglerová K, Zemanova Z, Tchinda J, Bradtke J, Thiede C, Haferlach C. Changes in cytogenetics and molecular genetics in acute myeloid leukemia from childhood to adult age groups. Cancer 2016; 122:3821-3830. [DOI: 10.1002/cncr.30220] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/09/2016] [Accepted: 06/30/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Ursula Creutzig
- Pediatric Hematology and Oncology, Hannover Medical School; Hannover Germany
| | - Martin Zimmermann
- Pediatric Hematology and Oncology, Hannover Medical School; Hannover Germany
| | - Dirk Reinhardt
- Department of Pediatric Hematology-Oncology; University of Duisburg-Essen; Essen Germany
| | - Mareike Rasche
- Department of Pediatric Hematology-Oncology; University of Duisburg-Essen; Essen Germany
| | - Christine von Neuhoff
- Department of Pediatric Hematology-Oncology; University of Duisburg-Essen; Essen Germany
| | | | - Michael Dworzak
- Department of Pediatrics, St. Anna Children's Hospital and Children's Cancer Research Institute; Medical University of Vienna; Vienna Austria
| | | | - Zuzana Zemanova
- Center of Oncocytogenetics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine; Charles University; Prague Czech Republic
| | - Joelle Tchinda
- Pediatric Hematology/Oncology; University of Zurich; Zurich Switzerland
| | - Jutta Bradtke
- Institute for Pathology; University Hospital of Giessen and Marburg; Giessen Germany
| | - Christian Thiede
- Medical Clinic and Policlinic I, Carl Gustav Carus University Hospital; Technical University; Dresden Germany
| | | |
Collapse
|
94
|
Metzeler KH, Herold T, Rothenberg-Thurley M, Amler S, Sauerland MC, Görlich D, Schneider S, Konstandin NP, Dufour A, Bräundl K, Ksienzyk B, Zellmeier E, Hartmann L, Greif PA, Fiegl M, Subklewe M, Bohlander SK, Krug U, Faldum A, Berdel WE, Wörmann B, Büchner T, Hiddemann W, Braess J, Spiekermann K. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood 2016; 128:686-98. [PMID: 27288520 DOI: 10.1182/blood-2016-01-693879] [Citation(s) in RCA: 396] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/03/2016] [Indexed: 12/14/2022] Open
Abstract
The clinical and prognostic relevance of many recently identified driver gene mutations in adult acute myeloid leukemia (AML) is poorly defined. We sequenced the coding regions or hotspot areas of 68 recurrently mutated genes in a cohort of 664 patients aged 18 to 86 years treated on 2 phase 3 trials of the German AML Cooperative Group (AMLCG). The median number of 4 mutations per patient varied according to cytogenetic subgroup, age, and history of previous hematologic disorder or antineoplastic therapy. We found patterns of significantly comutated driver genes suggesting functional synergism. Conversely, we identified 8 virtually nonoverlapping patient subgroups, jointly comprising 78% of AML patients, that are defined by mutually exclusive genetic alterations. These subgroups, likely representing distinct underlying pathways of leukemogenesis, show widely divergent outcomes. Furthermore, we provide detailed information on associations between gene mutations, clinical patient characteristics, and therapeutic outcomes in this large cohort of uniformly treated AML patients. In multivariate analyses including a comprehensive set of molecular and clinical variables, we identified DNMT3A and RUNX1 mutations as important predictors of shorter overall survival (OS) in AML patients <60 years, and particularly in those with intermediate-risk cytogenetics. NPM1 mutations in the absence of FLT3-ITD, mutated TP53, and biallelic CEBPA mutations were identified as important molecular prognosticators of OS irrespective of patient age. In summary, our study provides a comprehensive overview of the spectrum, clinical associations, and prognostic relevance of recurrent driver gene mutations in a large cohort representing a broad spectrum and age range of intensively treated AML patients.
Collapse
Affiliation(s)
- Klaus H Metzeler
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Herold
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maja Rothenberg-Thurley
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany
| | - Susanne Amler
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Maria C Sauerland
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Dennis Görlich
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Stephanie Schneider
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany
| | - Nikola P Konstandin
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany
| | - Annika Dufour
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany
| | - Kathrin Bräundl
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bianka Ksienzyk
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany
| | - Evelyn Zellmeier
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany
| | - Luise Hartmann
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philipp A Greif
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Fiegl
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany
| | - Marion Subklewe
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan K Bohlander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Utz Krug
- Department of Internal Medicine 3, Klinikum Leverkusen, Leverkusen, Germany
| | - Andreas Faldum
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, Hematology and Oncology, University of Münster, Münster, Germany
| | - Bernhard Wörmann
- Department of Medicine, Hematology, Oncology, Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - Thomas Büchner
- Department of Medicine A, Hematology and Oncology, University of Münster, Münster, Germany
| | - Wolfgang Hiddemann
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Braess
- Department of Oncology and Hematology, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Karsten Spiekermann
- Laboratory for Leukemia Diagnostics, Department of Internal Medicine III, Ludwig-Maximilans-Universität, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
95
|
De Kouchkovsky I, Abdul-Hay M. 'Acute myeloid leukemia: a comprehensive review and 2016 update'. Blood Cancer J 2016; 6:e441. [PMID: 27367478 PMCID: PMC5030376 DOI: 10.1038/bcj.2016.50] [Citation(s) in RCA: 881] [Impact Index Per Article: 97.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/03/2016] [Accepted: 05/19/2016] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults, with an incidence of over 20 000 cases per year in the United States alone. Large chromosomal translocations as well as mutations in the genes involved in hematopoietic proliferation and differentiation result in the accumulation of poorly differentiated myeloid cells. AML is a highly heterogeneous disease; although cases can be stratified into favorable, intermediate and adverse-risk groups based on their cytogenetic profile, prognosis within these categories varies widely. The identification of recurrent genetic mutations, such as FLT3-ITD, NMP1 and CEBPA, has helped refine individual prognosis and guide management. Despite advances in supportive care, the backbone of therapy remains a combination of cytarabine- and anthracycline-based regimens with allogeneic stem cell transplantation for eligible candidates. Elderly patients are often unable to tolerate such regimens, and carry a particularly poor prognosis. Here, we review the major recent advances in the treatment of AML.
Collapse
Affiliation(s)
- I De Kouchkovsky
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - M Abdul-Hay
- Department of Medicine, New York University School of Medicine, New York, NY, USA.,Department of Hematology/Oncology, New York University Perlmutter Cancer Center, New York, NY, USA
| |
Collapse
|
96
|
Austin R, Smyth MJ, Lane SW. Harnessing the immune system in acute myeloid leukaemia. Crit Rev Oncol Hematol 2016; 103:62-77. [PMID: 27247119 DOI: 10.1016/j.critrevonc.2016.04.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/13/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022] Open
|
97
|
Prevalence and Clinical Significance of FLT3 Mutation Status in Acute Myeloid Leukemia Patients: A Multicenter Study. Arch Med Res 2016; 47:172-9. [PMID: 27373815 DOI: 10.1016/j.arcmed.2016.06.003] [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] [Received: 11/07/2015] [Accepted: 06/08/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS FLT3-ITD mutations in acute myeloid leukemia (AML) are associated with a poor prognosis. In Latin America, little epidemiological data exist about these mutations and their influence on clinical evolution and prognosis. Standardization and well-established clinical correlation make FLT3 mutational analysis by molecular methods an invaluable tool to decide among treatment options and to determine AML prognosis. METHODS We assessed the prevalence of FLT3-ITD mutations in 138 patients with AML at four hematology referral centers from Mexico and Colombia. Molecular methods based on polymerase chain reaction (PCR) were employed for determining FLT3-ITD status. RESULTS Mutations were present in 28 patients indicating a prevalence of 20.28%. Median age was 47 years (5-96). The FLT3 mutation positive group was older, had higher WBC and hemoglobin values and lower platelet counts but without statistical significance. A not previously described mutation in the FLT3 gene was found in one patient involving a nucleotide exchange of timine for cytosine at the 66608 position. A high mortality was found in the FLT3-mutated group, 67.8 vs. 42.72% in the non-mutated group and median survival was 4.9 months vs. 20.4 months, p = 0.009. A mutated FLT3 did not confer poor prognosis to those with M3 AML. The mutated FLT3 population had poor overall survival (OS) despite hematoprogenitor stem cell transplantation (HSCT). CONCLUSION Prevalence of FLT3-ITD mutation in AML was present in a proportion comparable to other populations and, when present, was associated with a very poor prognosis.
Collapse
|
98
|
Kojima K, Ishizawa J, Andreeff M. Pharmacological activation of wild-type p53 in the therapy of leukemia. Exp Hematol 2016; 44:791-798. [PMID: 27327543 DOI: 10.1016/j.exphem.2016.05.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
Abstract
The tumor suppressor p53 is inactivated by mutations in the majority of human solid tumors. Conversely, p53 mutations are rare in leukemias and are only observed in a small fraction of the patient population, predominately in patients with complex karyotype acute myeloid leukemia or hypodiploid acute lymphoblastic leukemia. However, the loss of p53 function in leukemic cells is often caused by abnormalities in p53-regulatory proteins, including overexpression of MDM2/MDMX, deletion of CDKN2A/ARF, and alterations in ATM. For example, MDM2 inhibits p53-mediated transcription, promotes its nuclear export, and induces proteasome-dependent degradation. The MDM2 homolog MDMX is another direct regulator of p53 that inhibits p53-mediated transcription. Several small-molecule inhibitors and stapled peptides targeting MDM2 and MDMX have been developed and have recently entered clinical trials. The clinical trial results of the first clinically used MDM2 inhibitor, RG7112, illustrated promising p53 activation and apoptosis induction in leukemia cells as proof of concept. Side effects of RG7112 were most prominent in suppression of thrombopoiesis and gastrointestinal symptoms in leukemia patients. Predictive biomarkers for response to MDM2 inhibitors have been proposed, but they require further validation both in vitro and in vivo so that the accumulated knowledge concerning pathological p53 dysregulation in leukemia and novel molecular-targeted strategies to overcome this dysregulation can be translated safely and efficiently into novel clinical therapeutics.
Collapse
Affiliation(s)
- Kensuke Kojima
- Department of Hematology, Respiratory Medicine and Oncology, Division of Medicine, Saga University, Saga, Japan; Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
99
|
Rose D, Haferlach T, Schnittger S, Perglerová K, Kern W, Haferlach C. Subtype-specific patterns of molecular mutations in acute myeloid leukemia. Leukemia 2016; 31:11-17. [PMID: 27285584 DOI: 10.1038/leu.2016.163] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/11/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) can be grouped into morphologically or genetically defined subtypes. Today, the AML phenotype-genotype associations, that is, FAB/WHO (French-American-British/World Health Organization) definitions and recurrent molecular mutations, are not fully understood. Therefore, we evaluated the impact of molecular mutations on the AML differentiation stage by molecular profiling of 4373 adult de novo AML patients in 7 cytomorphological subtypes. We investigated mutations in 20 genes, including myeloid transcription factors (CEBPA, RUNX1), tumor suppressors (TP53, WT1), DNA modifiers (DNMT3A, IDH1/2, TET2), chromatin modifiers (ASXL1, MLL), signal transduction genes (FLT3, KRAS, NRAS) and NPM1. The most frequently mutated genes per cytomorphological subtype were RUNX1 in M0 (43%), NPM1 in M1 (42%), DNMT3A in M2 (26%), NPM1 in M4 (57%), M5a (49%) and M5b (70%) and TP53 in M6 (36%). Although some gene mutations were frequent in several cytomorphological subtypes, a series of associations of co-occurring mutations with distinct phenotypes were identified for molecularly defined subcohorts. FLT3, NPM1 and WT1 mutations were associated with an immature phenotype in myeloblastic AML, whereas other combinations involving ASXL1, RUNX1, MLL-PTD, CEBPA or KRAS were more frequent in myeloblastic AML with maturation. Within the NPM1 mutated subcohort, ASXL1 mutations were significantly associated with a monoblastic differentiation and DNMT3A mutations with a monocytic phenotype.
Collapse
Affiliation(s)
- D Rose
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - T Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| | | | | | - W Kern
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - C Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| |
Collapse
|
100
|
Role of ASXL1 and TP53 mutations in the molecular classification and prognosis of acute myeloid leukemias with myelodysplasia-related changes. Oncotarget 2016; 6:8388-96. [PMID: 25860933 PMCID: PMC4480760 DOI: 10.18632/oncotarget.3460] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/20/2015] [Indexed: 11/28/2022] Open
Abstract
Acute myeloid leukemias (AML) with myelodysplasia-related changes (AML-MRC) are defined by the presence of multilineage dysplasia (MLD), and/or myelodysplastic syndrome (MDS)-related cytogenetics, and/or previous MDS. The goal of this study was to identify distinct biological and prognostic subgroups based on mutations of ASXL1, RUNX1, DNMT3A, NPM1, FLT3 and TP53 in 125 AML-MRC patients according to the presence of MLD, cytogenetics and outcome. ASXL1 mutations (n=26, 21%) were associated with a higher proportion of marrow dysgranulopoiesis (mutant vs. wild-type: 75% vs. 55%, p=0.030) and were mostly found in intermediate cytogenetic AML (23/26) in which they predicted inferior 2-year overall survival (OS, mutant vs. wild-type: 14% vs. 37%, p=0.030). TP53 mutations (n=28, 22%) were mostly found in complex karyotype AML (26/28) and predicted poor outcome within unfavorable cytogenetic risk AML (mutant vs. wild-type: 9% vs. 40%, p=0.040). In multivariate analysis, the presence of either ASXL1 or TP53 mutation was the only independent factor associated with shorter OS (HR, 95%CI: 2.53, 1.40-4.60, p=0.002) while MLD, MDS-related cytogenetics and previous MDS history did not influence OS. We conclude that ASXL1 and TP53 mutations identify two molecular subgroups among AML-MRCs, with specific poor prognosis. This could be useful for future diagnostic and prognostic classifications.
Collapse
|