1
|
Potter N, Jovanovic J, Ivey A, Othman J, Thomas A, Gilkes A, Runglall M, Kanda A, Thomas I, Johnson S, Canham J, Villiers W, Knapper S, Khwaja A, McMullin MF, Cavenagh J, Overgaard UM, Clark RE, Solomon E, Freeman SD, Hills R, Burnett A, Russell N, Dillon R. Molecular monitoring versus standard clinical care in younger adults with acute myeloid leukaemia: results from the UK NCRI AML17 and AML19 randomised, controlled, phase 3 trials. Lancet Haematol 2025; 12:e346-e356. [PMID: 40306832 DOI: 10.1016/s2352-3026(25)00037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND In patients with acute myeloid leukaemia treated with curative intent, the detection of measurable residual disease (MRD) generally confers a poor prognosis. This study aimed to identify whether altering treatment based on MRD results can improve survival. METHODS In the UK NCRI AML17 and AML19 randomised, controlled, phase 3 trials, performed in the UK, Denmark, and New Zealand, we screened patients aged 16-60 years with newly diagnosed acute myeloid leukaemia for molecular markers suitable for disease monitoring, including NPM1 mutations and fusion genes. Patients with a marker were randomly assigned (2:1) to either sequential molecular MRD monitoring during treatment and for 3 years after, or standard clinical care only with no molecular monitoring. In the monitoring group, treating physicians decided whether and how to incorporate the MRD results into the patient's therapy, including in cases of MRD relapse. The primary endpoint was overall survival. Prespecified subgroup analysis of the primary outcome included analysis by molecular group (NPM1mut with FLT3-ITD, NPM1mut without FLT3-ITD, and fusion gene transcripts). Both trials were registered with ISRCTN, ISRCTN55675535 and ISRCTN78449203, and are completed. FINDINGS In the AML17 trial, 1836 patients were enrolled between June 1, 2012 and Dec 31, 2014. In the AML19 trial, 965 patients were enrolled between Nov 9, 2015, and Jan 23, 2018. 637 patients were randomly assigned across both trials (289 to MRD monitoring and 144 to no monitoring in AML17 and 136 to MRD monitoring and 68 to no monitoring in AML19). With a median follow-up time of 4·9 years (IQR 3·6-5·9), overall survival at 3 years was 70% (95% CI 66-75) in patients in the monitoring group and 73% (68-80) in patients in the no-monitoring group. Meta analysis of the two studies showed no difference in overall survival (hazard ratio [HR] 1·11, 95% CI 0·83-1·49; p=0·25). In the pre-specified subgroup analysis of the primary endpoint, overall survival at 3 years in patients with both NPM1 and FLT3 internal tandem duplication (ITD) mutations was 69% (95% CI 60-79) in the monitoring group and 58% (45-74) in the no-monitoring group (HR 0·53, 95% CI 0·31-0·91; p=0·021). However there was no difference in survival by randomisation in patients with NPM1 mutations without FLT3-ITD (overall survial 69% [95% CI 62-77] in the monitoring group and 78% [70-87] in the no monitoring group; HR 1·56, 95% CI 0·96-2·52) or those with fusion gene transcripts (overall survial 72% [95% CI 65-79] in the monitoring group and 77% [68-87] in the no monitoring group; HR 1·28, 95% CI 0·80-2·18). INTERPRETATION Sequential molecular MRD monitoring, coupled with MRD-guided treatment, did not improve overall survival in the entire study population; however, in the subgroup of patients with baseline NPM1 and FLT3 ITD mutations, we observed a survival benefit for MRD monitoring. FUNDING National Institute for Health Research, Blood Cancer UK, and Cancer Research UK.
Collapse
Affiliation(s)
- Nicola Potter
- Department of Medical and Molecular Genetics, King's College, London, UK
| | - Jelena Jovanovic
- Department of Medical and Molecular Genetics, King's College, London, UK
| | - Adam Ivey
- Department of Pathology, Alfred Hospital, Melbourne, VIC, Australia
| | - Jad Othman
- Department of Medical and Molecular Genetics, King's College, London, UK
| | - Abin Thomas
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | | | | | - Anju Kanda
- Department of Medical and Molecular Genetics, King's College, London, UK
| | - Ian Thomas
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Sean Johnson
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Joanna Canham
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - William Villiers
- Department of Medical and Molecular Genetics, King's College, London, UK
| | | | - Asim Khwaja
- Department of Haematology, University College London Hospital, London, UK
| | | | | | | | - Richard E Clark
- Molecular & Clinical Cancer Medicine, University of Liverpool, UK
| | - Ellen Solomon
- Department of Medical and Molecular Genetics, King's College, London, UK
| | - Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, UK
| | - Robert Hills
- Nuffield Department of Population Health, University of Oxford, UK
| | - Alan Burnett
- Department of Haematology, University of Glasgow, UK
| | - Nigel Russell
- Department of Haematology, University of Nottingham, UK; Department of Haematology, Guy's Hospital, London, UK
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, UK; Department of Haematology, Guy's Hospital, London, UK.
| |
Collapse
|
2
|
Loo S, Potter N, Ivey A, O'Nions J, Moon R, Jovanovic J, Fong CY, Anstee NS, Tiong IS, Othman J, Chua CC, Renshaw H, Baker R, Fleming S, Russell NH, Ritchie D, Bajel A, Hou HA, Dillon R, Wei AH. Pretransplant MRD detection of fusion transcripts is strongly prognostic in KMT2A-rearranged acute myeloid leukemia. Blood 2024; 144:2554-2557. [PMID: 39316646 DOI: 10.1182/blood.2024026605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
ABSTRACT Pretransplant detection of KMT2Ar measurable residual disease ≥0.001% by quantitative polymerase chain reaction was associated with significantly inferior posttransplant survival (2-year relapse-free survival 17% vs 59%; P = .001) and increased 2-year cumulative incidence of relapse (75% vs 25%, P = .0004).
Collapse
Affiliation(s)
- Sun Loo
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Nicola Potter
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - Adam Ivey
- Molecular Pathology, The Alfred Hospital, Melbourne, Australia
| | - Jenny O'Nions
- Department of Haematology, University College London NHS Foundation Trust, London, United Kingdom
- Specialist Integrated Haematology Malignancy Diagnostic Service, Health Services Laboratories, University College London Hospitals NHS Foundation Trust, University College London, London, United Kingdom
| | - Roderick Moon
- Molecular Pathology, The Alfred Hospital, Melbourne, Australia
| | - Jelena Jovanovic
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - Chun Y Fong
- Department of Haematology, Austin Hospital, Melbourne, Australia
| | - Natasha S Anstee
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Ing Soo Tiong
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Jad Othman
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Chong Chyn Chua
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Department of Haematology, Monash Health, Clayton, Australia
| | - Hanna Renshaw
- Department of Haematology, University College London NHS Foundation Trust, London, United Kingdom
| | - Robert Baker
- Department of Haematology, University College London NHS Foundation Trust, London, United Kingdom
- Specialist Integrated Haematology Malignancy Diagnostic Service, Health Services Laboratories, University College London Hospitals NHS Foundation Trust, University College London, London, United Kingdom
| | - Shaun Fleming
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, Australia
| | - Nigel H Russell
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - David Ritchie
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Ashish Bajel
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Hsin-An Hou
- Division of Hematology and Division of General Medicine, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Andrew H Wei
- Department of Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
3
|
Dillon R, Potter N, Freeman S, Russell N. How we use molecular minimal residual disease (MRD) testing in acute myeloid leukaemia (AML). Br J Haematol 2021; 193:231-244. [PMID: 33058194 DOI: 10.1111/bjh.17185] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years there have been major advances in the use of molecular diagnostic and monitoring techniques for patients with acute myeloid leukaemia (AML). Coupled with the simultaneous explosion of new therapeutic agents, this has sown the seeds for significant improvements to treatment algorithms. Here we show, using a selection of real-life examples, how molecular monitoring can be used to refine clinical decision-making and to personalise treatment in patients with AML with nucleophosmin (NPM1) mutations, core binding factor translocations and other fusion genes. For each case we review the established evidence base and provide practical recommendations where evidence is lacking or conflicting. Finally, we review important technical considerations that clinicians should be aware of in order to safely exploit these technologies as they undergo widespread implementation.
Collapse
Affiliation(s)
- Richard Dillon
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College, London, UK
- Department of Haematology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| | - Nicola Potter
- Cancer Genetics Laboratory, Department of Medical and Molecular Genetics, King's College, London, UK
| | - Sylvie Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Nigel Russell
- Department of Haematology, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
4
|
Dwivedi N, Mondal S, P. K. S, T. S, Sachdeva K, Bathula C, K. V, K. S. N, Damodar S, Dhar SK, Das M. Relative quantification of BCL2 mRNA for diagnostic usage needs stable uncontrolled genes as reference. PLoS One 2020; 15:e0236338. [PMID: 32785215 PMCID: PMC7423076 DOI: 10.1371/journal.pone.0236338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/02/2020] [Indexed: 01/21/2023] Open
Abstract
Dysregulation of BCL2 is a pathophysiology observed in haematological malignancies. For implementation of available treatment-options it is preferred to know the relative quantification of BCL2 mRNA with appropriate reference genes. For the choice of reference genes-(i) Reference Genes were selected by assessing variation of >60,000 genes from 4 RNA-seq datasets of haematological malignancies followed by filtering based on their GO biological process annotations and proximity of their chromosomal locations to known disease translocations. Selected genes were experimentally validated across various haematological malignancy samples followed by stability comparison using geNorm, NormFinder, BestKeeper and RefFinder. (ii) 43 commonly used Reference Genes were obtained from literature through extensive systematic review. Levels of BCL2 mRNA was assessed by qPCR normalized either by novel reference genes from this study or GAPDH, the most cited reference gene in literature and compared. The analysis showed PTCD2, PPP1R3B and FBXW9 to be the most unregulated genes across lymph-nodes, bone marrow and PBMC samples unlike the Reference Genes used in literature. BCL2 mRNA level shows a consistent higher expression in haematological malignancy patients when normalized by these novel Reference Genes as opposed to GAPDH, the most cited Reference Gene. These reference genes should also be applicable in qPCR platforms using Taqman probes and other model systems including cell lines and rodent models. Absence of sample from healthy-normal individual in diagnostic cases call for careful selection of Reference Genes for relative quantification of a biomarker by qPCR.BCL2 can be used as molecular diagnostics only if normalized with a set of reference genes with stable yet low levels of expression across different types of haematological malignancies.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/isolation & purification
- Bone Marrow/pathology
- Cell Line, Tumor
- Datasets as Topic
- Disease Models, Animal
- Feasibility Studies
- Gene Expression Regulation, Neoplastic
- Genes, Essential
- Hematologic Neoplasms/blood
- Hematologic Neoplasms/diagnosis
- Hematologic Neoplasms/genetics
- Hematologic Neoplasms/pathology
- Humans
- Leukocytes, Mononuclear
- Proto-Oncogene Proteins c-bcl-2/blood
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/isolation & purification
- RNA, Messenger/blood
- RNA, Messenger/genetics
- RNA, Messenger/isolation & purification
- RNA-Seq/standards
- Real-Time Polymerase Chain Reaction/standards
- Reference Standards
Collapse
Affiliation(s)
- Nehanjali Dwivedi
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
- MAHE, Manipal, India
| | - Sreejeta Mondal
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Smitha P. K.
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Sowmya T.
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Kartik Sachdeva
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Christopher Bathula
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Vishnupriyan K.
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Nataraj K. S.
- Department of Haematology, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Sharat Damodar
- Department of Haematology, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Sujan K. Dhar
- Beyond Antibody, InCite Labs, MSMF, MSMC, Narayana Health City, Bangalore, India
| | - Manjula Das
- Tumor Immunology Program, MSMF, MSMC, Narayana Health City, Bangalore, India
- Beyond Antibody, InCite Labs, MSMF, MSMC, Narayana Health City, Bangalore, India
| |
Collapse
|
5
|
Pérez-Sáenz MA, Rodriguez-Pinilla SM, Salgado RN, Carvajal N, Serrano C, Soto C, Serrano J, Atance M, López-Lorenzo JL, Requena L, Rojo F, Piris MA, Llamas P, Alonso-Dominguez JM. Three monocytic neoplasms in a single patient. Leuk Lymphoma 2020; 61:2523-2526. [PMID: 32449424 DOI: 10.1080/10428194.2020.1761966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- María Angeles Pérez-Sáenz
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Rocío N Salgado
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Nerea Carvajal
- Pathology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Serrano
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Soto
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juana Serrano
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mireia Atance
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - José Luis López-Lorenzo
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Requena
- Dermatology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Federico Rojo
- Pathology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel Angel Piris
- Pathology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pilar Llamas
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Manuel Alonso-Dominguez
- Hematology Department, University Hospital Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
6
|
Minimal Residual Disease in Acute Myeloid Leukemia of Adults: Determination, Prognostic Impact and Clinical Applications. Mediterr J Hematol Infect Dis 2016; 8:e2016052. [PMID: 27872732 PMCID: PMC5111512 DOI: 10.4084/mjhid.2016.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 09/12/2016] [Indexed: 02/06/2023] Open
Abstract
Pretreatment assessment of cytogenetic/genetic signature of acute myeloid leukemia (AML) has been consistently shown to play a major prognostic role but also to fail at predicting outcome on individual basis, even in low-risk AML. Therefore, we are in need of further accurate methods to refine the patients’ risk allocation process, distinguishing more adequately those who are likely to recur from those who are not. In this view, there is now evidence that the submicroscopic amounts of leukemic cells (called minimal residual disease, MRD), measured during the course of treatment, indicate the quality of response to therapy. Therefore, MRD might serve as an independent, additional biomarker to help to identify patients at higher risk of relapse. Detection of MRD requires the use of highly sensitive ancillary techniques, such as polymerase chain reaction (PCR) and multiparametric flow cytometry(MPFC). In the present manuscript, we will review the current approaches to investigate MRD and its clinical applications in AML management.
Collapse
|
7
|
Hirsch P, Zhang Y, Tang R, Joulin V, Boutroux H, Pronier E, Moatti H, Flandrin P, Marzac C, Bories D, Fava F, Mokrani H, Betems A, Lorre F, Favier R, Féger F, Mohty M, Douay L, Legrand O, Bilhou-Nabera C, Louache F, Delhommeau F. Genetic hierarchy and temporal variegation in the clonal history of acute myeloid leukaemia. Nat Commun 2016; 7:12475. [PMID: 27534895 PMCID: PMC4992157 DOI: 10.1038/ncomms12475] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/05/2016] [Indexed: 12/21/2022] Open
Abstract
In acute myeloid leukaemia (AML) initiating pre-leukaemic lesions can be identified through three major hallmarks: their early occurrence in the clone, their persistence at relapse and their ability to initiate multilineage haematopoietic repopulation and leukaemia in vivo. Here we analyse the clonal composition of a series of AML through these characteristics. We find that not only DNMT3A mutations, but also TET2, ASXL1 mutations, core-binding factor and MLL translocations, as well as del(20q) mostly fulfil these criteria. When not eradicated by AML treatments, pre-leukaemic cells with these lesions can re-initiate the leukaemic process at various stages until relapse, with a time-dependent increase in clonal variegation. Based on the nature, order and association of lesions, we delineate recurrent genetic hierarchies of AML. Our data indicate that first lesions, variegation and treatment selection pressure govern the expansion and adaptive behaviour of the malignant clone, shaping AML in a time-dependent manner. Pre-leukaemic clones, together with the propensity to cause disease in mice, are characterized by appearing early in myeloid leukaemia and being found at relapse. Here, the authors identify clones in human samples and find that they are characterized by hierarchically organized genetic lesions, which can be used to track evolution of the disease.
Collapse
Affiliation(s)
- Pierre Hirsch
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France.,AP-HP, Hôpital St Antoine, Service d'Hématologie clinique et de thérapie cellulaire, F-75012 Paris, France
| | - Yanyan Zhang
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 1170, CNRS GDR 3697 Micronit, 94805 Villejuif, France.,Institut Gustave Roussy, Univ Paris-Sud, Université Paris Saclay, 94805 Villejuif, France
| | - Ruoping Tang
- AP-HP, Hôpital St Antoine, Service d'Hématologie clinique et de thérapie cellulaire, F-75012 Paris, France
| | - Virginie Joulin
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 1170, CNRS GDR 3697 Micronit, 94805 Villejuif, France.,Institut Gustave Roussy, Univ Paris-Sud, Université Paris Saclay, 94805 Villejuif, France
| | - Hélène Boutroux
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France.,Department of Pediatric Hematology and Oncology, AP-HP, Hôpital Armand-Trousseau, F-75012 Paris, France
| | - Elodie Pronier
- Institut Gustave Roussy, Univ Paris-Sud, Université Paris Saclay, 94805 Villejuif, France
| | - Hannah Moatti
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France
| | - Pascale Flandrin
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France
| | - Christophe Marzac
- AP-HP, Hôpital Saint-Antoine &Hôpital Armand-Trousseau, Service d'hématologie biologique, F-75012 Paris, France
| | - Dominique Bories
- AP-HP, Hôpital Henri Mondor, Unité d'Hématologie Moléculaire, F-94010 Créteil, France
| | - Fanny Fava
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France
| | - Hayat Mokrani
- Institut Gustave Roussy, Univ Paris-Sud, Université Paris Saclay, 94805 Villejuif, France
| | - Aline Betems
- Institut Gustave Roussy, Univ Paris-Sud, Université Paris Saclay, 94805 Villejuif, France
| | - Florence Lorre
- AP-HP, Hôpital Saint-Antoine, Laboratoire commun de biologie et génétique moléculaires, F-75012 Paris, France
| | - Rémi Favier
- AP-HP, Hôpital Saint-Antoine &Hôpital Armand-Trousseau, Service d'hématologie biologique, F-75012 Paris, France
| | - Frédéric Féger
- AP-HP, Hôpital Saint-Antoine &Hôpital Armand-Trousseau, Service d'hématologie biologique, F-75012 Paris, France
| | - Mohamad Mohty
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France
| | - Luc Douay
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine &Hôpital Armand-Trousseau, Service d'hématologie biologique, F-75012 Paris, France
| | - Ollivier Legrand
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France.,AP-HP, Hôpital St Antoine, Service d'Hématologie clinique et de thérapie cellulaire, F-75012 Paris, France
| | - Chrystèle Bilhou-Nabera
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine &Hôpital Armand-Trousseau, Service d'hématologie biologique, F-75012 Paris, France
| | - Fawzia Louache
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 1170, CNRS GDR 3697 Micronit, 94805 Villejuif, France.,Institut Gustave Roussy, Univ Paris-Sud, Université Paris Saclay, 94805 Villejuif, France
| | - François Delhommeau
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, F-75012 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, GRC n°7, Groupe de Recherche Clinique sur les Myéloproliférations Aiguës et Chroniques MYPAC, F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine &Hôpital Armand-Trousseau, Service d'hématologie biologique, F-75012 Paris, France
| |
Collapse
|
8
|
Kayser S, Walter RB, Stock W, Schlenk RF. Minimal residual disease in acute myeloid leukemia--current status and future perspectives. Curr Hematol Malig Rep 2016; 10:132-44. [PMID: 25994952 DOI: 10.1007/s11899-015-0260-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In acute myeloid leukemia (AML), the achievement of a morphological complete remission (CR) is an important milestone on the road to cure. Still, the majority of patients who achieve a morphological CR will eventually relapse. Thus, morphological means are not sensitive enough to detect clinically relevant tumor burdens left behind after therapy. Over the last years, several methodologies, particularly multiparameter flow cytometry and polymerase chain reaction, have emerged that can detect, quantify, and monitor submicroscopic amounts of leukemia cells ("minimal residual disease", MRD). Newer techniques, such as next-generation sequencing, have not only changed our understanding of the molecular pathogenesis and clonal heterogeneity of AML but may also be used for MRD detection. Increasing evidence indicates that MRD could play an important role in dynamically refining disease risk and, perhaps, serve to fine-tune post-remission therapy in a risk-adapted manner, although the latter concept awaits validation through well-controlled trials. In this review, we discuss the current use of MRD measurements during AML treatment and highlight future perspectives.
Collapse
Affiliation(s)
- Sabine Kayser
- Department of Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany,
| | | | | | | |
Collapse
|
9
|
Comparative analysis of KRAS codon 12, 13, 18, 61, and 117 mutations using human MCF10A isogenic cell lines. Sci Rep 2015; 5:8535. [PMID: 25705018 PMCID: PMC4336936 DOI: 10.1038/srep08535] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/23/2015] [Indexed: 12/30/2022] Open
Abstract
KRAS mutations occur in one third of human cancers and cluster in several hotspots, with codons 12 and 13 being most commonly affected. It has been suggested that the position and type of amino acid exchange influence the transforming capacity of mutant KRAS proteins. We used MCF10A human mammary epithelial cells to establish isogenic cell lines that express different cancer-associated KRAS mutations (G12C, G12D, G12V, G13C, G13D, A18D, Q61H, K117N) at physiological or elevated levels, and investigated the biochemical and functional consequences of the different variants. The overall effects of low-expressing mutants were moderate compared to overexpressed variants, but allowed delineation of biological functions that were related to specific alleles rather than KRAS expression level. None of the mutations induced morphological changes, migratory abilities, or increased phosphorylation of ERK, PDK1, and AKT. KRAS-G12D, G12V, G13D, and K117N mediated EGF-independent proliferation, whereas anchorage-independent growth was primarily induced by K117N and Q61H. Both codon 13 mutations were associated with increased EGFR expression. Finally, global gene expression analysis of MCF10A-G13D versus MCF10A-G12D revealed distinct transcriptional changes. Together, we describe a useful resource for investigating the function of multiple KRAS mutations and provide insights into the differential effects of these variants in MCF10A cells.
Collapse
|
10
|
Ommen HB, Hokland P, Haferlach T, Abildgaard L, Alpermann T, Haferlach C, Kern W, Schnittger S. Relapse kinetics in acute myeloid leukaemias with MLL translocations or partial tandem duplications within the MLL gene. Br J Haematol 2014; 165:618-28. [PMID: 24611505 DOI: 10.1111/bjh.12792] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/10/2014] [Indexed: 01/03/2023]
Abstract
Correct action upon re-emergence of minimal residual disease in acute myeloid leukaemia (AML) patients has not yet been established. The applicability of demethylating agents and use of allogeneic stem cell transplantation will be dependent on pre-relapse AML growth rates. We here delineate molecular growth kinetics of AML harbouring MLL partial tandem duplication (MLL-PTD; 37 cases) compared to those harbouring MLL translocations (43 cases). The kinetics of MLL-PTD relapses was both significantly slower than those of MLL translocation positive ones (median doubling time: MLL-PTD: 24 d, MLL-translocations: 12 d, P = 0·015, Wilcoxon rank sum test), and displayed greater variation depending on additional mutations. Thus, MLL-PTD+ cases with additional RUNX1 mutations or FLT3-internal tandem duplication relapsed significantly faster than cases without one of those two mutations (Wilcoxon rank sum test, P = 0·042). As rapid relapses occurred in all MLL subgroups, frequent sampling are necessary to obtain acceptable relapse detection rates and times from molecular relapse to haematological relapse (blood sampling every second month: MLL-PTD: 75%/50 d; MLL translocations: 85%/25 d). In conclusion, in this cohort relapse kinetics is heavily dependent on AML subtype as well as additional genetic aberrations, with possibly great consequences for the rational choice of pre-emptive therapies.
Collapse
Affiliation(s)
- Hans B Ommen
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Abildgaard L, Ommen HB, Lausen B, Hasle H, Nyvold CG. A novel RT-qPCR assay for quantification of the MLL-MLLT3 fusion transcript in acute myeloid leukaemia. Eur J Haematol 2013; 91:394-8. [PMID: 23772754 DOI: 10.1111/ejh.12156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2013] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Patients with acute myeloid leukaemia (AML) of the monocytic lineage often lack molecular markers for minimal residual disease (MRD) monitoring. The MLL-MLLT3 fusion transcript found in patients with AML harbouring t(9;11) is amenable to RT-qPCR quantification but because of the heterogeneity of translocation break points, the MLL-MLLT3 fusion gene is a challenging target. We hypothesised that MRD monitoring using MLL-MLLT3 as a RT-qPCR marker is feasible in the majority of patients with t(9;11)-positive AML. METHODS Using a locked nucleic acid probe, we developed a sensitive RT-qPCR assay for quantification of the most common break point region of the MLL-MLLT3 fusion gene. Five paediatric patients with t(9;11)-positive AML were monitored using the MLL-MLLT3 assay. RESULTS A total of 43 bone marrow (BM) and 52 Peripheral blood (PB) samples were collected from diagnosis until follow-up. Two patients relapsed, and both were MRD positive in BM after first induction course. A total of three relapses occurred, and they were detected by RT-qPCR 3 wks before haematological relapse was diagnosed. CONCLUSION This MLL-MLLT3 RT-qPCR assay could be useful in MRD monitoring of a group of patients with AML who often lack reliable MRD markers.
Collapse
Affiliation(s)
- Lotte Abildgaard
- Department of Paediatrics, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | |
Collapse
|
12
|
Sarid N, Eshel R, Rothman R, Shpringer M, Perry C, Solar I, Dezorella N, Gepstein L, Canaani J, Naparstek E, Polliack A, Herishanu Y. Acute myeloid leukemia with 11q23/MLL rearrangement after 'FCR' regimen for chronic lymphocytic leukemia. Eur J Haematol 2012; 89:430-1. [PMID: 22816611 DOI: 10.1111/ejh.12001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
MESH Headings
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Antibodies, Monoclonal, Murine-Derived/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Base Sequence
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 9/genetics
- Cyclophosphamide/administration & dosage
- Cyclophosphamide/adverse effects
- DNA Probes/genetics
- Gene Rearrangement
- Histone-Lysine N-Methyltransferase
- Humans
- In Situ Hybridization, Fluorescence
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/genetics
- Male
- Middle Aged
- Myeloid-Lymphoid Leukemia Protein/genetics
- Neoplasms, Second Primary/etiology
- Neoplasms, Second Primary/genetics
- Rituximab
- Translocation, Genetic
- Vidarabine/administration & dosage
- Vidarabine/adverse effects
- Vidarabine/analogs & derivatives
Collapse
|
13
|
Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML. Blood 2009; 114:2220-31. [PMID: 19587375 DOI: 10.1182/blood-2009-03-213389] [Citation(s) in RCA: 250] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nucleophosmin (NPM1)-mutated acute myeloid leukemia (AML), which is recognized as a provisional entity in the World Health Organization 2008 classification of myeloid neoplasms, accounts for 30% of AML. We analyzed 1227 diagnostic and follow-up samples in 252 NPM1-mutated AML patients with 17 different NPM1 mutation-specific real-time quantitative polymerase chain reaction (RQ-PCR) assays. Paired diagnostic/relapse samples of 84 patients revealed stable NPM1 mutations in all cases, suggesting that they are pathogenetically early events and thus applicable for minimal residual disease detection. A total of 47 relapses were predictable because of an NPM1 mutation level (%NPM1/ABL1) increase of at least 1 log or in 15 cases because of NPM1 mutation levels not decreasing less than 3 log ranges. A high prognostic value of NPM1 levels was shown for 4 different intervals after therapy was initiated. Furthermore, thresholds of 0.1 and 0.01%NPM1/ABL1 during/after treatment discriminated between prognostic subgroups. Univariate analyses, including age, white blood cell count, blast count, CD34 positivity, FLT3 mutations status, FAB type, karyotype, NPM1 mutation type, and pretreatment NPM1 mutational level, showed that, besides NPM1 mutation level, only age and FLT3-LM mutation status were prognostically significant for EFS. Multivariate analysis, including age, FLT3-LM status, and NPM1 mutation level at different time points, demonstrated that NPM1 level was the most relevant prognostic factor during first-line treatment. Similar results were obtained in patients undergoing second-line chemotherapy or allogeneic stem cell transplantation.
Collapse
|
14
|
Haribaskar R, Pütz M, Schupp B, Skouloudaki K, Bietenbeck A, Walz G, Schäfer T. The planar cell polarity (PCP) protein Diversin translocates to the nucleus to interact with the transcription factor AF9. Biochem Biophys Res Commun 2009; 387:212-7. [PMID: 19591803 DOI: 10.1016/j.bbrc.2009.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 07/02/2009] [Indexed: 01/19/2023]
Abstract
The planar cell polarity (PCP) pathway, a beta-catenin-independent branch of the Wnt signaling pathway, orients cells and their appendages with respect to the body axes. Diversin, the mammalian homolog of the Drosophila PCP protein Diego, acts as a molecular switch that blocks beta-catenin-dependent and promotes beta-catenin-independent Wnt signaling. We report now that Diversin, containing several nuclear localization signals, translocates to the nucleus, where it interacts with the transcription factor AF9. Both Diversin and AF9 block canonical Wnt signaling; however, this occurs independently of each other, and does not require nuclear Diversin. In contrast, AF9 strongly augments the Diversin-driven activation of c-Jun N-terminal kinase (JNK)-dependent gene expression in the nucleus, and this augmentation largely depends on the presence of nuclear Diversin. Thus, our findings reveal that components of the PCP cascade translocate to the nucleus to participate in transcriptional regulation and PCP signaling.
Collapse
|
15
|
Alonso CN, Longo PLR, Gallego MS, Medina A, Felice MS. A novel AF9 breakpoint in MLL-AF9-positive acute monoblastic leukemia. Pediatr Blood Cancer 2008; 50:869-71. [PMID: 18000862 DOI: 10.1002/pbc.21393] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
MLL-AF9 is the most frequent MLL rearrangement in childhood acute myeloid leukemia (AML) and it may be also found in acute lymphoblastic leukemia (ALL) of patients younger than 1-year-old (infants). We report a novel AF9 breakpoint site, located between previously reported sites A and B, detected in an infant who was diagnosed with AML-FAB M5. The occurrence of this new breakpoint should be considered when designing RT-PCR assays for the screening of MLL abnormalities. The precise characterization of the MLL-AF9 transcript is important to carry out the minimal residual disease analysis during the follow-up of the patients.
Collapse
Affiliation(s)
- Cristina N Alonso
- Molecular Biology Laboratory, Department of Hematology-Oncology, Hospital de Pediatría SAMIC "Prof. Dr. J. P. Garrahan", Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
16
|
Kern W, Haferlach C, Haferlach T, Schnittger S. Monitoring of minimal residual disease in acute myeloid leukemia. Cancer 2007; 112:4-16. [DOI: 10.1002/cncr.23128] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
17
|
Kern W, Schoch C, Haferlach T, Schnittger S. Monitoring of minimal residual disease in acute myeloid leukemia. Crit Rev Oncol Hematol 2005; 56:283-309. [PMID: 16213150 DOI: 10.1016/j.critrevonc.2004.06.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2004] [Revised: 06/24/2004] [Accepted: 06/24/2004] [Indexed: 11/17/2022] Open
Abstract
Monitoring minimal residual disease (MRD) becomes increasingly important in the risk-adapted management of patients with acute myeloid leukemia (AML). The two most sensitive and quantitative methods for MRD detection are multiparameter flow cytometry (MFC) and real-time polymerase chain reaction (QRT-PCR). Fusion gene-specific PCR in AML is based on the RNA level, and thus in contrast to MFC expression levels rather than cell counts are assessed. For both methods independent prognostic values have been shown. The strong power of MFC has been shown mainly in the assessment of early clearance of the malignant clone. MRD levels in AML with fusion genes have the strongest prognostic power after the end of consolidation therapy. In addition, with QRT-PCR highly predictive initial expression levels can be assessed. With both methods early detection of relapse is possible. So far, validated PCR-based MRD was done with fusion genes that are detectable in only 20-25% of all AML MFC is superior since it is applicable for most AML. However, QRT-PCR is still more sensitive in most cases. Thus, it is desirable to establish new molecular markers for PCR-based studies. Large clinical trials will determine the role and place of immunologic and PCR-based monitoring in the prognostic stratification of patients with AML.
Collapse
Affiliation(s)
- Wolfgang Kern
- Laboratory for Leukemia Diagnostics, Ludwig-Maximilians-University, University Hospital Grosshadern, Department of Internal Medicine III, 81366 Muenchen, Germany.
| | | | | | | |
Collapse
|
18
|
Jansen MWJC, van der Velden VHJ, van Dongen JJM. Efficient and easy detection of MLL-AF4, MLL-AF9 and MLL-ENL fusion gene transcripts by multiplex real-time quantitative RT-PCR in TaqMan and LightCycler. Leukemia 2005; 19:2016-8. [PMID: 16151462 DOI: 10.1038/sj.leu.2403939] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Bench AJ, Erber WN, Scott MA. Molecular genetic analysis of haematological malignancies: I. Acute leukaemias and myeloproliferative disorders. ACTA ACUST UNITED AC 2005; 27:148-71. [PMID: 15938721 DOI: 10.1111/j.1365-2257.2005.00701.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular genetic techniques are now routinely applied to haematological malignancies within a clinical laboratory setting. The detection of genetic rearrangements not only assists with diagnosis and treatment decisions, but also adds important prognostic information. In addition, genetic rearrangements associated with leukaemia can be used as molecular markers allowing the detection of low levels of residual disease. This review will concentrate on the application of molecular genetic techniques to the acute leukaemias and myeloprolferative disorders.
Collapse
Affiliation(s)
- A J Bench
- Haemato-Oncology Diagnostic Service, Department of Haematology, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK.
| | | | | |
Collapse
|
20
|
Maroc N, Morel A, Beillard E, De La Chapelle AL, Fund X, Mozziconacci MJ, Dupont M, Cayuela JM, Gabert J, Koki A, Fert V, Hermitte F. A diagnostic biochip for the comprehensive analysis of MLL translocations in acute leukemia. Leukemia 2004; 18:1522-30. [PMID: 15322560 DOI: 10.1038/sj.leu.2403439] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Reciprocal rearrangements of the MLL gene are among the most common chromosomal abnormalities in both Acute Lymphoblastic and Myeloid Leukemia. The MLL gene, located on the 11q23 chromosomal band, is involved in more than 40 recurrent translocations. In the present study, we describe the development and validation of a biochip-based assay designed to provide a comprehensive molecular analysis of MLL rearrangements when used in a standard clinical pathology laboratory. A retrospective blind study was run with cell lines (n=5), and MLL positive and negative patient samples (n=31), to evaluate assay performance. The limits of detection determined on cell line data were 10(-1), and the precision studies yielded 100% repeatability and 98% reproducibility. The study shows that the device can detect frequent (AF4, AF6, AF10, ELL or ENL) as well as rare partner genes (AF17, MSF). The identified fusion transcripts can then be used as molecular phenotypic markers of disease for the precise evaluation of minimal residual disease by RQ-PCR. This biochip-based molecular diagnostic tool allows, in a single experiment, rapid and accurate identification of MLL gene rearrangements among 32 different fusion gene (FG) partners, precise breakpoint positioning and comprehensive screening of all currently characterized MLL FGs.
Collapse
Affiliation(s)
- N Maroc
- IPSOGEN SAS, Case 923, 163, Av. de Luminy, Marseille Cedex 9, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Raanani P, Ben-Bassat I. Detection of minimal residual disease in acute myelogenous leukemia. Acta Haematol 2004; 112:40-54. [PMID: 15179004 DOI: 10.1159/000077559] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acute myelogenous leukemia (AML) is considered to be in complete remission when fewer than 5% of the cells in bone marrow are blasts. Nevertheless, approximately two thirds of patients relapse due to persisting leukemic blasts. The persistence of these cells, below the threshold of morphological detection, is termed minimal residual disease (MRD) and various methods are used for its detection. These methods include classical cytogenetics, fluorescence in situ hybridization, qualitative and quantitative RT-PCR and multiparametric flow cytometry. Currently, less than half of the AML patients have a specific marker detectable by RT-PCR techniques. The major specific molecular markers are involvement of the MLL gene with up to 50 different partners and partial tandem duplications, the core binding factor leukemias with AML1/ETO and CBFbeta/MYH11 rearrangements, PML/RARalpha in acute promyelocytic leukemia, internal tandem duplications and mutations of FLT3 and some other rare translocations. In addition, several other genes show abnormal expression levels in AML, including the Wilms tumor gene, the PRAME gene and Ig/TCR rearrangements. Most of these genetic abnormalities can be detected by qualitative but more importantly by quantitative RT-PCR. The kinetics of disappearance of molecular markers in AML differs between the various types of leukemias, although at least a 2 log reduction of transcript after induction chemotherapy is necessary for long-term remission in all types. Conversely, the change of PCR from negativity to positivity is highly predictive of relapse. Whereas in acute lymphoblastic leukemia, multiparametric flow cytometry is an established method for MRD detection, this is less so in AML. The reason is the absence of well-characterized leukemia-specific antigens and the existence of phenotypic changes at relapse. On the other hand, this method is convenient due to its simplicity and universal applicability. In conclusion, several methods can be used for MRD detection in AML patients; each has its pros and cons. Several issues still remain to be settled including the choice of the best method and the timing for MRD monitoring and above all the practical clinical implications of MRD in the various types of AML.
Collapse
Affiliation(s)
- P Raanani
- Institute of Hematology, Chaim Sheba Medical Center, Tel Hashomer and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | |
Collapse
|