1
|
George BM, Eleftheriou M, Yankova E, Perr J, Chai P, Nestola G, Almahayni K, Evans S, Damaskou A, Hemberger H, Lebedenko CG, Rak J, Yu Q, Bapcum E, Russell J, Bagri J, Volk RF, Spiekermann M, Stone RM, Giotopoulos G, Huntly BJP, Baxter J, Camargo F, Liu J, Zaro BW, Vassiliou GS, Möckl L, de la Rosa J, Flynn RA, Tzelepis K. Treatment of acute myeloid leukemia models by targeting a cell surface RNA-binding protein. Nat Biotechnol 2025:10.1038/s41587-025-02648-2. [PMID: 40269321 DOI: 10.1038/s41587-025-02648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
Immunotherapies for acute myeloid leukemia (AML) and other cancers are limited by a lack of tumor-specific targets. Here we discover that RNA-binding proteins and glycosylated RNAs (glycoRNAs) form precisely organized nanodomains on cancer cell surfaces. We characterize nucleophosmin (NPM1) as an abundant cell surface protein (csNPM1) on a variety of tumor types. With a focus on AML, we observe csNPM1 on blasts and leukemic stem cells but not on normal hematopoietic stem cells. We develop a monoclonal antibody to target csNPM1, which exhibits robust anti-tumor activity in multiple syngeneic and xenograft models of AML, including patient-derived xenografts, without observable toxicity. We find that csNPM1 is expressed in a mutation-agnostic manner on primary AML cells and may therefore offer a general strategy for detecting and treating AML. Surface profiling and in vivo work also demonstrate csNPM1 as a target on solid tumors. Our data suggest that csNPM1 and its neighboring glycoRNA-cell surface RNA-binding protein (csRBP) clusters may serve as an alternative antigen class for therapeutic targeting or cell identification.
Collapse
Affiliation(s)
- Benson M George
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Eleftheriou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Eliza Yankova
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Jonathan Perr
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Peiyuan Chai
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Gianluca Nestola
- Department of Physics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Karim Almahayni
- Department of Physics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Siân Evans
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Aristi Damaskou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Helena Hemberger
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Charlotta G Lebedenko
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Justyna Rak
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Qi Yu
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Ece Bapcum
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - James Russell
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Jaana Bagri
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Regan F Volk
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | | | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George Giotopoulos
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Brian J P Huntly
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Joanna Baxter
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Fernando Camargo
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Jie Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Balyn W Zaro
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - George S Vassiliou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Leonhard Möckl
- Max Planck Institute for the Science of Light, Erlangen, Germany
- Faculty of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Faculty of Sciences, Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jorge de la Rosa
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Cambridge Institute of Science, Altos Labs, Cambridge, UK
| | - Ryan A Flynn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Konstantinos Tzelepis
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK.
- Wellcome Trust Sanger Institute, Hinxton, UK.
| |
Collapse
|
2
|
Lecornec N, Duchmann M, Itzykson R. Single-cell sequencing applications in acute myeloid leukemia. Leuk Lymphoma 2025; 66:175-189. [PMID: 39496597 DOI: 10.1080/10428194.2024.2422833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of malignancies with poor prognosis. AML result from the proliferation of immature myeloid cells blocked at a variable stage of differentiation. Beyond inter-patient heterogeneity, AMLs are characterized by genetic and phenotypic intra-patient heterogeneity. Despite major advances in deciphering AML biology with bulk sequencing studies, pivotal questions remain unanswered. Analyses at the single-cell level could thus transform our understanding of these neoplasms. We review recent progresses in single-cell sequencing technologies from cell processing to bioinformatic pipelines. We next discuss how single-cell applications have helped understand the genetic and functional intra-leukemic heterogeneity, emphasizing aspects related to leukemic stem cells, clonal evolution and measurable residual disease (MRD) monitoring. We finally delineate how single-cell technologies could be implemented in routine clinical practice to improve patient management.
Collapse
Affiliation(s)
- Nicolas Lecornec
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département d'Immuno-Hématologie Pédiatrique, Hôpital Robert-Debré, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Matthieu Duchmann
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Raphael Itzykson
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
3
|
Papadopoulou V, Schiavini G, Stalder G, Basset V, Schoumans J, Nabergoj M, Schaller M. Characteristics and Prognosis of "Acute Promyelocytic Leukemia-like" Nucleophosmin-1-Mutated Acute Myeloid Leukemia in a Retrospective Patient Cohort. Biomedicines 2024; 12:2282. [PMID: 39457595 PMCID: PMC11505509 DOI: 10.3390/biomedicines12102282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background: AML with NPM1 mutation is the largest subcategory of AML, representing about 35% of AML cases. It is characterized by CD34 negativity, which suggests a relatively differentiated state of the bulk of leukemic blasts. Notably, a significant subset of NPM1-mutated AML cases also exhibit HLA-DR negativity, classifying them as "double-negative", and mimicking, therefore, the CD34- HLA-DR- immunophenotype of acute promyelocytic leukemia (APL). Objectives: This study focuses on the "acute promyelocytic leukemia-like" ("APL-like") subset of NPM1-mutated AML, which can be challenging to distinguish from APL at presentation, prior to confirming RARa translocations. We aim to investigate the hematologic and immunophenotypic parameters that may aid to its distinction from APL. Additionally, we explore differences in genetic profile and prognosis between "APL-like" and "non-APL-like" NPM1-mutated AML cases. Methods: We conducted a retrospective evaluation of 77 NPM1-mutated AML cases and 28 APL cases. Results: Morphological characteristics, hematologic parameters (such as DD/WBC and PT/WBC), and specific immunophenotypic markers (including SSC, CD64, and CD4) can assist in the early distinction of "APL-like" NPM1-mutated AML from APL. Regarding differences in genetic profiles and outcomes between "APL-like" and non-"APL-like" NPM1-mutated AML cases, we observed a significantly higher incidence of IDH1/2 /TET2 mutations, along with a significantly lower incidence of DNMT3A mutations in the "APL-like" subset compared to the non-"APL-like" subset. The frequency of Ras-pathway and FLT3 mutations did not differ between these last two groups, nor did their prognoses. Conclusions: Our findings contribute to a comprehensive characterization of NPM1-mutated AML, enhancing diagnostic accuracy and aiding in the detailed classification of the disease. This information may potentially guide targeted therapies or differentiation-based treatment strategies.
Collapse
Affiliation(s)
- Vasiliki Papadopoulou
- Hematology Service and Laboratory, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Giulia Schiavini
- Hematology Service and Laboratory, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Gregoire Stalder
- Hematology Service and Laboratory, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Hematology Service, Department of Oncology, Cantonal Hospital of Valais, 1951 Sion, Switzerland
| | - Valentin Basset
- Hematology Service and Laboratory, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Jacqueline Schoumans
- Oncogenetics Unit, Hematology Service and Laboratory, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Mitja Nabergoj
- Hematology Service, Department of Oncology, Cantonal Hospital of Valais, 1951 Sion, Switzerland
| | - Muriel Schaller
- Hematology Service and Laboratory, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| |
Collapse
|
4
|
Khan AA, James D, Andresen V, Atkey J, Bradbury R, Cargo C, Dillon R, Gjertsen BT, Goldstone AR, Leach R, Lock D, Narayanan M, Russell N, Verigou EA, Green S, Fielding AK, Falini B. Pre-B acute lymphoblastic leukemia presenting with NPM1 and FLT3 mutations. Am J Hematol 2024; 99:2018-2022. [PMID: 38567798 DOI: 10.1002/ajh.27301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/23/2024] [Accepted: 03/11/2024] [Indexed: 10/25/2024]
Affiliation(s)
- Alesia A Khan
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Daniel James
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Vibeke Andresen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Julie Atkey
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Rachel Bradbury
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Catherine Cargo
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Richard Dillon
- Department of Medical and Molecular Genetics, Guy's and King's Hospitals, London, UK
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Antony R Goldstone
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Richard Leach
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Daniel Lock
- Hematological Malignancy Diagnostic Service, Leeds Teaching Hospitals, Leeds, UK
| | - Mayanka Narayanan
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Nigel Russell
- Department of Medical and Molecular Genetics, Guy's and King's Hospitals, London, UK
| | - Eleni-Anna Verigou
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Simone Green
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Adele K Fielding
- Centre for Blood Research, Hull University Teaching Hospitals NHS Trust, Hull, UK
- Centre for Blood research, University of York, York, UK
| | - Brunangelo Falini
- Institute of Hematology and Center for Haemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
5
|
Harrop S, Nguyen PC, Robinson S, Nguyen T, Tiong IS, Came N, Baldwin K, Nguyen V, Chan KL, Blombery P, Westerman D. Immunophenotypic characterisation of acute myeloid leukaemia with UBTF tandem duplications. Br J Haematol 2024; 204:2492-2495. [PMID: 38544472 DOI: 10.1111/bjh.19427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 03/14/2024] [Indexed: 06/15/2024]
Affiliation(s)
- Sean Harrop
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Phillip C Nguyen
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Samuel Robinson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Tamia Nguyen
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ing Soo Tiong
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Neil Came
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Kylie Baldwin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Vuong Nguyen
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kah Lok Chan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Piers Blombery
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - David Westerman
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Matarraz S, Leoz P, Yeguas-Bermejo A, van der Velden V, Bras AE, Sánchez Gallego JI, Lecrevisse Q, Ayala-Bueno R, Teodosio C, Criado I, González-González M, Flores-Montero J, Avendaño A, Vidriales MB, Chillón MC, González T, García-Sanz R, Prieto Conde MI, Villamor N, Magnano L, Colado E, Fernández P, Sonneveld E, Philippé J, Reiterová M, Caballero Berrocal JC, Diaz-Gálvez FJ, Ramos F, Dávila Valls J, Manjón Sánchez R, Solano Tovar J, Calvo X, García Alonso L, Arenillas L, Alonso S, Fonseca A, Quirós Caso C, van Dongen JJM, Orfao A. Baseline immunophenotypic profile of bone marrow leukemia cells in acute myeloid leukemia with nucleophosmin-1 gene mutation: a EuroFlow study. Blood Cancer J 2023; 13:132. [PMID: 37666856 PMCID: PMC10477264 DOI: 10.1038/s41408-023-00909-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023] Open
Affiliation(s)
- Sergio Matarraz
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Pilar Leoz
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - Ana Yeguas-Bermejo
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - Vincent van der Velden
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anne E Bras
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jose I Sánchez Gallego
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Rosa Ayala-Bueno
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Cristina Teodosio
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Ignacio Criado
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - María González-González
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Juan Flores-Montero
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - Alejandro Avendaño
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - María B Vidriales
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - María C Chillón
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - Teresa González
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - Ramón García-Sanz
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - María I Prieto Conde
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Hematology Department, University Hospital of Salamanca, CIBERONC (CB16/12/00233), IBSAL, Accelerator program and Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca), Salamanca, Spain
| | - Neus Villamor
- Hematology Service, Hospital Clinic, Barcelona, Spain
| | - Laura Magnano
- Hematology Service, Hospital Clinic, Barcelona, Spain
| | - Enrique Colado
- Hematology Department and Laboratory Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Paula Fernández
- FACS/Stem Cell Laboratory, Kantonsspital Aarau, Aarau, Switzerland
| | | | - Jan Philippé
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Michaela Reiterová
- CLIP-Department of Pediatric Hematology and Oncology, Second Medical Faculty, Charles University and University Hospital Motol, Prague, Czech Republic
| | | | | | - Fernando Ramos
- Department of Hematology, Complejo Asistencial Universitario de León, León, Spain
| | | | | | - Jackeline Solano Tovar
- Department of Hematology, Complejo Asistencial Universitario de Palencia, Palencia, Spain
| | - Xavier Calvo
- Pathology Service, Hospital del Mar, Barcelona, Spain
| | | | | | - Sara Alonso
- Hematology Department and Laboratory Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ariana Fonseca
- Hematology Department and Laboratory Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Covadonga Quirós Caso
- Hematology Department and Laboratory Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jacques J M van Dongen
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (IBMCC; CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
7
|
Sendker S, Awada A, Domagalla S, Sendker M, Orhan E, Hoffmeister LM, Antoniou E, Niktoreh N, Reinhardt D, von Neuhoff N, Schneider M. RUNX1 mutation has no prognostic significance in paediatric AML: a retrospective study of the AML-BFM study group. Leukemia 2023; 37:1435-1443. [PMID: 37188777 PMCID: PMC10317839 DOI: 10.1038/s41375-023-01919-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023]
Abstract
In acute myeloid leukaemia (AML) RUNX1 mutation is characterised by certain clinicopathological features with poor prognosis and adverse risk by the European LeukemiaNet recommendation. Though initially considered as provisional category, the recent World Health Organisation (WHO) classification of 2022 removed RUNX1-mutated AML from the unique entity. However, the significance of RUNX1 mutation in paediatric AML remains unclear. We retrospectively analysed a German cohort of 488 paediatric patients with de novo AML, enroled in the AMLR12 or AMLR17 registry of the AML-BFM Study Group (Essen, Germany). A total of 23 paediatric AML patients (4.7%) harboured RUNX1 mutations, 18 of which (78%) had RUNX1 mutation at initial diagnosis. RUNX1 mutations were associated with older age, male gender, number of coexisting alterations and presence of FLT3-ITD but mutually exclusive of KRAS, KIT and NPM1 mutation. RUNX1 mutations did not prognostically impact overall or event-free survival. Response rates did not differ between patients with and without RUNX1 mutations. This comprehensive study, comprising the largest analysis of RUNX1 mutation in a paediatric cohort to date, reveals distinct but not unique clinicopathologic features, with no prognostic significance of RUNX1-mutated paediatric AML. These results broaden the perspective on the relevance of RUNX1 alterations in leukaemogenesis in AML.
Collapse
Affiliation(s)
- Stephanie Sendker
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Amani Awada
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Sophia Domagalla
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Michael Sendker
- University of Applied Sciences for Economics and Management (FOM), 20357, Hamburg, Germany
| | - Eser Orhan
- Centre for Research Acceleration in Paediatrics GmbH, 30175, Hannover, Germany
| | - Lina Marie Hoffmeister
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Evangelia Antoniou
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Naghmeh Niktoreh
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Dirk Reinhardt
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Nils von Neuhoff
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Markus Schneider
- Department of Paediatric Hematology and Oncology, University Children's Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
| |
Collapse
|
8
|
Tan J, Zhong M, Hu Y, Pan G, Yao J, Tang Y, Duan H, Jiang Y, Shan W, Lin J, Liu Y, Huang J, Zheng H, Zhou Y, Fu G, Li Z, Xu B, Zha J. Ritanserin suppresses acute myeloid leukemia by inhibiting DGKα to downregulate phospholipase D and the Jak-Stat/MAPK pathway. Discov Oncol 2023; 14:118. [PMID: 37392305 DOI: 10.1007/s12672-023-00737-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023] Open
Abstract
Refractory or relapsed (R/R) AML is the most challenging form of AML to treat. Due to frequent genetic mutations, therapy alternatives are limited. Here, we identified the role of ritanserin and its target DGKα in AML. Several AML cell lines and primary patient cells were treated with ritanserin and subjected to cell proliferation, apoptosis and gene analyses with CCK-8 assay, Annexin V/PI assay and Western blotting, respectively. We also evaluated the function of the ritanserin target diacylglycerol kinase alpha (DGKα) in AML by bioinformatics. In vitro experiments have revealed that ritanserin inhibits AML progression in a dose- and time-dependent manner, and it shows an anti-AML effect in xenograft mouse models. We further demonstrated that the expression of DGKα was elevated in AML and correlated with poor survival. Mechanistically, ritanserin negatively regulates SphK1 expression through PLD signaling, also inhibiting the Jak-Stat and MAPK signaling pathways via DGKα. These findings suggest that DGKα may be an available therapeutic target and provide effective preclinical evidence of ritanserin as a promising treatment for AML.
Collapse
Affiliation(s)
- Jinshui Tan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Mengya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yanyan Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Guangchao Pan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jingwei Yao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yuanfang Tang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Hongpeng Duan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yuelong Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Weihang Shan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jiaqi Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Yating Liu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jiewen Huang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361002, Fujian, China
| | - Huijian Zheng
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Yong Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Zhifeng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China.
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China.
| |
Collapse
|
9
|
Srinivasan Rajsri K, Roy N, Chakraborty S. Acute Myeloid Leukemia Stem Cells in Minimal/Measurable Residual Disease Detection. Cancers (Basel) 2023; 15:2866. [PMID: 37345204 PMCID: PMC10216329 DOI: 10.3390/cancers15102866] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by an abundance of incompletely matured or immature clonally derived hematopoietic precursors called leukemic blasts. Rare leukemia stem cells (LSCs) that can self-renew as well as give rise to leukemic progenitors comprising the bulk of leukemic blasts are considered the cellular reservoir of disease initiation and maintenance. LSCs are widely thought to be relatively resistant as well as adaptive to chemotherapy and can cause disease relapse. Therefore, it is imperative to understand the molecular bases of LSC forms and functions during different stages of disease progression, so we can more accurately identify these cells and design therapies to target them. Irrespective of the morphological, cytogenetic, and cellular heterogeneity of AML, the uniform, singularly important and independently significant prognosticator of disease response to therapy and patient outcome is measurable or minimal residual disease (MRD) detection, defined by residual disease detection below the morphology-based 5% blast threshold. The importance of LSC identification and frequency estimation during MRD detection, in order to make MRD more effective in predicting disease relapse and modifying therapeutic regimen is becoming increasingly apparent. This review focuses on summarizing functional and cellular composition-based LSC identification and linking those studies to current techniques of MRD detection to suggest LSC-inclusive MRD detection as well as outline outstanding questions that need to be addressed to improve the future of AML clinical management and treatment outcomes.
Collapse
Affiliation(s)
- Kritika Srinivasan Rajsri
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; (K.S.R.); (N.R.)
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Nainita Roy
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; (K.S.R.); (N.R.)
| | - Sohini Chakraborty
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; (K.S.R.); (N.R.)
| |
Collapse
|
10
|
Li C, Wang Z, Wei B, Liu Z, Li B, Kang H, Wang J, Liu J, Wang Q, Guo H, Wu X, Liu N, Luo J. Upregulation of ROBO3 promotes proliferation, migration and adhesion of AML cells and affects the survival of AML patients. Biochem Biophys Res Commun 2023; 661:1-9. [PMID: 37084487 DOI: 10.1016/j.bbrc.2023.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy, which is the most common and severe acute leukemia in adults. Its occurrence, development and prognosis are affected by many factors, and more research is still needed to further guide its treatment. Here, we found that roundabout3 (ROBO3) was associated with poor prognosis in AML through bioinformatics analysis. We then found that overexpression of ROBO3 promoted AML cell proliferation, adhesion and migration while knockdown of ROBO3 had opposite effects. We subsequently found that ROBO3 regulated CD34 expression in AML cells, and this regulatory effect may be achieved through the Hippo-YAP pathway. The inhibitors of this pathway, K-975 and verteporfin, showed an inhibitory effect on AML cells with high ROBO3 expression. ROBO3 was also found to be significantly increased in bone marrow samples from AML patients. Our research indicates that ROBO3 plays an important role in the development of AML, which suggests that ROBO3 can be a prognostic biomarker and potential therapeutic target for AML.
Collapse
Affiliation(s)
- Chaonan Li
- Department of Hematology, Key Laboratory of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China; Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Zhen Wang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Binghui Wei
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zechen Liu
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Bei Li
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Hening Kang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jue Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Junle Liu
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qingyu Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hongming Guo
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Xiaoli Wu
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Na Liu
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Jianmin Luo
- Department of Hematology, Key Laboratory of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
11
|
Maag AH, Swanton H, Kull M, Vegi NM, Feuring M. Immunophenotypical profiling of myeloid neoplasms with erythroid predominance using mass cytometry (CyTOF). Cytometry A 2023. [PMID: 36647792 DOI: 10.1002/cyto.a.24716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 12/10/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
Acute erythroid leukemia (AEL) is a disease continuum between Myelodysplastic syndrome (MDS) and Acute myeloid leukemia (AML) with the cellular hallmark of uncontrolled proliferation and impaired differentiation of erythroid progenitor cells. First described by Giovanni di Guglielmo in 1917 AEL accounts for less than 5% of all de novo AML cases. There have been efforts to characterize AEL at a molecular level, describing recurrent alterations in TP53, NPM1 and FLT3 genes. A genomic analysis of AEL cases confirmed its complexity. Despite these advances, the biology underlying erythroid proliferations remains unclear and the prognosis is dismal with a median survival of only 3 months for pure erythroid leukemia (PEL). Marker combinations suitable for the identification and characterization of leukemic stem cell (LSC) candidates, monitoring measurable residual disease (MRD) during chemotherapy treatment and the development of innovative targeted therapies are missing. Here, we developed a mass cytometry panel for an in-depth characterization of erythroid and myeloid blast cell populations from human AEL bone marrow samples in comparison to other AML subtypes and healthy counterparts. A total of 8 AEL samples were analyzed and compared to 28 AML samples from different molecular subtypes, healthy bone marrow counterparts (n = 5) and umbilical cord blood (n = 6) using high-dimensional mass cytometry. Identification of erythroid and myeloid blast populations in high-dimensional mass cytometry data enabled a refined view on erythroblast differentiation stages present in AEL erythroid blasts and revealed immunophenotypical profiles specific to AEL. Profiling of phenotypic LSCs revealed aberrant erythroid marker expression in the CD34+ CD38- stem cell compartment. In addition, the identification of novel candidate surface marker combinations and aberrancies might enhance clinical diagnostics of AEL. We present a high-parameter mass cytometry approach feasible for immunophenotypical analysis of blast and stem cell populations in myeloid neoplasms with erythroid predominance laying the foundation for more precise experimental approaches in the future.
Collapse
Affiliation(s)
- Abdul-Habib Maag
- Comprehensive Cancer Center, Institute of Experimental Cancer Research, University Hospital of Ulm, Ulm, Germany
| | - Helen Swanton
- Comprehensive Cancer Center, Institute of Experimental Cancer Research, University Hospital of Ulm, Ulm, Germany
| | - Miriam Kull
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Naidu M Vegi
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Michaela Feuring
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
12
|
Current status and future perspectives in targeted therapy of NPM1-mutated AML. Leukemia 2022; 36:2351-2367. [PMID: 36008542 PMCID: PMC9522592 DOI: 10.1038/s41375-022-01666-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/09/2022]
Abstract
Nucleophosmin 1 (NPM1) is a nucleus-cytoplasmic shuttling protein which is predominantly located in the nucleolus and exerts multiple functions, including regulation of centrosome duplication, ribosome biogenesis and export, histone assembly, maintenance of genomic stability and response to nucleolar stress. NPM1 mutations are the most common genetic alteration in acute myeloid leukemia (AML), detected in about 30–35% of adult AML and more than 50% of AML with normal karyotype. Because of its peculiar molecular and clinico-pathological features, including aberrant cytoplasmic dislocation of the NPM1 mutant and wild-type proteins, lack of involvement in driving clonal hematopoiesis, mutual exclusion with recurrent cytogenetic abnormalities, association with unique gene expression and micro-RNA profiles and high stability at relapse, NPM1-mutated AML is regarded as a distinct genetic entity in the World Health Organization (WHO) classification of hematopoietic malignancies. Starting from the structure and functions of NPM1, we provide an overview of the potential targeted therapies against NPM1-mutated AML and discuss strategies aimed at interfering with the oligomerization (compound NSC348884) and the abnormal traffic of NPM1 (avrainvillamide, XPO1 inhibitors) as well as at inducing selective NPM1-mutant protein degradation (ATRA/ATO, deguelin, (-)-epigallocatechin-3-gallate, imidazoquinoxaline derivatives) and at targeting the integrity of nucleolar structure (actinomycin D). We also discuss the current therapeutic results obtained in NPM1-mutated AML with the BCL-2 inhibitor venetoclax and the preliminary clinical results using menin inhibitors targeting HOX/MEIS1 expression. Finally, we review various immunotherapeutic approaches in NPM1-mutated AML, including immune check-point inhibitors, CAR and TCR T-cell-based therapies against neoantigens created by the NPM1 mutations.
Collapse
|
13
|
Shi Y, Xue Y, Wang C, Yu L. Nucleophosmin 1: from its pathogenic role to a tantalizing therapeutic target in acute myeloid leukemia. Hematology 2022; 27:609-619. [PMID: 35621728 DOI: 10.1080/16078454.2022.2067939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Nucleophosmin 1 (NPM1, also known as B23) is a multifunctional protein involved in a variety of cellular processes, including ribosomal maturation, centrosome replication, maintenance of genomic stability, cell cycle control, and apoptosis. NPM1 is the most commonly mutated gene in adult acute myeloid leukemia (AML) and is present in approximately 40% of all AML cases. The underlying mechanisms of mutant NPM1 (NPM1mut) in leukemogenesis remain unclear. This review summarizes the structure and physiological function of NPM1, mechanisms underlying the pathogenesis of NPM1-mutated AML, and the potential role of NPM1 as a therapeutic target. It is reported that dysfunctional NPM1 might cause AML pathogenesis via its role as a protein chaperone, inhibiting differentiation of leukemia stem cells and regulation of non-coding RNAs. Besides conventional chemotherapies, NPM1 is a promising therapeutic target against AML that warrants further investigation. NPM1-based therapeutic strategies include inducing nucleolar relocalisation of NPM1 mutants, interfering with NPM1 oligomerization, and NPM1 as an immune response target.
Collapse
Affiliation(s)
- Yuye Shi
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yuhao Xue
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China
| | - Chunling Wang
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Liang Yu
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
14
|
Pianigiani G, Rocchio F, Peruzzi S, Andresen V, Bigerna B, Sorcini D, Capurro M, Gjertsen BT, Sportoletti P, Di Ianni M, Martelli MP, Brunetti L, Falini B. The absent/low expression of CD34 in NPM1-mutated AML is not related to cytoplasmic dislocation of NPM1 mutant protein. Leukemia 2022; 36:1931-1934. [PMID: 35568767 PMCID: PMC9252915 DOI: 10.1038/s41375-022-01593-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Giulia Pianigiani
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Francesca Rocchio
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy.,Research and Early Development, Dompé Farmaceutici S.p.A, Napoli, Italy
| | - Sara Peruzzi
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Vibeke Andresen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Barbara Bigerna
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Daniele Sorcini
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Michela Capurro
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, Haematology Section, Haukeland University Hospital, Bergen, Norway
| | - Paolo Sportoletti
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Maria Paola Martelli
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Lorenzo Brunetti
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy.,Department of Molecular and Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Brunangelo Falini
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy.
| |
Collapse
|
15
|
Cell Origin-Dependent Cooperativity of Mutant Dnmt3a and Npm1 in Clonal Hematopoiesis and Myeloid Malignancy. Blood Adv 2022; 6:3666-3677. [PMID: 35413095 DOI: 10.1182/bloodadvances.2022006968] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/31/2022] [Indexed: 11/20/2022] Open
Abstract
In adult acute myeloid leukemia (AML), acquisition of driver somatic mutations may be preceded by a benign state termed clonal hematopoiesis (CH). To develop therapeutic strategies to prevent leukemia development from CH, it is important to understand the mechanisms by which CH-driving and AML-driving mutations cooperate. Here, we use mice with inducible mutant alleles common in human CH (DNMT3AR882; mouse Dnmt3aR878H) and AML (NPM1c; mouse Npm1cA). We find that Dnmt3aR878H/+ hematopoietic stem cells (HSCs), but not multipotent progenitor cell (MPP) subsets, have reduced expression of cytokine and pro-inflammatory transcriptional signatures and a functional competitive advantage over their wild-type counterparts. Dnmt3aR878H/+ HSCs are the most potent cell type transformed by Npm1cA, generating myeloid malignancies in which few additional cooperating somatic mutation events were detected. At a molecular level, Npm1cA in cooperation with Dnmt3aR878H acutely increased accessibility of a distinct set of promoters in HSCs compared to MPP cells. These promoters were enriched for cell cycling, PI3K/AKT/mTOR signaling, stem cell signatures, and targets of transcription factors including NFAT and the chromatin binding factor HMGB1, which have been implicated in human AML. These results demonstrate cooperativity between pre-existing Dnmt3aR878H and Npm1cA at the chromatin level, where specific loci altered in accessibility by Npm1cA are dependent on cell context as well as Dnmt3a mutation status. These findings have implications for biological understanding and therapeutic intervention into transformation from CH to AML.
Collapse
|
16
|
Darici S, Zavatti M, Braglia L, Accordi B, Serafin V, Horne GA, Manzoli L, Palumbo C, Huang X, Jørgensen HG, Marmiroli S. Synergistic cytotoxicity of dual PI3K/mTOR and FLT3 inhibition in FLT3-ITD AML cells. Adv Biol Regul 2021; 82:100830. [PMID: 34555701 DOI: 10.1016/j.jbior.2021.100830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 01/17/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy, characterized by a heterogeneous genetic landscape and complex clonal evolution, with poor outcomes. Mutation at the internal tandem duplication of FLT3 (FLT3-ITD) is one of the most common somatic alterations in AML, associated with high relapse rates and poor survival due to the constitutive activation of the FLT3 receptor tyrosine kinase and its downstream effectors, such as PI3K signaling. Thus, aberrantly activated FLT3-kinase is regarded as an attractive target for therapy for this AML subtype, and a number of small molecule inhibitors of this kinase have been identified, some of which are approved for clinical practice. Nevertheless, acquired resistance to these molecules is often observed, leading to severe clinical outcomes. Therapeutic strategies to tackle resistance include combining FLT3 inhibitors with other antileukemic agents. Here, we report on the preclinical activity of the combination of the FLT3 inhibitor quizartinib with the dual PI3K/mTOR inhibitor PF-04691502 in FLT3-ITD cells. Briefly, we show that the association of these two molecules displays synergistic cytotoxicity in vitro in FLT3-ITD AML cells, triggering 90% cell death at nanomolar concentrations after 48 h.
Collapse
Affiliation(s)
- Salihanur Darici
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy; Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Manuela Zavatti
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy
| | - Luca Braglia
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Benedetta Accordi
- Department of Woman and Child Health, Haemato-Oncology Laboratory, University of Padua, Via Giustiniani 3 and IRP Città Della Speranza, Corso Stati Uniti 4, 35128, Padua, Italy
| | - Valentina Serafin
- Department of Woman and Child Health, Haemato-Oncology Laboratory, University of Padua, Via Giustiniani 3 and IRP Città Della Speranza, Corso Stati Uniti 4, 35128, Padua, Italy
| | - Gillian A Horne
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carla Palumbo
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK.
| | - Heather G Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Sandra Marmiroli
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| |
Collapse
|
17
|
How I diagnose and treat NPM1-mutated AML. Blood 2021; 137:589-599. [PMID: 33171486 DOI: 10.1182/blood.2020008211] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations of the nucleophosmin (NPM1) gene, encoding for a nucleolar multifunctional protein, occur in approximately one-third of adult acute myeloid leukemia (AML). NPM1-mutated AML exhibits unique molecular, pathological, and clinical features, which led to its recognition as distinct entity in the 2017 World Health Organization (WHO) classification of myeloid neoplasms. Although WHO criteria for the diagnosis of NPM1-mutated AML are well established, its distinction from other AML entities may be difficult. Moreover, the percentage of blasts required to diagnose NPM1-mutated AML remains controversial. According to the European LeukemiaNet (ELN), determining the mutational status of NPM1 (together with FLT3) is mandatory for accurate relapse-risk assessment. NPM1 mutations are ideal targets for measurable residual disease (MRD) monitoring, since they are AML specific, frequent, very stable at relapse, and do not drive clonal hematopoiesis of undetermined significance. MRD monitoring by quantitative polymerase chain reaction of NPM1-mutant transcripts, possibly combined with ELN genetic-based risk stratification, can guide therapeutic decisions after remission. Furthermore, immunohistochemistry can be very useful in selected situations, such as diagnosis of NPM1-mutated myeloid sarcoma. Herein, we present 4 illustrative cases of NPM1-mutated AML that address important issues surrounding the biology, diagnosis, and therapy of this common form of leukemia.
Collapse
|
18
|
CD200 expression marks leukemia stem cells in human AML. Blood Adv 2021; 4:5402-5413. [PMID: 33147339 DOI: 10.1182/bloodadvances.2020001802] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/07/2020] [Indexed: 12/25/2022] Open
Abstract
The leukemia stem cell (LSC) populations of acute myeloid leukemia (AML) exhibit phenotypic, genetic, and functional heterogeneity that contribute to therapy failure and relapse. Progress toward understanding the mechanistic basis for therapy resistance in LSCs has been hampered by difficulties in isolating cell fractions that enrich for the entire heterogeneous population of LSCs within individual AML samples. We previously reported that CD200 gene expression is upregulated in LSC-containing AML fractions. Here, we show that CD200 is present on a greater proportion of CD45dim blasts compared with more differentiated CD45high cells in AML patient samples. In 75% (49 of 65) of AML cases we examined, CD200 was expressed on ≥10% of CD45dim blasts; of these, CD200 identified LSCs within the blast population in 9 of 10 (90%) samples tested in xenotransplantation assays. CD200+ LSCs could be isolated from CD200+ normal HSCs with the use of additional markers. Notably, CD200 expression captured both CD34- and CD34+ LSCs within individual AML samples. Analysis of highly purified CD200+ LSC-containing fractions from NPM1-mutated AMLs, which are commonly CD34-, exhibited an enrichment of primitive gene expression signatures compared with unfractionated cells. Overall, our findings support CD200 as a novel LSC marker that is able to capture the entire LSC compartment from AML patient samples, including those with NPM1 mutation.
Collapse
|
19
|
Swatler J, Turos-Korgul L, Kozlowska E, Piwocka K. Immunosuppressive Cell Subsets and Factors in Myeloid Leukemias. Cancers (Basel) 2021; 13:cancers13061203. [PMID: 33801964 PMCID: PMC7998753 DOI: 10.3390/cancers13061203] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Effector immune system cells have the ability to kill tumor cells. However, as a cancer (such as leukemia) develops, it inhibits and evades the effector immune response. Such a state of immunosuppression can be driven by several factors – receptors, soluble cytokines, as well as by suppressive immune cells. In this review, we describe factors and cells that constitute immunosuppressive microenvironment of myeloid leukemias. We characterize factors of direct leukemic origin, such as inhibitory receptors, enzymes and extracellular vesicles. Furthermore, we describe suppressive immune cells, such as myeloid derived suppressor cells and regulatory T cells. Finally, we sum up changes in these drivers of immune evasion in myeloid leukemias during therapy. Abstract Both chronic myeloid leukemia and acute myeloid leukemia evade the immune response during their development and disease progression. As myeloid leukemia cells modify their bone marrow microenvironment, they lead to dysfunction of cytotoxic cells, such as CD8+ T cells or NK cells, simultaneously promoting development of immunosuppressive regulatory T cells and suppressive myeloid cells. This facilitates disease progression, spreading of leukemic blasts outside the bone marrow niche and therapy resistance. The following review focuses on main immunosuppressive features of myeloid leukemias. Firstly, factors derived directly from leukemic cells – inhibitory receptors, soluble factors and extracellular vesicles, are described. Further, we outline function, properties and origin of main immunosuppressive cells - regulatory T cells, myeloid derived suppressor cells and macrophages. Finally, we analyze interplay between recovery of effector immunity and therapeutic modalities, such as tyrosine kinase inhibitors and chemotherapy.
Collapse
Affiliation(s)
- Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
| | - Laura Turos-Korgul
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
| | - Ewa Kozlowska
- Department of Immunology, Institute of Functional Biology and Ecology, University of Warsaw, 02-096 Warsaw, Poland;
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
- Correspondence:
| |
Collapse
|
20
|
Zeisig BB, Fung TK, Zarowiecki M, Tsai CT, Luo H, Stanojevic B, Lynn C, Leung AYH, Zuna J, Zaliova M, Bornhauser M, von Bonin M, Lenhard B, Huang S, Mufti GJ, So CWE. Functional reconstruction of human AML reveals stem cell origin and vulnerability of treatment-resistant MLL-rearranged leukemia. Sci Transl Med 2021; 13:eabc4822. [PMID: 33627486 DOI: 10.1126/scitranslmed.abc4822] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 01/08/2021] [Indexed: 01/13/2023]
Abstract
Chemoresistance remains the major challenge for successful treatment of acute myeloid leukemia (AML). Although recent mouse studies suggest that treatment response of genetically and immunophenotypically indistinguishable AML can be influenced by their different cells of origin, corresponding evidence in human disease is still largely lacking. By combining prospective disease modeling using highly purified human hematopoietic stem or progenitor cells with retrospective deconvolution study of leukemia stem cells (LSCs) from primary patient samples, we identified human hematopoietic stem cells (HSCs) and common myeloid progenitors (CMPs) as two distinctive origins of human AML driven by Mixed Lineage Leukemia (MLL) gene fusions (MLL-AML). Despite LSCs from either MLL-rearranged HSCs or MLL-rearranged CMPs having a mature CD34-/lo/CD38+ immunophenotype in both a humanized mouse model and primary patient samples, the resulting AML cells exhibited contrasting responses to chemotherapy. HSC-derived MLL-AML was highly resistant to chemotherapy and expressed elevated amounts of the multispecific anion transporter ABCC3. Inhibition of ABCC3 by shRNA-mediated knockdown or with small-molecule inhibitor fidaxomicin, currently used for diarrhea associated with Clostridium difficile infection, effectively resensitized HSC-derived MLL-AML toward standard chemotherapeutic drugs. This study not only functionally established two distinctive origins of human LSCs for MLL-AML and their role in mediating chemoresistance but also identified a potential therapeutic avenue for stem cell-associated treatment resistance by repurposing a well-tolerated antidiarrhea drug already used in the clinic.
Collapse
Affiliation(s)
- Bernd B Zeisig
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK
- Department of Haematological Medicine, King's College Hospital, London SE5 9RS, UK
| | - Tsz Kan Fung
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK
- Department of Haematological Medicine, King's College Hospital, London SE5 9RS, UK
| | - Magdalena Zarowiecki
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK
| | - Chiou Tsun Tsai
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK
| | - Huacheng Luo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Boban Stanojevic
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK
- Laboratory for Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Science, University of Belgrade, 11000 Belgrade, Serbia
| | - Claire Lynn
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK
| | - Anskar Y H Leung
- Department of Medicine, The University of Hong Kong, Pokfulam Road, HKSAR, China
| | - Jan Zuna
- CLIP, Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague 5, Czech Republic
| | - Marketa Zaliova
- CLIP, Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague 5, Czech Republic
| | | | - Malte von Bonin
- Department of Medicine, University Hospital, 01307 Dresden, Germany
| | - Boris Lenhard
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
- Computational Regulatory Genomics, MRC London Institute of Medical Sciences, London W12 0NN, UK
- Sars International Centre for Marine Molecular Biology, University of Bergen, N-5008 Bergen, Norway
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Ghulam J Mufti
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK
- Department of Haematological Medicine, King's College Hospital, London SE5 9RS, UK
| | - Chi Wai Eric So
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College, London SE5 9NU, UK.
- Department of Haematological Medicine, King's College Hospital, London SE5 9RS, UK
| |
Collapse
|
21
|
Perriello VM, Gionfriddo I, Rossi R, Milano F, Mezzasoma F, Marra A, Spinelli O, Rambaldi A, Annibali O, Avvisati G, Di Raimondo F, Ascani S, Falini B, Martelli MP, Brunetti L. CD123 Is Consistently Expressed on NPM1-Mutated AML Cells. Cancers (Basel) 2021; 13:cancers13030496. [PMID: 33525388 PMCID: PMC7865228 DOI: 10.3390/cancers13030496] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary One-third of adult acute myeloid leukemia (AML) harbors NPM1 mutations. A deep knowledge of the distribution of selected antigens on the surface of NPM1-mutated AML cells may help optimizing new therapies for this frequent AML subtype. CD123 is known to be expressed on leukemic cells but also on healthy hematopoietic and endothelial cells, although at lower levels. Differences in antigen densities between AML and healthy cells may enlighten therapeutic windows, where targeting CD123 could be effective without triggering “on-target off-tumor” toxicities. Here, we perform a thorough analysis of CD123 expression demonstrating high expression of this antigen on both NPM1-mutated bulk leukemic cells and CD34+CD38− cells. Abstract NPM1-mutated (NPM1mut) acute myeloid leukemia (AML) comprises about 30% of newly diagnosed AML in adults. Despite notable advances in the treatment of this frequent AML subtype, about 50% of NPM1mut AML patients treated with conventional treatment die due to disease progression. CD123 has been identified as potential target for immunotherapy in AML, and several anti-CD123 therapeutic approaches have been developed for AML resistant to conventional therapies. As this antigen has been previously reported to be expressed by NPM1mut cells, we performed a deep flow cytometry analysis of CD123 expression in a large cohort of NPM1mut and wild-type samples, examining the whole blastic population, as well as CD34+CD38− leukemic cells. We demonstrate that CD123 is highly expressed on NPM1mut cells, with particularly high expression levels showed by CD34+CD38− leukemic cells. Additionally, CD123 expression was further enhanced by FLT3 mutations, which frequently co-occur with NPM1 mutations. Our results identify NPM1-mutated and particularly NPM1/FLT3 double-mutated AML as disease subsets that may benefit from anti-CD123 targeted therapies.
Collapse
Affiliation(s)
- Vincenzo Maria Perriello
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Ilaria Gionfriddo
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Roberta Rossi
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Francesca Milano
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Federica Mezzasoma
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Andrea Marra
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Orietta Spinelli
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy; (O.S.); (A.R.)
| | - Alessandro Rambaldi
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy; (O.S.); (A.R.)
- Department of Oncology and Hematology, University of Milan, 20122 Milan, Italy
| | - Ombretta Annibali
- Hematology and Stem Cell Transplant Unit, Campus Biomedico University Hospital, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Giuseppe Avvisati
- Hematology and Stem Cell Transplant Unit, Campus Biomedico University Hospital, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Francesco Di Raimondo
- Hematology and Bone Marrow Transplant Unit, Catania University Hospital, 95125 Catania, Italy;
| | - Stefano Ascani
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Pathology, Santa Maria Hospital, 05100 Terni, Italy
| | - Brunangelo Falini
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
| | - Maria Paola Martelli
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Correspondence: (M.P.M.); (L.B.)
| | - Lorenzo Brunetti
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Correspondence: (M.P.M.); (L.B.)
| |
Collapse
|
22
|
Diagnostic and therapeutic pitfalls in NPM1-mutated AML: notes from the field. Leukemia 2021; 35:3113-3126. [PMID: 33879827 PMCID: PMC8056374 DOI: 10.1038/s41375-021-01222-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/21/2021] [Accepted: 03/09/2021] [Indexed: 02/02/2023]
Abstract
Mutations of Nucleophosmin (NPM1) are the most common genetic abnormalities in adult acute myeloid leukaemia (AML), accounting for about 30% of cases. NPM1-mutated AML has been recognized as distinct entity in the 2017 World Health Organization (WHO) classification of lympho-haematopoietic neoplasms. WHO criteria allow recognition of this leukaemia entity and its distinction from AML with myelodysplasia-related changes, AML with BCR-ABL1 rearrangement and AML with RUNX1 mutations. Nevertheless, controversial issues include the percentage of blasts required for the diagnosis of NPM1-mutated AML and whether cases of NPM1-mutated myelodysplasia and chronic myelomonocytic leukaemia do exist. Evaluation of NPM1 and FLT3 status represents a major pillar of the European LeukemiaNet (ELN) genetic-based risk stratification model. Moreover, NPM1 mutations are particularly suitable for assessing measurable residual disease (MRD) since they are frequent, stable at relapse and do not drive clonal haematopoiesis. Ideally, combining monitoring of MRD with the ELN prognostication model can help to guide therapeutic decisions. Here, we provide examples of instructive cases of NPM1-mutated AML, in order to provide criteria for the appropriate diagnosis and therapy of this frequent leukaemia entity.
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Nucleophosmin (NPM1) mutations are encountered in myeloid neoplasia and are present in ~ 30% of de novo acute myeloid leukemia cases. This review summarizes features of mutant NPM1-related disease, with a particular emphasis on recent discoveries relevant to disease monitoring, prognostication, and therapeutic intervention. RECENT FINDINGS Recent studies have shown that HOX/MEIS gene overexpression is central to the survival of NPM1-mutated cells. Two distinct classes of small molecule drugs, BH3 mimetics and menin-MLL interaction inhibitors, have demonstrated exquisite leukemic cell toxicity in preclinical AML models associated with HOX/MEIS overexpression, and the former of these has shown efficacy in older treatment-naïve NPM1-mutated AML patients. The results of ongoing clinical trials further investigating these compounds will be of particular importance and may alter the clinical management of patients with NPM1-mutated myeloid neoplasms. Significant scientific advancements over the last decade, including improved sequencing and disease monitoring techniques, have fostered a much deeper understanding of mutant NPM1 disease biology, prognostication, and opportunities for therapeutic intervention. These discoveries have led to the development of clinical assays that permit the detection and monitoring of mutant NPM1 and have paved the way for future investigation of targeted therapeutics using emerging cutting-edge techniques.
Collapse
Affiliation(s)
- Sanjay S Patel
- Division of Hematopathology, Weill Cornell Medical College, New York, NY, USA
| | - Michael J Kluk
- Division of Hematopathology, Weill Cornell Medical College, New York, NY, USA
| | - Olga K Weinberg
- Department of Pathology, Boston Children's Hospital, 300 Longwood Avenue, Bader 126.2, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Gupta M, Jafari K, Rajab A, Wei C, Mazur J, Tierens A, Hyjek E, Musani R, Porwit A. Radar plots facilitate differential diagnosis of acute promyelocytic leukemia and NPM1+ acute myeloid leukemia by flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:409-420. [PMID: 33301193 PMCID: PMC8359362 DOI: 10.1002/cyto.b.21979] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/09/2020] [Accepted: 11/24/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Acute promyelocytic leukemia (APL) is one of the most life-threatening hematological emergencies and requires a prompt correct diagnosis by cytomorphology and flow cytometry (FCM) with later confirmation by cytogenetics/molecular genetics. However, nucleophosmin 1 muted acute myeloid leukemia (NPM1+ AML) can mimic APL, especially the hypogranular variant of APL. Our study aimed to develop a novel, Radar plot-based FCM strategy to distinguish APLs and NPM1+ AMLs quickly and accurately. METHOD Diagnostic samples from 52 APL and 32 NPM1+ AMLs patients were analyzed by a 3-tube panel of 10-color FCM. Radar plots combining all markers were constructed for each tube. Percentages of positive leukemic cells and mean fluorescence intensity were calculated for all the markers. RESULTS APL showed significantly higher expression of CD64, CD2, and CD13, whereas more leukemic cells were positive for CD11b, CD11c, CD15, CD36, and HLA-DR in NPM1+ AMLs. Radar plots featured CD2 expression, a lack of a monocytic component, lack of expression of HLA-DR and CD15, and a lack of a prominent CD11c+ population as recurring characteristics of APL. The presence of blasts with low SSC, presence of at least some monocytes, some expression of HLA-DR and/or CD15, and a prominent CD11c population were recurrent characteristics of NPM1+ AMLs. Radar plot analysis could confidently separate all hypergranular APL cases from any NPM1+ AML and in 90% of cases between variant APL and blastic NPM1+ AML. CONCLUSION Radar plots can potentially add to differential diagnostics as they exhibit characteristic patterns distinguishing APL and different types of NPM1+ AMLs.
Collapse
Affiliation(s)
- Monali Gupta
- Immunophenotyping Laboratory, Viapath Analytics LLP, Department of Hematology, Kings College Hospital, London, UK.,Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada
| | - Katayoon Jafari
- Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada.,Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Amr Rajab
- Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada.,Medical-Scientific Department, Lifelabs Medical Laboratory Services, Toronto, Ontario, Canada
| | - Cuihong Wei
- Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada.,Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Joanna Mazur
- Department of Humanization of Medicine and Sexology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland.,Department of Child and Adolescent Health, Institute of Mother and Child, Warsaw, Poland
| | - Anne Tierens
- Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada
| | - Elizabeth Hyjek
- Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada.,Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Rumina Musani
- Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada
| | - Anna Porwit
- Department of Pathobiology and Laboratory Medicine, Division of Hematopathology, University Health Network, Toronto, Ontario, Canada.,Faculty of Medicine, Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Multiparametric in situ imaging of NPM1-mutated acute myeloid leukemia reveals prognostically-relevant features of the marrow microenvironment. Mod Pathol 2020; 33:1380-1388. [PMID: 32051557 DOI: 10.1038/s41379-020-0498-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022]
Abstract
Ancillary testing during the initial workup of acute myeloid leukemia (AML) is largely performed using aspirated materials. We utilized multiplex immunofluorescence (MIF) imaging with digital image analysis to perform an in situ analysis of the microenvironment in NPM1-mutated AML using diagnostic bone marrow biopsy tissues (N = 17) and correlated these findings with diagnostic next-generation sequencing (NGS, N = 17), flow cytometry (FC, N = 14), and first remission (CR1) NPM1-specific molecular MRD (n = 16) data. The total CD3-positive T-cell percentages correlated positively between FC and MIF (r = 0.53, p = 0.05), but were significantly lower by MIF (1.62% vs. 3.4%, p = 0.009). The percentage of mutant NPM1-positive (NPM1c+) cells ranged from 9.7 to 90.8% (median 45.4%) and did not correlate with the NPM1 mutant allele fraction by NGS (p > 0.05). The percentage of CD34+/NPM1c+ cells ranged from 0 to 1.8% (median 0.07%). The percentage of NPM1c+ cells correlated inversely (34% vs. 62%, p = 0.03), while the percentages of CD3-/NPM1c- cells (64% vs. 35%, p = 0.03), and specifically CD3-/CD4-/NPM1c- cells (26% vs. 13%, p = 0.04), correlated positively with subsequent MRD. Discordances between MIF and FC/NGS data suggest that aspirate materials are likely an imperfect reflection of the core biopsy tissue. Furthermore, increased numbers of NPM1 wild-type cells within the microenvironment at diagnosis correlate with the subsequent presence of MRD.
Collapse
|
26
|
Beziat G, Tavitian S, Bertoli S, Huguet F, Largeaud L, Luquet I, Vergez F, Rieu J, Bories P, Delabesse E, Récher C. Dactinomycin in acute myeloid leukemia with NPM1 mutations. Eur J Haematol 2020; 105:302-307. [DOI: 10.1111/ejh.13438] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Guillaume Beziat
- Service d'Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Suzanne Tavitian
- Service d'Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Sarah Bertoli
- Service d'Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
- Université Toulouse III Paul Sabatier Toulouse France
| | - Françoise Huguet
- Service d'Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Laetitia Largeaud
- Université Toulouse III Paul Sabatier Toulouse France
- Laboratoire d’Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Isabelle Luquet
- Laboratoire d’Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - François Vergez
- Laboratoire d’Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Jean‐Baptiste Rieu
- Laboratoire d’Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Pierre Bories
- Réseau Onco‐Occitanie Institut Universitaire du Cancer de Toulouse‐Oncopole Toulouse France
| | - Eric Delabesse
- Université Toulouse III Paul Sabatier Toulouse France
- Laboratoire d’Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Christian Récher
- Service d'Hématologie Institut Universitaire du Cancer de Toulouse Oncopole Centre Hospitalier Universitaire de Toulouse Toulouse France
- Université Toulouse III Paul Sabatier Toulouse France
| |
Collapse
|
27
|
Diaz de la Guardia R, González-Silva L, López-Millán B, Rodríguez-Sevilla JJ, Baroni ML, Bueno C, Anguita E, Vives S, Palomo L, Lapillonne H, Varela I, Menendez P. Bone Marrow Clonogenic Myeloid Progenitors from NPM1-Mutated AML Patients Do Not Harbor the NPM1 Mutation: Implication for the Cell-Of-Origin of NPM1+ AML. Genes (Basel) 2020; 11:genes11010073. [PMID: 31936647 PMCID: PMC7017102 DOI: 10.3390/genes11010073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/05/2020] [Accepted: 01/07/2020] [Indexed: 11/24/2022] Open
Abstract
The cell-of-origin of NPM1- and FLT3-mutated acute myeloid leukemia (AML) is still a matter of debate. Here, we combined in vitro clonogenic assays with targeted sequencing to gain further insights into the cell-of-origin of NPM1 and FLT3-ITD-mutated AML in diagnostic bone marrow (BM) from nine NPM1+/FLT3-ITD (+/−) AMLs. We reasoned that individually plucked colony forming units (CFUs) are clonal and reflect the progeny of a single stem/progenitor cell. NPM1 and FLT3-ITD mutations seen in the diagnostic blasts were found in only 2/95 and 1/57 individually plucked CFUs, suggesting that BM clonogenic myeloid progenitors in NPM1-mutated and NPM1/FLT3-ITD-mutated AML patients do not harbor such molecular lesions. This supports previous studies on NPM1 mutations as secondary mutations in AML, likely acquired in an expanded pool of committed myeloid progenitors, perhaps CD34−, in line with the CD34−/low phenotype of NPM1-mutated AMLs. This study has important implications on the cell-of-origin of NPM1+ AML, and reinforces that therapeutic targeting of either NPM1 or FLT3-ITD mutations might only have a transient clinical benefit in debulking the leukemia, but is unlikely to be curative since will not target the AML-initiating/preleukemic cells. The absence of NPM1 and FLT3-ITD mutations in normal clonogenic myeloid progenitors is in line with their absence in clonal hematopoiesis of indeterminate potential.
Collapse
Affiliation(s)
- Rafael Diaz de la Guardia
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; (R.D.d.l.G.); (B.L.-M.); (J.J.R.-S.); (M.L.B.); (C.B.)
| | - Laura González-Silva
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, 39011 Santander, Spain; (L.G.-S.); (I.V.)
| | - Belén López-Millán
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; (R.D.d.l.G.); (B.L.-M.); (J.J.R.-S.); (M.L.B.); (C.B.)
| | - Juan José Rodríguez-Sevilla
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; (R.D.d.l.G.); (B.L.-M.); (J.J.R.-S.); (M.L.B.); (C.B.)
| | - Matteo L. Baroni
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; (R.D.d.l.G.); (B.L.-M.); (J.J.R.-S.); (M.L.B.); (C.B.)
| | - Clara Bueno
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; (R.D.d.l.G.); (B.L.-M.); (J.J.R.-S.); (M.L.B.); (C.B.)
| | - Eduardo Anguita
- Hematology and hemotherapy Department, Hospital Clínico San Carlos, IMDL, IdISSC, Departamento de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Susana Vives
- Hematology Department, ICO-Hospital Germans Trias i Pujol, 08916 Badalona, Spain; (S.V.); (L.P.)
- Josep Carreras Leukemia Research Institute, Universitat Autònoma Barcelona, 08193 Barcelona, Spain
| | - Laura Palomo
- Hematology Department, ICO-Hospital Germans Trias i Pujol, 08916 Badalona, Spain; (S.V.); (L.P.)
- Josep Carreras Leukemia Research Institute, Universitat Autònoma Barcelona, 08193 Barcelona, Spain
| | - Helene Lapillonne
- Sorbonne Université, INSERM, Centre de recherche Saint-Antoine CRSA, AP-HP, Hôspital Armand Trousseau, Haematology Laboratory, F-75012 Paris, France;
| | - Ignacio Varela
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, 39011 Santander, Spain; (L.G.-S.); (I.V.)
| | - Pablo Menendez
- Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; (R.D.d.l.G.); (B.L.-M.); (J.J.R.-S.); (M.L.B.); (C.B.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBER-ONC), ISCIII, 08036 Barcelona, Spain
- Instituciò Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
- Correspondence:
| |
Collapse
|
28
|
Mitchell K, Steidl U. Targeting Immunophenotypic Markers on Leukemic Stem Cells: How Lessons from Current Approaches and Advances in the Leukemia Stem Cell (LSC) Model Can Inform Better Strategies for Treating Acute Myeloid Leukemia (AML). Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036251. [PMID: 31451539 DOI: 10.1101/cshperspect.a036251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapies targeting cell-surface antigens in acute myeloid leukemia (AML) have been tested over the past 20 years with limited improvement in overall survival. Recent advances in the understanding of AML pathogenesis support therapeutic targeting of leukemia stem cells as the most promising avenue toward a cure. In this review, we provide an overview of the evolving leukemia stem cell (LSC) model, including evidence of the cell of origin, cellular and molecular disease architecture, and source of relapse in AML. In addition, we explore limitations of current targeted strategies utilized in AML and describe the various immunophenotypic antigens that have been proposed as LSC-directed therapeutic targets. We draw lessons from current approaches as well as from the (pre)-LSC model to suggest criteria that immunophenotypic targets should meet for more specific and effective elimination of disease-initiating clones, highlighting in detail a few targets that we suggest fit these criteria most completely.
Collapse
Affiliation(s)
- Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, New York 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
29
|
Brennan L, Narendran A. Cancer Stem Cells in the Development of Novel Therapeutics for Refractory Pediatric Leukemia. Stem Cells Dev 2019; 28:1277-1287. [PMID: 31364487 DOI: 10.1089/scd.2019.0035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although treatment strategies for pediatric leukemia have improved overall survival rates in the recent past, relapse rates in certain subgroups such as infant leukemia remain unacceptably high. Despite undergoing extensive chemotherapy designed to target the rapidly proliferating leukemia cells, many of these children experience relapse. In refractory leukemia, the existence of cell populations with stemness characteristics, termed leukemia stem cells (LSCs), which remain quiescent and subsequently replenish the blast population, has been described. A significant body of evidence exists, derived largely from xenograft models of adult acute myeloid leukemia, to support the idea that LSCs may play a fundamental role in refractory disease. In addition, clinical studies have also linked LSCs with increased minimal residual disease, higher relapse rate, and decreased survival rates in these patients. Recently, a number of reports have addressed effective ways to utilize new-generation genomic sequencing and transcriptomic analyses to identify targeted therapeutic agents aimed at LSCs, while sparing normal hematopoietic stem cells. These data underscore the value of timely translation of knowledge from adult studies to the unique molecular and physiological characteristics seen in pediatric leukemia. We aim to summarize this article in the rapidly expanding field of stem cell biology in hematopoietic malignancies, focusing particularly on relevant preclinical models and novel targeted therapeutics, and their applicability to childhood leukemia.
Collapse
Affiliation(s)
| | - Aru Narendran
- Division of Pediatric Hematology, Oncology and Transplant, POETIC Laboratory for Novel Therapeutics Discovery in Pediatric Oncology, Alberta Children's Hospital, Calgary, Canada
| |
Collapse
|
30
|
The acetyltransferase GCN5 maintains ATRA-resistance in non-APL AML. Leukemia 2019; 33:2628-2639. [PMID: 31576004 DOI: 10.1038/s41375-019-0581-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/04/2019] [Indexed: 02/06/2023]
Abstract
To date, only one subtype of acute myeloid leukemia (AML), acute promyelocytic leukemia (APL) can be effectively treated by differentiation therapy utilizing all-trans retinoic acid (ATRA). Non-APL AMLs are resistant to ATRA. Here we demonstrate that the acetyltransferase GCN5 contributes to ATRA resistance in non-APL AML via aberrant acetylation of histone 3 lysine 9 (H3K9ac) residues maintaining the expression of stemness and leukemia associated genes. We show that inhibition of GCN5 unlocks an ATRA-driven therapeutic response. This response is potentiated by coinhibition of the lysine demethylase LSD1, leading to differentiation in most non-APL AML. Induction of differentiation was not correlated to a specific AML subtype, cytogenetic, or mutational status. Our study shows a previously uncharacterized role of GCN5 in maintaining the immature state of leukemic blasts and identifies GCN5 as a therapeutic target in AML. The high efficacy of the combined epigenetic treatment with GCN5 and LSD1 inhibitors may enable the use of ATRA for differentiation therapy of non-APL AML. Furthermore, it supports a strategy of combined targeting of epigenetic factors to improve treatment, a concept potentially applicable for a broad range of malignancies.
Collapse
|
31
|
Acute Myeloid Leukemia Stem Cell Heterogeneity and Its Clinical Relevance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1139:153-169. [DOI: 10.1007/978-3-030-14366-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
Potter N, Miraki-Moud F, Ermini L, Titley I, Vijayaraghavan G, Papaemmanuil E, Campbell P, Gribben J, Taussig D, Greaves M. Single cell analysis of clonal architecture in acute myeloid leukaemia. Leukemia 2019; 33:1113-1123. [PMID: 30568172 PMCID: PMC6451634 DOI: 10.1038/s41375-018-0319-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 11/09/2022]
Abstract
We used single cell Q-PCR on a micro-fluidic platform (Fluidigm) to analyse clonal, genetic architecture and phylogeny in acute myeloid leukaemia (AML) using selected mutations. Ten cases of NPM1c mutant AML were screened for 111 mutations that are recurrent in AML and cancer. Clonal architectures were relatively simple with one to six sub-clones and were branching in some, but not all, patients. NPM1 mutations were secondary or sub-clonal to other driver mutations (DNM3TA, TET2, WT1 and IDH2) in all cases. In three of the ten cases, single cell analysis of enriched CD34+/CD33- cells revealed a putative pre-leukaemic sub-clone, undetectable in the bulk CD33+ population that had one or more driver mutations but lacked NPM1c. Cells from all cases were transplanted into NSG mice and in most (8/10), more than one sub-clone (#2-5 sub-clones) transplanted. However, the dominant regenerating sub-clone in 9/10 cases was NPM1+ and this sub-clone was either dominant or minor in the diagnostic sample from which it was derived. This study provides further evidence, at the single cell level, for genetic variegation in sub-clones and stem cells in acute leukaemia and demonstrates both a preferential order of mutation accrual and parallel evolution of sub-clones.
Collapse
Affiliation(s)
- Nicola Potter
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | | | - Luca Ermini
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Ian Titley
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | | | | | | | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
33
|
Touzet L, Dumezy F, Roumier C, Berthon C, Bories C, Quesnel B, Preudhomme C, Boyer T. CD9 in acute myeloid leukemia: Prognostic role and usefulness to target leukemic stem cells. Cancer Med 2019; 8:1279-1288. [PMID: 30740913 PMCID: PMC6434215 DOI: 10.1002/cam4.2007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/14/2018] [Accepted: 01/15/2019] [Indexed: 01/04/2023] Open
Abstract
CD9 is a cell surface protein and belongs to the tetraspanin family. Its role in carcinomagenesis has been widely studied in solid tumors but remains controversial, depending on the cancer type. Although CD9 seems to be associated with unfavorable outcome and disease progression in acute lymphoblastic leukemia (ALL), this marker has not yet been studied in acute myeloid leukemia (AML). First, we explored its prognostic role and its association with biological factors in a cohort of 112 AML patients treated with intensive chemotherapy. CD9 was expressed in 40% of AML and was associated with a favorable outcome (event‐free survival and relapse‐free survival) in univariate (P = 0.009 and P = 0.048, respectively) and multivariate (P = 0.004 and P = 0.039, respectively) analyses. Interestingly, CD9 expression was different between the more immature physiologic and AML cells (CD34+CD38−) as it was also expressed in AML on putative leukemic stem cells (LSCs) but not on hematopoietic stem cells (HSCs). Hence, CD9 could be a very relevant marker for minimal residual disease (MRD) monitoring in AML based on LSC targeting.
Collapse
Affiliation(s)
- Lucas Touzet
- Laboratory of Hematology, CHU Lille, Lille, France
| | | | | | | | | | | | | | - Thomas Boyer
- Laboratory of Hematology, CHU Lille, Lille, France
| |
Collapse
|
34
|
Kaur M, Drake AC, Hu G, Rudnick S, Chen Q, Phennicie R, Attar R, Nemeth J, Gaudet F, Chen J. Induction and Therapeutic Targeting of Human NPM1c + Myeloid Leukemia in the Presence of Autologous Immune System in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:1885-1894. [PMID: 30710044 DOI: 10.4049/jimmunol.1800366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022]
Abstract
Development of targeted cancer therapy requires a thorough understanding of mechanisms of tumorigenesis as well as mechanisms of action of therapeutics. This is challenging because by the time patients are diagnosed with cancer, early events of tumorigenesis have already taken place. Similarly, development of cancer immunotherapies is hampered by a lack of appropriate small animal models with autologous human tumor and immune system. In this article, we report the development of a mouse model of human acute myeloid leukemia (AML) with autologous immune system for studying early events of human leukemogenesis and testing the efficacy of immunotherapeutics. To develop such a model, human hematopoietic stem/progenitor cells (HSPC) are transduced with lentiviruses expressing a mutated form of nucleophosmin (NPM1), referred to as NPM1c. Following engraftment into immunodeficient mice, transduced HSPCs give rise to human myeloid leukemia, whereas untransduced HSPCs give rise to human immune cells in the same mice. The de novo AML, with CD123+ leukemic stem or initiating cells (LSC), resembles NPM1c+ AML from patients. Transcriptional analysis of LSC and leukemic cells confirms similarity of the de novo leukemia generated in mice with patient leukemia and suggests Myc as a co-operating factor in NPM1c-driven leukemogenesis. We show that a bispecific conjugate that binds both CD3 and CD123 eliminates CD123+ LSCs in a T cell-dependent manner both in vivo and in vitro. These results demonstrate the utility of the NPM1c+ AML model with an autologous immune system for studying early events of human leukemogenesis and for evaluating efficacy and mechanism of immunotherapeutics.
Collapse
Affiliation(s)
- Mandeep Kaur
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Adam C Drake
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Guangan Hu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Qingfeng Chen
- Institute for Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673
| | - Ryan Phennicie
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ricardo Attar
- Janssen Pharmaceuticals, Inc., Springhouse, PA 19477; and
| | - Jeffrey Nemeth
- Janssen Pharmaceuticals, Inc., Springhouse, PA 19477; and
| | | | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139; .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
35
|
Di Natale C, La Manna S, Malfitano AM, Di Somma S, Florio D, Scognamiglio PL, Novellino E, Netti PA, Marasco D. Structural insights into amyloid structures of the C-terminal region of nucleophosmin 1 in type A mutation of acute myeloid leukemia. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:637-644. [PMID: 30710643 DOI: 10.1016/j.bbapap.2019.01.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/11/2019] [Accepted: 01/26/2019] [Indexed: 12/22/2022]
Abstract
Acute myeloid leukemia (AML) is a clinically and a molecularly heterogeneous disease characterized by the accumulation of undifferentiated and uncontrolled proliferation of hematopoietic progenitor cells. The sub-group named "AML with gene mutations" includes mutations in nucleophosmin (NPM1) assumed as a distinct leukemic entity. NPM1 is an abundant multifunctional protein belonging to the nucleoplasmin family of nuclear chaperones. AML mutated protein is translocated into the cytoplasm (NPM1c+) retaining all functional domains except the loss of a unique NoLs (nucleolar localization signal) at the C-term domain (CTD) and the subsequent disruption of a three helix bundle as tertiary structure. The oligomeric state of NPM1 is of outmost importance for its biological roles and our previous studies linked an aggregation propensity of distinct regions of CTD to leukomogenic potentials of AML mutations. Here we investigated a polypeptide spanning the third and second helices of the bundle of type A mutated CTD. By a combination of several techniques, we ascertained the amyloid character of the aggregates and of fibrils resulting from a self-recognition mechanism. Further amyloid assemblies resulted cytoxic in MTT assay strengthening a new idea of a therapeutic strategy in AML consisting in the self-degradation of mutated NPM1.
Collapse
Affiliation(s)
- Concetta Di Natale
- Department of Pharmacy, University of Naples "Federico II", Italy; Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
| | - Sara La Manna
- Department of Pharmacy, University of Naples "Federico II", Italy
| | | | - Sarah Di Somma
- Department of Translational Medicine, University of Naples "Federico II", Italy
| | - Daniele Florio
- Department of Pharmacy, University of Naples "Federico II", Italy
| | | | - Ettore Novellino
- Department of Pharmacy, University of Naples "Federico II", Italy
| | - Paolo Antonio Netti
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
| | - Daniela Marasco
- Department of Pharmacy, University of Naples "Federico II", Italy.
| |
Collapse
|
36
|
Current Outlook on Autophagy in Human Leukemia: Foe in Cancer Stem Cells and Drug Resistance, Friend in New Therapeutic Interventions. Int J Mol Sci 2019; 20:ijms20030461. [PMID: 30678185 PMCID: PMC6387281 DOI: 10.3390/ijms20030461] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 01/07/2023] Open
Abstract
Autophagy is an evolutionarily conserved cellular recycling process in cell homeostasis and stress adaptation. It confers protection and promotes survival in response to metabolic/environmental stress, and is upregulated in response to nutrient deprivation, hypoxia, and chemotherapies. Autophagy is also known to sustain malignant cell growth and contributes to cancer stem cell survival when challenged by cytotoxic and/or targeted therapies, a potential mechanism of disease persistence and drug resistance that has gathered momentum. However, different types of human leukemia utilize autophagy in complex, context-specific manners, and the molecular and cellular mechanisms underlying this process involve multiple protein networks that will be discussed in this review. There is mounting preclinical evidence that targeting autophagy can enhance the efficacy of cancer therapies. Chloroquine and other lysosomal inhibitors have spurred initiation of clinical trials and demonstrated that inhibition of autophagy restores chemosensitivity of anticancer drugs, but with limited autophagy-dependent effects. Intriguingly, several autophagy-specific inhibitors, with better therapeutic indexes and lower toxicity, have been developed. Promising preclinical studies with novel combination approaches as well as potential challenges to effectively eradicate drug-resistant cells, particularly cancer stem cells, in human leukemia are also detailed in this review.
Collapse
|
37
|
Zhang W, Zhao C, Zhao J, Zhu Y, Weng X, Chen Q, Sun H, Mi JQ, Li J, Zhu J, Chen Z, Pandolfi PP, Chen S, Yan X, Xu J. Inactivation of PBX3 and HOXA9 by down-regulating H3K79 methylation represses NPM1-mutated leukemic cell survival. Am J Cancer Res 2018; 8:4359-4371. [PMID: 30214626 PMCID: PMC6134928 DOI: 10.7150/thno.26900] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/24/2018] [Indexed: 11/05/2022] Open
Abstract
Acute myeloid leukemia (AML) with an NPM1 mutation (NPMc+) has a distinct gene expression signature and displays molecular abnormalities similar to mixed lineage leukemia (MLL), including aberrant expression of the PBX3 and HOXA gene cluster. However, it is unclear if the aberrant expression of PBX3 and HOXA is essential for the survival of NPM1-mutated leukemic cells. Methods: Using the gene expression profiling of TCGA and E-MTAB-3444 datasets, we screened for high co-expression of PBX3 and HOXA9 in NPMc+ leukemia patients. We performed NPMc+ depletion and overexpression experiments to examine aberrant H3K79 methylation through epigenetic regulation. Through RNA interference technology and small-molecule inhibitor treatment, we evaluated the effect of methyl-modified H3K79 on cell survival and explored the possible underlying mechanism. Results: We showed that NPMc+ increased the expression of PBX3 and HOXA9, which are both poor prognosis indicators in AML. High PBX3 and HOXA9 expression was accompanied by increased dimethylated and trimethylated H3K79 in transgenic murine Lin-Sca-1+c-Kit+ cells and human NPMc+ leukemia cells. Using chromatin immunoprecipitation sequencing (ChIP-seq) assays of NPMc+ cells, we determined that hypermethylated H3K79 was present at the expressed HOXA9 gene but not the PBX3 gene. PBX3 expression was positively regulated by HOXA9, and a reduction in either PBX3 or HOXA9 resulted in NPMc+ cell apoptosis. Importantly, an inhibitor of DOT1L, EPZ5676, effectively and selectively promoted NPMc+ human leukemic cell apoptosis by reducing HOXA9 and PBX3 expression. Conclusion: Our data indicate that NPMc+ leukemic cell survival requires upregulation of PBX3 and HOXA9, and this action can be largely attenuated by a DOT1L inhibitor.
Collapse
|
38
|
Cosimato V, Scalia G, Raia M, Gentile L, Cerbone V, Visconte F, Statuto T, Valvano L, D'Auria F, Calice G, Graziano D, Musto P, Del Vecchio L. Surface endoglin (CD105) expression on acute leukemia blast cells: an extensive flow cytometry study of 1002 patients. Leuk Lymphoma 2018; 59:2242-2245. [PMID: 29334284 DOI: 10.1080/10428194.2017.1416366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | | | | | | | | | | | - Teodora Statuto
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Luciana Valvano
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Fiorella D'Auria
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Giovanni Calice
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Daniela Graziano
- c Antonio Cardarelli Hospital , Unit of Transfusional Medicine , Naples , Italy
| | - Pellegrino Musto
- d Scientific Direction, IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Luigi Del Vecchio
- a CEINGE Biotecnologie Avanzate , Naples , Italy.,e Department of Molecular Medicine and Medical Biotechnologies (DMMBM) , Federico II University , Naples , Italy
| |
Collapse
|
39
|
|
40
|
Draper JE, Sroczynska P, Fadlullah MZH, Patel R, Newton G, Breitwieser W, Kouskoff V, Lacaud G. A novel prospective isolation of murine fetal liver progenitors to study in utero hematopoietic defects. PLoS Genet 2018; 14:e1007127. [PMID: 29300724 PMCID: PMC5754050 DOI: 10.1371/journal.pgen.1007127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/26/2017] [Indexed: 12/29/2022] Open
Abstract
In recent years, highly detailed characterization of adult bone marrow (BM) myeloid progenitors has been achieved and, as a result, the impact of somatic defects on different hematopoietic lineage fate decisions can be precisely determined. Fetal liver (FL) hematopoietic progenitor cells (HPCs) are poorly characterized in comparison, potentially hindering the study of the impact of genetic alterations on midgestation hematopoiesis. Numerous disorders, for example infant acute leukemias, have in utero origins and their study would therefore benefit from the ability to isolate highly purified progenitor subsets. We previously demonstrated that a Runx1 distal promoter (P1)-GFP::proximal promoter (P2)-hCD4 dual-reporter mouse (Mus musculus) model can be used to identify adult BM progenitor subsets with distinct lineage preferences. In this study, we undertook the characterization of the expression of Runx1-P1-GFP and P2-hCD4 in FL. Expression of P2-hCD4 in the FL immunophenotypic Megakaryocyte-Erythroid Progenitor (MEP) and Common Myeloid Progenitor (CMP) compartments corresponded to increased granulocytic/monocytic/megakaryocytic and decreased erythroid specification. Moreover, Runx1-P2-hCD4 expression correlated with several endogenous cell surface markers' expression, including CD31 and CD45, providing a new strategy for prospective identification of highly purified fetal myeloid progenitors in transgenic mouse models. We utilized this methodology to compare the impact of the deletion of either total RUNX1 or RUNX1C alone and to determine the fetal HPCs lineages most substantially affected. This new prospective identification of FL progenitors therefore raises the prospect of identifying the underlying gene networks responsible with greater precision than previously possible.
Collapse
Affiliation(s)
- Julia E. Draper
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Patrycja Sroczynska
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Muhammad Z. H. Fadlullah
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Rahima Patel
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Gillian Newton
- Molecular Biology Core Facility, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Wolfgang Breitwieser
- Molecular Biology Core Facility, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Valerie Kouskoff
- Division of Developmental Biology & Medicine, Michael Smith Building, The University of Manchester, Manchester, United Kingdom
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
41
|
Bunaciu RP, MacDonald RJ, Gao F, Johnson LM, Varner JD, Wang X, Nataraj S, Guzman ML, Yen A. Potential for subsets of wt-NPM1 primary AML blasts to respond to retinoic acid treatment. Oncotarget 2017; 9:4134-4149. [PMID: 29423110 PMCID: PMC5790527 DOI: 10.18632/oncotarget.23642] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/09/2017] [Indexed: 01/16/2023] Open
Abstract
Acute myeloid leukemia (AML) has high mortality rates, perhaps reflecting a lack of understanding of the molecular diversity in various subtypes and a lack of known actionable targets. There are currently 12 open clinical trials for AML using combination therapeutic modalities including all-trans retinoic acid (RA). Mutant nucleophosmin-1, proposed as a possible marker for RA response, is the criterion for recruiting patients in three active RA phase 3 clinical trials. We tested the ability of RA alone or in combination with either bosutinib (B) or 6-formylindolo(3,2-b) carbazole (F) to induce conversion of 12 de novo AML samples toward a more differentiated phenotype. We assessed levels of expression of cell surface markers associated with differentiation, aldehyde dehydrogenase activity, and glucose uptake activity. Colony formation capacity was reduced with the combined treatment of RA and B or F, and correlated with modulation of a c-Cbl/Lyn/c-Raf-centered signalsome. Combination treatment was in most cases more effective than RA alone. Based on their responses to the treatments, some primary leukemic samples cluster closer to HL-60 cells than to other primary samples, suggesting that they may represent a hitherto undefined AML subtype that is potentially responsive to RA in a combination differentiation therapy.
Collapse
Affiliation(s)
- Rodica P Bunaciu
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | | | - Feng Gao
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA.,Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA.,Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Lynn M Johnson
- Cornell Statistical Unit, Cornell University, Ithaca, NY, USA
| | - Jeffrey D Varner
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Sarah Nataraj
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Monica L Guzman
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Andrew Yen
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| |
Collapse
|
42
|
Deguelin induced differentiation of mutated NPM1 acute myeloid leukemia in vivo and in vitro. Anticancer Drugs 2017; 28:723-738. [PMID: 28471807 DOI: 10.1097/cad.0000000000000494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Nucleophosmin (NPM1), a restricted nucleolar localization protein, shuttles between the nucleus and the cytoplasm. Mutated (Mt)-NPM1 protein, which has aberrant cytoplasmic dislocation of nucleophosmin, occurs in approximately one-third of acute myeloid leukemia cases. Deguelin, a rotenoid isolated from several plant species, is a strong antitumor agent. NOD/SCID mice xenografted with human Mt-NPM1 OCI/AML3 cell lines served as in-vivo models. Wright-Giemsa staining and flow cytometry analysis were used for differentiation assays. Associated molecular events were assessed by western blot and histological analyses. Kaplan-Meier estimates were used to calculate survival. Deguelin toxicity in mice was assessed by immunohistochemistry staining and serum markers. Clinical samples were differentiated by flow cytometry analysis. Deguelin induced differentiation by downregulating the Mt-NPM1 protein levels, which was accompanied by a decrease in SIRT1, p21, and HDAC1 and an increase in CEBPβ and granulocyte colony-stimulating factor receptor protein expression levels. A low-deguelin dose prolonged survival compared with the control group, and there were no apparent lesions to the brain, liver, heart, and kidney in vivo. In clinical samples, deguelin induced the differentiation of fresh blasts with Mt-NPM1 protein, but not with the wild-type NPM1 protein. Taken together, these findings further provide new evidence that the Mt-NPM1 protein plays an important role in inducing differentiation in vivo and in vitro. Mutated NPM1 protein may be a therapeutic target of deguelin in acute myeloid leukemia with the NPM1 mutation.
Collapse
|
43
|
Her Z, Yong KSM, Paramasivam K, Tan WWS, Chan XY, Tan SY, Liu M, Fan Y, Linn YC, Hui KM, Surana U, Chen Q. An improved pre-clinical patient-derived liquid xenograft mouse model for acute myeloid leukemia. J Hematol Oncol 2017; 10:162. [PMID: 28985760 PMCID: PMC5639594 DOI: 10.1186/s13045-017-0532-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 09/29/2017] [Indexed: 11/10/2022] Open
Abstract
Background Xenotransplantation of patient-derived AML (acute myeloid leukemia) cells in NOD-scid Il2rγnull (NSG) mice is the method of choice for evaluating this human hematologic malignancy. However, existing models constructed using intravenous injection in adult or newborn NSG mice have inferior engraftment efficiency, poor peripheral blood engraftment, or are difficult to construct. Methods Here, we describe an improved AML xenograft model where primary human AML cells were injected into NSG newborn pups intrahepatically. Results Introduction of primary cells from AML patients resulted in high levels of engraftment in peripheral blood, spleen, and bone marrow (BM) of recipient mice. The phenotype of engrafted AML cells remained unaltered during serial transplantation. The mice developed features that are consistent with human AML including spleen enlargement and infiltration of AML cells into multiple organs. Importantly, we demonstrated that although leukemic stem cell activity is enriched and mediated by CD34+CD117+ subpopulation, CD34+CD117− subpopulation can acquire CD34+CD117+ phenotype through de-differentiation. Lastly, we evaluated the therapeutic potential of Sorafenib and Regorafenib in this AML model and found that periphery and spleen AML cells are sensitive to these treatments, whereas BM provides a protective environment to AML. Conclusions Collectively, our improved model is robust, easy-to-construct, and reliable for pre-clinical AML studies. Electronic supplementary material The online version of this article (10.1186/s13045-017-0532-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhisheng Her
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Kylie Su Mei Yong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Kathirvel Paramasivam
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Wilson Wei Sheng Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Xue Ying Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Min Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yeh Ching Linn
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Kam Man Hui
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore.,Division of Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
| | - Uttam Surana
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore. .,Department of Pharmacology, National University of Singapore, Singapore, Singapore. .,Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore. .,Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China. .,Division of Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore. .,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
44
|
Chopra A, Soni S, Pati H, Kumar D, Diwedi R, Verma D, Vishwakama G, Bakhshi S, Kumar S, Gogia A, Kumar R. Nucleophosmin mutation analysis in acute myeloid leukaemia: Immunohistochemistry as a surrogate for molecular techniques. Indian J Med Res 2017; 143:763-768. [PMID: 27748301 PMCID: PMC5094116 DOI: 10.4103/0971-5916.192027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background & objectives: Mutation of nucleophosmin (NPM1) gene in the absence of FLT3-ITD (FMS related tyrosine kinase 3 - internal tandem duplications) mutation carries a good prognosis in cytogenetically normal acute myeloid leukaemia (AML). NPM1, a multifunctional nucleolar phosphoprotein that shuttles between nucleus and cytoplasm, gets trapped in the cytoplasm when mutated. Immunohistochemical (IHC) demonstration of its aberrant cytoplasmic location (NPMc+) has been suggested as a simple substitute for the standard screening molecular method. This study was aimed to assess the diagnostic utility of IHC on formalin fixed bone marrow biopsies in comparison with the reference molecular method (allele specific oligonucleotide - polymerase chain reaction; ASO-PCR) to predict NPM1 mutation status in AML patients. Methods: NPM protein IHC was performed using mouse anti-NPM monoclonal antibody on 35 paraffin-embedded bone marrow biopsies of patients with primary AML of any French-American-British (FAB) subtype. Results of IHC were compared with those of ASO-PCR. Results: Of the 35 AML patients, 21 (60%) were positive for NPM1 exon 12 gene mutation by ASO-PCR, 19 (90.47%) of these 21 were NPMc+. Thirteen of the 35 patients were negative by both the methods. One NPMc+ patient was not detected by ASO-PCR. IHC had a sensitivity and specificity of 90 and 93 per cent, respectively, compared to the molecular screening gold standard. Interpretation & conclusions: Mutation of NPM1 determined by the widely available and inexpensive IHC agrees closely with results of the standard molecular methods. Thus, technically and financially not well endowed laboratories can provide the prognostically and potentially therapeutically important information on NPM1 mutation using IHC.
Collapse
Affiliation(s)
- Anita Chopra
- Department of Laboratory Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sushant Soni
- Department of Laboratory Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Haraprasad Pati
- Department of Haematology, All India Institute of Medical Sciences, New Delhi, India
| | - Dev Kumar
- Department of Laboratory Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Rahul Diwedi
- Department of Laboratory Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Deepak Verma
- Department of Laboratory Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Garima Vishwakama
- Department of Laboratory Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Suman Kumar
- Command Hospital (Eastern Command), Kolkata, India
| | - Ajay Gogia
- Department of Medical Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Rajive Kumar
- Department of Laboratory Oncology, Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
45
|
Biology and relevance of human acute myeloid leukemia stem cells. Blood 2017; 129:1577-1585. [PMID: 28159741 DOI: 10.1182/blood-2016-10-696054] [Citation(s) in RCA: 332] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022] Open
Abstract
Evidence of human acute myeloid leukemia stem cells (AML LSCs) was first reported nearly 2 decades ago through the identification of rare subpopulations of engrafting cells in xenotransplantation assays. These AML LSCs were shown to reside at the apex of a cellular hierarchy that initiates and maintains the disease, exhibiting properties of self-renewal, cell cycle quiescence, and chemoresistance. This cancer stem cell model offers an explanation for chemotherapy resistance and disease relapse and implies that approaches to treatment must eradicate LSCs for cure. More recently, a number of studies have both refined and expanded our understanding of LSCs and intrapatient heterogeneity in AML using improved xenotransplant models, genome-scale analyses, and experimental manipulation of primary patient cells. Here, we review these studies with a focus on the immunophenotype, biological properties, epigenetics, genetics, and clinical associations of human AML LSCs and discuss critical questions that need to be addressed in future research.
Collapse
|
46
|
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
|
47
|
Yanagisawa B, Ghiaur G, Smith BD, Jones RJ. Translating leukemia stem cells into the clinical setting: Harmonizing the heterogeneity. Exp Hematol 2016; 44:1130-1137. [PMID: 27693555 PMCID: PMC5110366 DOI: 10.1016/j.exphem.2016.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/23/2016] [Indexed: 01/01/2023]
Abstract
Considerable evidence suggests that rare leukemia cells with stem cell features, including self-renewal capacity and drug resistance, are primarily responsible for both disease maintenance and relapses. Traditionally, these so-called leukemia stem cells (LSCs) have been identified in the laboratory by their ability to engraft acute myeloid leukemia (AML) into immunocompromised mice. For many years, only those rare AML cells characterized by a hematopoietic stem cell (HSC) CD34+CD38- phenotype were believed capable of generating leukemia in immunocompromised mice. However, more recently, significant heterogeneity in the phenotypes of those AML cells that can engraft immunocompromised mice has been demonstrated. AML cells that engraft immunocompromised mice have also been shown to not necessarily represent either the founder clone or those cells responsible for relapse. A recent study found that the most immature phenotype present in an AML correlated with genetically defined risk groups and outcomes, but was heterogeneous. Patients with AML cells expressing a primitive HSC phenotype (CD34+CD38- with high aldehyde dehydrogenase activity) manifested significantly lower complete remission rates, as well as poorer event-free and overall survivals. Leukemias in which the most primitive cells displayed more mature phenotypes were associated with better outcomes. The strong clinical correlations suggest that the most immature phenotype detectable within a patient's AML might serve as a biomarker for "clinically relevant" LSCs.
Collapse
Affiliation(s)
- Breann Yanagisawa
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Gabriel Ghiaur
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - B Douglas Smith
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Richard J Jones
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
48
|
Ma HS, Robinson TM, Small D. Potential role for all- trans retinoic acid in nonpromyelocytic acute myeloid leukemia. Int J Hematol Oncol 2016; 5:133-142. [PMID: 30302214 DOI: 10.2217/ijh-2016-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/08/2017] [Indexed: 11/21/2022] Open
Abstract
All-trans retinoic acid (ATRA) has been very successful in the subtype of acute myelogenous leukemia known as acute promyelocytic leukemia due to targeted reactivation of retinoic acid signaling. There has been great interest in applying this form of differentiation therapy to other cancers, and numerous clinical trials have been initiated. However, ATRA as monotherapy has thus far shown little benefit in nonacute promyelocytic leukemia acute myelogenous leukemia. Here, we review the literature on the use of ATRA in combination with chemotherapy, epigenetic modifying agents and targeted therapy, highlighting specific patient populations where the addition of ATRA to existing therapies may provide benefit. Furthermore, we discuss the impact of recent whole genome sequencing efforts in leading the design of rational combinatorial approaches.
Collapse
Affiliation(s)
- Hayley S Ma
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Tara M Robinson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Donald Small
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
49
|
Laranjeira ABA, Yang SX. Therapeutic target discovery and drug development in cancer stem cells for leukemia and lymphoma: from bench to the clinic. Expert Opin Drug Discov 2016; 11:1071-1080. [PMID: 27626707 DOI: 10.1080/17460441.2016.1236785] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Cancer stem cells (CSCs), also known as tumor initialing cells, have self-renewal capacity and are believed to play an important role in residual disease or tumor relapse. CSCs exhibit characteristic slow growth rate and are resistant to conventional chemotherapy/radiotherapy in experimental models. The type of cells commonly employs aberrant activity of the embryonic signal transduction pathways - Notch, Hedgehog (Hh), and Wnt - for uncontrolled proliferation and survival. Areas covered: The following article discusses key genetic and molecular alterations in Notch, Hh and Wnt pathways and drugs targeting the alterations for the treatment of leukemia and lymphoma. Expert opinion: Early signs of signal agent activity have been observed in certain types of leukemia and lymphoma with experimental therapeutics targeting the embryonic pathways in the CSC signaling network. However, clinical development of agents that inhibit the Wnt/β-catenin, Notch and Hh signaling appear to be more complex in relapsed or refractory malignancies. A strategy to effectively target signaling may rely on early application of biomarkers representative of the active signaling nodes companion to the molecularly targeted agents. Biomarkers for efficacy could potentially guide selective treatment of hematological malignancies or cancer with drugs that target the embryonic pathways.
Collapse
Affiliation(s)
- Angelo B A Laranjeira
- a National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Sherry X Yang
- a National Clinical Target Validation Laboratory, Division of Cancer Treatment and Diagnosis , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
50
|
Quek L, Otto GW, Garnett C, Lhermitte L, Karamitros D, Stoilova B, Lau IJ, Doondeea J, Usukhbayar B, Kennedy A, Metzner M, Goardon N, Ivey A, Allen C, Gale R, Davies B, Sternberg A, Killick S, Hunter H, Cahalin P, Price A, Carr A, Griffiths M, Virgo P, Mackinnon S, Grimwade D, Freeman S, Russell N, Craddock C, Mead A, Peniket A, Porcher C, Vyas P. Genetically distinct leukemic stem cells in human CD34- acute myeloid leukemia are arrested at a hemopoietic precursor-like stage. J Exp Med 2016; 213:1513-35. [PMID: 27377587 PMCID: PMC4986529 DOI: 10.1084/jem.20151775] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/19/2016] [Indexed: 12/16/2022] Open
Abstract
Quek and colleagues identify human leukemic stem cells (LSCs) present in CD34− AML. In-depth characterization of the functional and clonal aspects of CD34− LSCs indicates that most are similar to myeloid precursors. Our understanding of the perturbation of normal cellular differentiation hierarchies to create tumor-propagating stem cell populations is incomplete. In human acute myeloid leukemia (AML), current models suggest transformation creates leukemic stem cell (LSC) populations arrested at a progenitor-like stage expressing cell surface CD34. We show that in ∼25% of AML, with a distinct genetic mutation pattern where >98% of cells are CD34−, there are multiple, nonhierarchically arranged CD34+ and CD34− LSC populations. Within CD34− and CD34+ LSC–containing populations, LSC frequencies are similar; there are shared clonal structures and near-identical transcriptional signatures. CD34− LSCs have disordered global transcription profiles, but these profiles are enriched for transcriptional signatures of normal CD34− mature granulocyte–macrophage precursors, downstream of progenitors. But unlike mature precursors, LSCs express multiple normal stem cell transcriptional regulators previously implicated in LSC function. This suggests a new refined model of the relationship between LSCs and normal hemopoiesis in which the nature of genetic/epigenetic changes determines the disordered transcriptional program, resulting in LSC differentiation arrest at stages that are most like either progenitor or precursor stages of hemopoiesis.
Collapse
Affiliation(s)
- Lynn Quek
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK Department of Hematology, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| | - Georg W Otto
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Catherine Garnett
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Ludovic Lhermitte
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Dimitris Karamitros
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Bilyana Stoilova
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - I-Jun Lau
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK Department of Hematology, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| | - Jessica Doondeea
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Batchimeg Usukhbayar
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Alison Kennedy
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Marlen Metzner
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Nicolas Goardon
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Adam Ivey
- Department of Genetics, King's College London, London WC2R 2LS, England, UK
| | - Christopher Allen
- Cancer Institute, University College London, London WC1E 6BT, England, UK
| | - Rosemary Gale
- Cancer Institute, University College London, London WC1E 6BT, England, UK
| | - Benjamin Davies
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| | - Alexander Sternberg
- Department of Hematology, Great Western Hospital National Health Service Foundation Trust, Swindon SN3 6BB, England, UK
| | - Sally Killick
- Department of Hematology, Royal Bournemouth and Christchurch Hospital National Health Service Trust, Bournemouth BH7 7DW, England, UK
| | - Hannah Hunter
- Department of Hematology, Plymouth Hospitals National Health Service Trust, Plymouth PL6 8DH, England, UK
| | - Paul Cahalin
- Department of Hematology, Blackpool, Fylde and Wyre Hospitals National Health Service Trust, Blackpool FY3 8NR, England, UK
| | - Andrew Price
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| | - Andrew Carr
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| | - Mike Griffiths
- West Midlands Regional Genetics Laboratory, Birmingham B15 2TG, England, UK
| | - Paul Virgo
- Department of Immunology, North Bristol National Health Service Trust, Bristol BS10 5NB, England, UK
| | - Stephen Mackinnon
- Cancer Institute, University College London, London WC1E 6BT, England, UK Department of Hematology, University College London Hospital National Health Service Foundation Trust, London NW1 2BU, England, UK
| | - David Grimwade
- Department of Genetics, King's College London, London WC2R 2LS, England, UK
| | - Sylvie Freeman
- School of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, England, UK Department of Haematology, University Hospitals Birmingham National Health Service Foundation Trust, Birmingham B15 2TH, England, UK
| | - Nigel Russell
- Centre for Clinical Hematology, Nottingham University Hospitals National Health Service Trust, Nottingham NG5 1PB, England, UK
| | - Charles Craddock
- Department of Clinical Haematology, University of Birmingham, Birmingham B15 2TT, England, UK Department of Clinical Haematology, University Hospitals Birmingham National Health Service Foundation Trust, Birmingham B15 2TH, England, UK
| | - Adam Mead
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK Department of Hematology, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| | - Andrew Peniket
- Department of Hematology, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| | - Catherine Porcher
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK
| | - Paresh Vyas
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX1 2JD, England, UK Department of Hematology, Oxford University Hospital National Health Service Trust, Oxford OX3 9DU, England, UK
| |
Collapse
|