51
|
Pan X, Gao M, Sun Y, Zhou Y, Wang K, Wang Y, Xu L, Zhang X, Huang X, Zhao X. Significance of WT1 and multiparameter flow cytometry assessment in patients with chronic myelomonocytic leukemia receiving allogeneic hematopoietic stem cell transplantation. Int J Lab Hematol 2022; 44:510-517. [DOI: 10.1111/ijlh.13788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/10/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Xinan Pan
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Mengge Gao
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Yuqian Sun
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Yang Zhou
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Ke Wang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Yu Wang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Lanping Xu
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Xiaohui Zhang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Xiaojun Huang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
- Peking‐Tsinghua Center for Life Sciences Beijing China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies Chinese Academy of Medical Sciences Beijing China
| | - Xiao‐Su Zhao
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies Chinese Academy of Medical Sciences Beijing China
| |
Collapse
|
52
|
Emergence of clone with PHF6 nonsense mutation in chronic myelomonocytic leukemia at relapse after allogeneic HCT. Int J Hematol 2022; 115:748-752. [PMID: 34988909 DOI: 10.1007/s12185-021-03284-7] [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: 09/10/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 10/19/2022]
Abstract
Disease relapse is a major cause of treatment failure after allogeneic hematopoietic cell transplantation (HCT) and the mechanisms of relapse remain unclear. We encountered a 58-year-old man with chronic myelomonocytic leukemia (CMML) that relapsed after haploidentical HCT from his daughter. Peripheral blood samples collected at HCT and at relapse were analyzed, and CD14+/CD16- monocytes that typically accumulate in CMML were isolated by flow cytometry. Whole-exome sequencing of the monocytes revealed 8 common mutations in CMML at HCT. In addition, a PHF6 nonsense mutation not detected at HCT was detected at relapse. RNA sequencing could not detect changes in expression of HLA or immune-checkpoint molecules, which are important mechanisms of immune evasion. However, gene set enrichment analysis (GSEA) revealed that a TNF-α signaling pathway was downregulated at relapse. Ubiquitination of histone H2B at lysine residue 120 (H2BK120ub) at relapse was significantly decreased at the protein level, indicating that PHF6 loss might downregulate a TNF-α signaling pathway by reduction of H2BK120ub. This case illustrates that PHF6 loss contributes to a competitive advantage for the clone under stress conditions and leads to relapse after HCT.
Collapse
|
53
|
Sorigue M, Arenillas L, Xicoy B, Andrade-Campos M, Navarro JT, Ferrer A, Zamora L, Calvo X. Monocyte subset distribution in myeloproliferative and myelodysplastic/myeloproliferative neoplasms with monocytosis. Leuk Res 2021; 112:106771. [PMID: 34902713 DOI: 10.1016/j.leukres.2021.106771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/16/2021] [Accepted: 12/05/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Marc Sorigue
- Department of Hematology, ICO-Badalona, Hospital Germans Trias i Pujol, Functional Cytomics- IJC, LCMN, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Leonor Arenillas
- Laboratori de Citologia Hematològica, Servei de Patologia, Grup de Recerca Translacional en Neoplàsies Hematològiques (GRETNHE), IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Blanca Xicoy
- Department of Hematology, ICO-Badalona, Hospital Germans Trias i Pujol, IJC, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Marcio Andrade-Campos
- Servei d'Hematologia Clínica, Grup de Recerca Clínica Aplicada en Neoplàsies Hematològiques, IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Jose-Tomas Navarro
- Department of Hematology, ICO-Badalona, Hospital Germans Trias i Pujol, IJC, LCMN, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Ana Ferrer
- Laboratori de Citologia Hematològica, Servei de Patologia, Grup de Recerca Translacional en Neoplàsies Hematològiques (GRETNHE), IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Lurdes Zamora
- Department of Hematology, ICO-Badalona, Hospital Germans Trias i Pujol, IJC, LCMN, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Xavier Calvo
- Laboratori de Citologia Hematològica, Servei de Patologia, Grup de Recerca Translacional en Neoplàsies Hematològiques (GRETNHE), IMIM Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
54
|
Eisenwort G, Sadovnik I, Keller A, Ivanov D, Peter B, Berger D, Stefanzl G, Bauer K, Slavnitsch K, Greiner G, Gleixner KV, Sperr WR, Willmann M, Sill H, Bettelheim P, Geissler K, Deininger M, Rülicke T, Valent P. Phenotypic characterization of leukemia-initiating stem cells in chronic myelomonocytic leukemia. Leukemia 2021; 35:3176-3187. [PMID: 33785864 PMCID: PMC7611912 DOI: 10.1038/s41375-021-01227-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/04/2021] [Accepted: 03/11/2021] [Indexed: 12/27/2022]
Abstract
Chronic myelomonocytic leukemia (CMML) is a stem cell-derived neoplasm characterized by dysplasia, uncontrolled expansion of monocytes, and substantial risk to transform to secondary acute myeloid leukemia (sAML). So far, little is known about CMML-initiating cells. We found that leukemic stem cells (LSC) in CMML reside in a CD34+/CD38- fraction of the malignant clone. Whereas CD34+/CD38- cells engrafted NSGS mice with overt CMML, no CMML was produced by CD34+/CD38+ progenitors or the bulk of CD34- monocytes. CMML LSC invariably expressed CD33, CD117, CD123 and CD133. In a subset of patients, CMML LSC also displayed CD52, IL-1RAP and/or CLL-1. CMML LSC did not express CD25 or CD26. However, in sAML following CMML, the LSC also expressed CD25 and high levels of CD114, CD123 and IL-1RAP. No correlations between LSC phenotypes, CMML-variant, mutation-profiles, or clinical course were identified. Pre-incubation of CMML LSC with gemtuzumab-ozogamicin or venetoclax resulted in decreased growth and impaired engraftment in NSGS mice. Together, CMML LSC are CD34+/CD38- cells that express a distinct profile of surface markers and target-antigens. During progression to sAML, LSC acquire or upregulate certain cytokine receptors, including CD25, CD114 and CD123. Characterization of CMML LSC should facilitate their enrichment and the development of LSC-eradicating therapies.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Antigens, CD34/immunology
- Antigens, CD34/metabolism
- Apoptosis
- Case-Control Studies
- Cell Proliferation
- Female
- Humans
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelomonocytic, Chronic/complications
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Middle Aged
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Phenotype
- Prognosis
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Gregor Eisenwort
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Irina Sadovnik
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Alexandra Keller
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Daniel Ivanov
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Barbara Peter
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Daniela Berger
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Stefanzl
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Karin Bauer
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Katharina Slavnitsch
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Georg Greiner
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Ihr Labor, Medical Diagnostic Laboratories, Vienna, Austria
| | - Karoline V Gleixner
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Michael Willmann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department for Companion Animals and Horses, Clinic for Internal Medicine, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | | | - Klaus Geissler
- Medical School, Sigmund Freud University, Vienna, Austria
| | - Michael Deininger
- Division of Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Thomas Rülicke
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Peter Valent
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
55
|
Abstract
Chronic myelomonocytic leukemia (CMML) is a rare and challenging type of myeloproliferative neoplasm. Poor prognosis and high mortality, associated predominantly with progression to secondary acute myeloid leukemia (sAML), is still an unsolved problem. Despite a growing body of knowledge about the molecular repertoire of this disease, at present, the prognostic significance of CMML-associated mutations is controversial. The absence of available CMML cell lines and the small number of patients with CMML make pre-clinical testing and clinical trials complicated. Currently, specific therapy for CMML has not been approved; most of the currently available therapeutic approaches are based on myelodysplastic syndrome (MDS) and other myeloproliferative neoplasm (MNP) studies. In this regard, the development of the robust CMML animal models is currently the focus of interest. This review describes important studies concerning animal models of CMML, examples of methodological approaches, and the obtained hematologic phenotypes.
Collapse
|
56
|
Gallì A, Todisco G, Catamo E, Sala C, Elena C, Pozzi S, Bono E, Ferretti VV, Rizzo E, Molteni E, Zibellini S, Sarchi M, Boveri E, Ferrari J, Fiorelli N, Camaschella C, Gasparini P, Toniolo D, Cazzola M, Malcovati L. Relationship between clone metrics and clinical outcome in clonal cytopenia. Blood 2021; 138:965-976. [PMID: 34255818 DOI: 10.1182/blood.2021011323] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/21/2021] [Indexed: 01/05/2023] Open
Abstract
Clonal cytopenia of undetermined significance (CCUS) is associated with an increased risk of developing a myeloid neoplasm with myelodysplasia (MN). To identify the features of the mutant clone(s) that is associated with clinical phenotype and progression, we studied the following cohorts of individuals: 311 patients with idiopathic cytopenia of undetermined significance (ICUS), 532 community-dwelling individuals without hematologic phenotype (n = 355) or with unexplained anemia (n = 177), and 592 patients with overt MN. Ninety-two of 311 (30%) patients with ICUS carried a somatic genetic lesion that signaled CCUS. Clonal hematopoiesis (CH) was detected in 19.7% and 27.7% of nonanemic and anemic community-dwelling individuals, respectively. Different mutation patterns and variant allele frequencies (VAFs) (clone metrics parameters) were observed in the conditions studied. Recurrent mutation patterns exhibited different VAFs associated with marrow dysplasia (0.17-0.48), indicating variable clinical expressivity of mutant clones. Unsupervised clustering analysis based on mutation profiles identified 2 major clusters, characterized by isolated DNMT3A mutations (CH-like cluster) or combinatorial mutation patterns (MN-like cluster), and showing different overall survival (HR, 1.8). In patients with CCUS, the 2 clusters had different risk of progression to MN (HR, 2.7). Within the MN-like cluster, distinct subsets with different risk of progression to MN were identified based on clone metrics. These findings unveil marked variability in the clinical expressivity of myeloid driver genes and underline the limitations of morphologic dysplasia for clinical staging of mutant hematopoietic clones. Clone metrics appears to be critical for informing clinical decision-making in patients with clonal cytopenia.
Collapse
Affiliation(s)
- Anna Gallì
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
| | - Gabriele Todisco
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eulalia Catamo
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Elena
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
| | - Sara Pozzi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Elisa Bono
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | | | | | - Silvia Zibellini
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
| | - Martina Sarchi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Emanuela Boveri
- Department of Pathology, IRCCS Fondazione Policlinico San Matteo, Pavia, Italy
| | - Jacqueline Ferrari
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Nicolas Fiorelli
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Gasparini
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Mario Cazzola
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Luca Malcovati
- Department of Hematology Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
57
|
Romano A, Giusti M, Di Giorgio M, Lumera G, Laura Parrinello N, Cosentino S, Ippolito M, Villari L, Alberto Palumbo G, Di Raimondo F, Santo Signorelli S. The first description of a singular case of synchronous chronic myelomonocytic leukemia and diffuse large b-cell lymphoma. Clin Case Rep 2021; 9:e03817. [PMID: 34589219 PMCID: PMC8458837 DOI: 10.1002/ccr3.3817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/08/2020] [Accepted: 11/18/2020] [Indexed: 11/06/2022] Open
Abstract
In CMML, neoplastic monocytes can be distinguished based on their immunophenotype. Supportive care myeloid growth factors in concomitant extranodal non-Hodgkin Lymphoma are safe.
Collapse
Affiliation(s)
- Alessandra Romano
- Dipartimento di Chirurgia e Specialità Medico ChirurgicheSezione di EmatologiaUniversità degli Studi di CataniaCataniaItaly
| | - Michele Giusti
- Department of Clinical and Experimental MedicineUniversità degli Studi di CataniaCataniaItaly
- UO Medicina GeneraleAOU Policlinico di Catania, Presidio RodolicoCataniaItaly
| | | | - Giovanni Lumera
- Department of Clinical and Experimental MedicineUniversità degli Studi di CataniaCataniaItaly
- UO Medicina GeneraleAOU Policlinico di Catania, Presidio RodolicoCataniaItaly
| | | | - Sebastiano Cosentino
- Dipartimento Tecnologie AvanzateUOC Medicina Nucleare ‐ Centro PETAOE, “Cannizzaro” CataniaCataniaItaly
| | - Massimo Ippolito
- Dipartimento Tecnologie AvanzateUOC Medicina Nucleare ‐ Centro PETAOE, “Cannizzaro” CataniaCataniaItaly
| | - Loredana Villari
- UO Anatomia PatologicaAOU Policlinico di Catania, Presidio San MarcoCataniaItaly
| | - Giuseppe Alberto Palumbo
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”Sezione di EmatologiaUniversità degli Studi di CataniaCataniaItaly
| | - Francesco Di Raimondo
- Dipartimento di Chirurgia e Specialità Medico ChirurgicheSezione di EmatologiaUniversità degli Studi di CataniaCataniaItaly
- UOC EmatologiaAOU Policlinico di Catania, Presidio RodolicoCataniaItaly
| | - Salvatore Santo Signorelli
- Department of Clinical and Experimental MedicineUniversità degli Studi di CataniaCataniaItaly
- UO Medicina GeneraleAOU Policlinico di Catania, Presidio RodolicoCataniaItaly
| |
Collapse
|
58
|
Ferrone CK, Wong H, Semenuk L, Werunga B, Snetsinger B, Zhang X, Zhang G, Lui J, Richard-Carpentier G, Crocker S, Good D, Hay AE, Quest G, Carson N, Feilotter HE, Rauh MJ. Validation, Implementation, and Clinical Impact of the Oncomine Myeloid Targeted-Amplicon DNA and RNA Ion Semiconductor Sequencing Assay. J Mol Diagn 2021; 23:1292-1305. [PMID: 34365012 DOI: 10.1016/j.jmoldx.2021.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 05/04/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022] Open
Abstract
The identification of clinically significant genes recurrently mutated in myeloid malignancies necessitates expanding diagnostic testing with higher throughput, such as targeted next-generation sequencing. We present validation of the Thermo Fisher Oncomine Myeloid Next-Generation Sequencing Panel (OMP), targeting 40 genes and 29 fusion drivers recurrently mutated in myeloid malignancies. The study includes data from a sample exchange between two Canadian hospitals demonstrating high concordance for detection of DNA and RNA aberrations. Clinical validation demonstrates high accuracy, sensitivity, and specificity of the OMP, with a lower limit of detection of 5% for single-nucleotide variants and 10% for insertions/deletions. Prospective sequencing was performed for 187 samples from 168 unique patients presenting with suspected or confirmed myeloid malignancy and other hematological conditions to assess clinical impact of identifying variants. Of detected variants, 48% facilitated or clarified diagnoses, 29% affected prognoses, and 25% had the potential to influence clinical management. Of note, OMP was essential to identifying patients with premalignant clonal states likely contributing to cytopenias. We also found that the detection of even a single variant by the OMP assay, versus 0 variants, was predictive of overall survival, independent of age, sex, or diagnosis (P = 0.03). This study demonstrates that molecular profiling of myeloid malignancies with the OMP represents a promising strategy to advance molecular diagnostics.
Collapse
Affiliation(s)
- Christina K Ferrone
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Henry Wong
- Molecular Genetics Laboratory, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Laura Semenuk
- Molecular Genetics Laboratory, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Barnaba Werunga
- Division of Genetics, Department of Lab Medicine and Pathology, Saint John Regional Hospital, Saint John, New Brunswick, Canada
| | - Brooke Snetsinger
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Xiao Zhang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Grace Zhang
- Division of Hematology, Department of Medicine, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Janet Lui
- Division of Hematology, Department of Medicine, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | | | - Susan Crocker
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada; Cytogenetics Laboratory, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - David Good
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Annette E Hay
- Division of Hematology, Department of Medicine, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Graeme Quest
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Nancy Carson
- Division of Genetics, Department of Lab Medicine and Pathology, Saint John Regional Hospital, Saint John, New Brunswick, Canada
| | - Harriet E Feilotter
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada; Molecular Genetics Laboratory, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Michael J Rauh
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
59
|
Hochman MJ, Savani BN, Jain T. Examining disease boundaries: Genetics of myelodysplastic/myeloproliferative neoplasms. EJHAEM 2021; 2:607-615. [PMID: 35844680 PMCID: PMC9175746 DOI: 10.1002/jha2.264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) are clonal myeloid malignancies that are characterized by dysplasia resulting in cytopenias as well as proliferative features such as thrombocytosis or splenomegaly. Recent studies have better defined the genetics underlying this diverse group of disorders. Trisomy 8, monosomy 7, and loss of Y chromosome are the most common cytogenetic abnormalities seen. Chronic myelomonocytic leukemia (CMML) likely develops from early clones with TET2 mutations that drive granulomonocytic differentiation. Mutations in SRSF2 are common and those in the RAS-MAPK pathway are typically implicated in disease with a proliferative phenotype. Several prognostic systems have incorporated genetic features, with ASXL1 most consistently demonstrating worse prognosis. Atypical chronic myeloid leukemia (aCML) is most known for granulocytosis with marked dysplasia and often harbors ASXL1 mutations, but SETBP1 and ETNK1 are more specific to this disease. MDS/MPN with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T) most commonly involves spliceosome mutations (namely SF3B1) and mutations in the JAK-STAT pathway. Finally, MDS/MPN-unclassifiable (MDS/MPN-U) is least characterized but a significant fraction carries mutations in TP53. The remaining patients have clinical and/or genetic features similar to the other MDS/MPNs, suggesting there is room to better characterize this entity. Evolution from age-related clonal hematopoiesis to MDS/MPN likely depends on the order of mutation acquisition and interactions between various biologic factors. Genetics will continue to play a critical role in our understanding of these illnesses and advancing patient care.
Collapse
Affiliation(s)
- Michael J. Hochman
- Division of Hematological Malignancies and Bone Marrow TransplantationSidney Kimmel Comprehensive Cancer CenterJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Bipin N. Savani
- Division of Hematology and OncologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow TransplantationSidney Kimmel Comprehensive Cancer CenterJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
60
|
Co-mutation pattern, clonal hierarchy, and clone size concur to determine disease phenotype of SRSF2 P95-mutated neoplasms. Leukemia 2021; 35:2371-2381. [PMID: 33349666 DOI: 10.1038/s41375-020-01106-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 10/29/2020] [Accepted: 11/27/2020] [Indexed: 01/29/2023]
Abstract
Somatic mutations in splicing factor genes frequently occur in myeloid neoplasms. While SF3B1 mutations are associated with myelodysplastic syndromes (MDS) with ring sideroblasts, SRSF2P95 mutations are found in different disease categories, including MDS, myeloproliferative neoplasms (MPN), myelodysplastic/myeloproliferative neoplasms (MDS/MPN), and acute myeloid leukemia (AML). To identify molecular determinants of this phenotypic heterogeneity, we explored molecular and clinical features of a prospective cohort of 279 SRSF2P95-mutated cases selected from a population of 2663 patients with myeloid neoplasms. Median number of somatic mutations per subject was 3. Multivariate regression analysis showed associations between co-mutated genes and clinical phenotype, including JAK2 or MPL with myelofibrosis (OR = 26.9); TET2 with monocytosis (OR = 5.2); RAS-pathway genes with leukocytosis (OR = 5.1); and STAG2, RUNX1, or IDH1/2 with blast phenotype (MDS or AML) (OR = 3.4, 1.9, and 2.1, respectively). Within patients with SRSF2-JAK2 co-mutation, JAK2 dominance was invariably associated with clinical feature of MPN, whereas SRSF2 mutation was dominant in MDS/MPN. Within patients with SRSF2-TET2 co-mutation, clinical expressivity of monocytosis was positively associated with co-mutated clone size. This study provides evidence that co-mutation pattern, clone size, and hierarchy concur to determine clinical phenotype, tracing relevant genotype-phenotype associations across disease entities and giving insight on unaccountable clinical heterogeneity within current WHO classification categories.
Collapse
|
61
|
Kaehler M, Cascorbi I. Pharmacogenomics of Impaired Tyrosine Kinase Inhibitor Response: Lessons Learned From Chronic Myelogenous Leukemia. Front Pharmacol 2021; 12:696960. [PMID: 34262462 PMCID: PMC8273252 DOI: 10.3389/fphar.2021.696960] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
The use of small molecules became one key cornerstone of targeted anti-cancer therapy. Among them, tyrosine kinase inhibitors (TKIs) are especially important, as they were the first molecules to proof the concept of targeted anti-cancer treatment. Since 2001, TKIs can be successfully used to treat chronic myelogenous leukemia (CML). CML is a hematologic neoplasm, predominantly caused by reciprocal translocation t(9;22)(q34;q11) leading to formation of the so-called BCR-ABL1 fusion gene. By binding to the BCR-ABL1 kinase and inhibition of downstream target phosphorylation, TKIs, such as imatinib or nilotinib, can be used as single agents to treat CML patients resulting in 80 % 10-year survival rates. However, treatment failure can be observed in 20-25 % of CML patients occurring either dependent or independent from the BCR-ABL1 kinase. Here, we review approved TKIs that are indicated for the treatment of CML, their side effects and limitations. We point out mechanisms of TKI resistance focusing either on BCR-ABL1-dependent mechanisms by summarizing the clinically observed BCR-ABL1-mutations and their implications on TKI binding, as well as on BCR-ABL1-independent mechanisms of resistances. For the latter, we discuss potential mechanisms, among them cytochrome P450 implications, drug efflux transporter variants and expression, microRNA deregulation, as well as the role of alternative signaling pathways. Further, we give insights on how TKI resistance could be analyzed and what could be learned from studying TKI resistance in CML in vitro.
Collapse
Affiliation(s)
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
62
|
Machherndl-Spandl S, Jäger E, Barna A, Gurbisz M, Marschon R, Graf T, Graf E, Geissler C, Hoermann G, Nösslinger T, Pfeilstöcker M, Bettelheim P, Zach O, Weltermann A, Heibl S, Thaler J, Zebisch A, Sill H, Stauder R, Webersinke G, Kusec R, Ulsperger E, Schneeweiss B, Öhler L, Germing U, Valent P, Tüchler H, Geissler K. Impact of age on the cumulative risk of transformation in patients with chronic myelomonocytic leukaemia. Eur J Haematol 2021; 107:265-274. [PMID: 33998054 PMCID: PMC8480146 DOI: 10.1111/ejh.13647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 12/19/2022]
Abstract
In older patients with chronic myelomonocytic leukaemia (CMML) and limited life expectancy due to age and or comorbidities, it is particularly important to consider the risk of transformation for individualised treatment decisions. There is limited information on potential differences between younger and older CMML patients regarding the cumulative risk of transformation as well as haematological, molecular and biologic characteristics. We analysed data from the Austrian Biodatabase for CMML (ABCMML) to compare these parameters in 518 CMML patients. Categorisation of patients into 3 age-related groups: <60 years, 60-79 years and ≥80 years, showed a significantly lower risk of transformation at higher age by competing risk analysis, with a 4-year risk of 39%, 23% and 13%, respectively (P < .0001). The lower probability of transformation was associated with a lower percentage of blast cells in the peripheral blood (PB) of older patients. Furthermore, we provide a simple score based on age, PB blasts and platelet counts that allowed us to define subgroups of CMML patients with a different cumulative transformation risk, including a low-risk group with a transformation risk of only 5%. Our findings may facilitate reasonable treatment decisions in elderly patients with CMML.
Collapse
Affiliation(s)
- Sigrid Machherndl-Spandl
- Department of Internal Medicine I with Hematology, Stem Cell Transplantation, Hemostasis and Medical Oncology, Ordensklinikum Elisabethinen Hospital, Linz, Austria
| | - Eva Jäger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Agnes Barna
- Blood Transfusion Service, Blood Transfusion Service for Upper Austria, Austrian Red Cross, Linz, Austria
| | - Michael Gurbisz
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Renate Marschon
- Laboratory for molecular and genetic diagnostics, Ordensklinikum Linz, Linz, Austria
| | - Temeida Graf
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, Vienna, Austria
| | - Elmir Graf
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, Vienna, Austria
| | | | - Gregor Hoermann
- MLL Munich Leukemia Laboratory, Munich, Germany.,Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria
| | - Thomas Nösslinger
- Department of Internal Medicine III, Hanusch Hospital, Vienna, Austria
| | | | - Peter Bettelheim
- Department of Internal Medicine I with Hematology, Stem Cell Transplantation, Hemostasis and Medical Oncology, Ordensklinikum Elisabethinen Hospital, Linz, Austria
| | - Otto Zach
- Laboratory for molecular and genetic diagnostics, Ordensklinikum Linz, Linz, Austria
| | - Ansgar Weltermann
- Department of Internal Medicine I with Hematology, Stem Cell Transplantation, Hemostasis and Medical Oncology, Ordensklinikum Elisabethinen Hospital, Linz, Austria
| | - Sonja Heibl
- Department of Internal Medicine IV, Hospital Wels-Grieskirchen, Wels, Austria
| | - Josef Thaler
- Department of Internal Medicine IV, Hospital Wels-Grieskirchen, Wels, Austria
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, Graz, Austria.,Otto-Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Reinhard Stauder
- Internal Medicine V with Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerald Webersinke
- Laboratory for molecular and genetic diagnostics, Ordensklinikum Linz, Linz, Austria
| | - Rajko Kusec
- Internal Medicine V with Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria.,School of Medicine, University Hospital Dubrava, University of Zagreb, Zagreb, Croatia
| | - Ernst Ulsperger
- Department of Internal Medicine, Hospital Horn, Horn, Austria
| | - Bruno Schneeweiss
- Department of Internal Medicine, Hospital Kirchdorf, Kirchdorf, Austria
| | - Leopold Öhler
- Department of Internal Medicine/Oncology, St. Josef Hospital, Vienna, Austria
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria.,Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Heinz Tüchler
- Department of Internal Medicine III, Hanusch Hospital, Vienna, Austria
| | - Klaus Geissler
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, Vienna, Austria.,Sigmund Freud University, Vienna, Austria
| |
Collapse
|
63
|
Chan O, Renneville A, Padron E. Chronic myelomonocytic leukemia diagnosis and management. Leukemia 2021; 35:1552-1562. [PMID: 33714974 DOI: 10.1038/s41375-021-01207-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/23/2021] [Accepted: 02/18/2021] [Indexed: 01/31/2023]
Abstract
Chronic myelomonocytic leukemia (CMML) is a rare, heterogeneous myeloid malignancy classified as a myelodysplastic syndromes/myeloproliferative neoplasm (MDS/MPN) overlap syndrome by the World Health Organization (WHO). Its initial presentation can be incidental or associated with myelodysplastic or myeloproliferative symptoms and up to 20% of patients harbor a concurrent inflammatory or autoimmune condition. Persistent monocytosis is the hallmark of CMML but diagnosis can be challenging. Increased understanding of human monocyte subsets, chromosomal abnormalities, and somatic gene mutations have led to more accurate diagnosis and improved prognostication. A number of risk stratification systems have been developed and validated but using those that incorporate molecular information such as CMML Prognostic Scoring System (CPSS)-Mol, Mayo Molecular, and Groupe Francophone des Myelodysplasies (GFM) are preferred. Symptom-directed approaches forms the basis of CMML management. Outcomes vary substantially depending on risk ranging from observation for a number of years to rapidly progressive disease and acute myeloid leukemia (AML) transformation. Patients who are low risk but with symptoms from cytopenias or proliferative features such as splenomegaly may be treated with hypomethylating agents (HMAs) or cytoreductive therapy, respectively, with the goal of durable symptoms control. Allogeneic hematopoietic cell transplantation should be considered for intermediate to high risk patients. The lack of effective pharmaceutical options has generated interest in novel therapeutics for this disease, and early phase clinical trial results are promising.
Collapse
Affiliation(s)
- Onyee Chan
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Eric Padron
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
64
|
Oligomonocytic and overt chronic myelomonocytic leukemia show similar clinical, genomic, and immunophenotypic features. Blood Adv 2021; 4:5285-5296. [PMID: 33108455 DOI: 10.1182/bloodadvances.2020002206] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/20/2020] [Indexed: 12/19/2022] Open
Abstract
Oligomonocytic chronic myelomonocytic leukemia (OM-CMML) is defined as those myelodysplastic syndromes (MDSs) or myelodysplastic/myeloproliferative neoplasms, unclassifiable with relative monocytosis (≥10% monocytes) and a monocyte count of 0.5 to <1 × 109/L. These patients show clinical and genomic features similar to those of overt chronic myelomonocytic leukemia (CMML), although most of them are currently categorized as MDS, according to the World Health Organization 2017 classification. We analyzed the clinicopathologic features of 40 patients with OM-CMML with well-annotated immunophenotypic and molecular data and compared them to those of 56 patients with overt CMML. We found similar clinical, morphological, and cytogenetic features. In addition, OM-CMML mirrored the well-known complex molecular profile of CMML, except for the presence of a lower percentage of RAS pathway mutations. In this regard, of the different genes assessed, only CBL was found to be mutated at a significantly lower frequency. Likewise, the OM-CMML immunophenotypic profile, assessed by the presence of >94% classical monocytes (MO1s) and CD56 and/or CD2 positivity in peripheral blood monocytes, was similar to overt CMML. The MO1 percentage >94% method showed high accuracy for predicting CMML diagnosis (sensitivity, 90.7%; specificity, 92.2%), even when considering OM-CMML as a subtype of CMML (sensitivity, 84.9%; specificity, 92.1%) in our series of 233 patients (39 OM-CMML, 54 CMML, 23 MDS, and 15 myeloproliferative neoplasms with monocytosis and 102 reactive monocytosis). These results support the consideration of OM-CMML as a distinctive subtype of CMML.
Collapse
|
65
|
Laboratory Evaluation and Pathological Workup of Neoplastic Monocytosis - Chronic Myelomonocytic Leukemia and Beyond. Curr Hematol Malig Rep 2021; 16:286-303. [PMID: 33945086 DOI: 10.1007/s11899-021-00625-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW Monocytosis is a distinct but non-specific manifestation of various physiologic and pathologic conditions. Among hematopoietic stem cell neoplasms, depending on the criteria used for disease classification, monocytosis may be a consistent and integral component of diseases such as chronic myelomonocytic leukemia or acute myeloid leukemia with monocytic differentiation, or it may represent an inconsistent finding that often provides a clue to the underlying genetic changes driving the neoplasm. The purpose of this review is to provide the readers with a laboratory-based approach to neoplastic monocytosis. RECENT FINDINGS In-depth elucidation of the genomic landscape of myeloid neoplasms within the past few years has broadened our understanding of monocytosis and its implications for diagnosis and prognosis. Genetic findings also shed light on potential disease response - or lack thereof - to various therapeutic agents used in the setting of myeloid neoplasms. In this review, we provide our approach to diagnose neoplastic monocytosis in the context of case-based studies while incorporating the most recent literature on this topic.
Collapse
|
66
|
Genomic stratification of myelodysplastic/myeloproliferative neoplasms, unclassifiable: Sorting through the unsorted. Leukemia 2021; 35:3329-3333. [PMID: 33931725 DOI: 10.1038/s41375-021-01258-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 01/04/2023]
|
67
|
Chang YH. Myelodysplastic syndromes and overlap syndromes. Blood Res 2021; 56:S51-S64. [PMID: 33935036 PMCID: PMC8094000 DOI: 10.5045/br.2021.2021010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematological neoplasms characterized by ineffective hematopoiesis, morphologic dysplasia, and cytopenia. MDS overlap syndromes include various disorders, such as myelodysplastic/myeloproliferative neoplasms and hypoplastic MDS with aplastic anemia characteristics. MDS overlap syndromes share the characteristics of other diseases, which make differential diagnoses challenging. Advances in genomic studies have led to the discovery of frequent mutations in MDS and overlap syndromes; however, most of the mutations are not specific for the diagnosis of these diseases. The molecular characteristics of the overlap syndromes usually do not show a just "in-between" form but rather heterogeneous features. Established diagnostic criteria for these diseases based on clinical, morphologic, and laboratory features are still useful when combined with genomic data. It is expected that further studies for MDS and overlap syndromes will place emphasis on the roles of mutations as therapeutic targets and prognostic indicators.
Collapse
Affiliation(s)
- Yoon Hwan Chang
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
68
|
Kwon J. Diagnosis and treatment of chronic myelomonocytic leukemia. Blood Res 2021; 56:S5-S16. [PMID: 33935030 PMCID: PMC8094002 DOI: 10.5045/br.2021.2020321] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/20/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal disorder of hematopoietic cells and is a complex of heterogeneous conditions with both myeloproliferative and myelodysplastic features. The diagnosis of CMML is made using morphologic criteria including monocyte-dominant leukocytosis, dysplastic changes, and increased blasts in the bone marrow. Recently, the identification of monocyte subtypes in peripheral blood using multiparameter flow cytometry has been actively studied. Chromosomal abnormalities are the basis of CMML risk stratification, and mutations in several genes including ASXL1 are known to be important not only for the diagnosis and treatment of this disease but also for predicting its prognosis. The standard treatment principles for CMML have not yet been clearly defined; however, hypomethylating agents are mainly considered the frontline therapy in most cases. Although allogeneic hematopoietic stem cell transplantation has limited applications owing to its toxicity, it still plays an important role as the only curative treatment option. Researchers are continuing to develop new drugs for CMML treatment and to prove their clinical usefulness. This review summarizes what is known to date on the diagnosis, treatment, and prognostic factors of CMML and presents future directions by analyzing recent research trends.
Collapse
Affiliation(s)
- Jihyun Kwon
- Division of Hematology and Oncology, Department of Internal Medicine, Chungbuk National University College of Medicine, Chungbuk National University Hospital, Cheongju, Korea
| |
Collapse
|
69
|
Mangaonkar AA, Tande AJ, Bekele DI. Differential Diagnosis and Workup of Monocytosis: A Systematic Approach to a Common Hematologic Finding. Curr Hematol Malig Rep 2021; 16:267-275. [PMID: 33880680 PMCID: PMC8057007 DOI: 10.1007/s11899-021-00618-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 12/19/2022]
Abstract
Purpose of Review Monocytosis is a frequently encountered clinical condition that needs appropriate investigation due to a broad range of differential diagnoses. This review is meant to summarize the latest literature in the diagnostic testing and interpretation and offer a stepwise diagnostic approach for a patient presenting with monocytosis. Recent Findings Basic studies have highlighted the phenotypic and functional heterogeneity in the monocyte compartment. Studies, both translational and clinical, have provided insights into why monocytosis occurs and how to distinguish the different etiologies. Flow cytometry studies have illustrated that monocyte repartitioning can distinguish chronic myelomonocytic leukemia, a prototypical neoplasm with monocytosis from other reactive or neoplastic causes. Summary In summary, we provide an algorithmic approach to the diagnosis of a patient presenting with monocytosis and expect this document to serve as a reference guide for clinicians.
Collapse
Affiliation(s)
| | - Aaron J Tande
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN, USA
| | - Delamo I Bekele
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
70
|
Jian J, Qiao Y, Li Y, Guo Y, Ma H, Liu B. Mutations in chronic myelomonocytic leukemia and their prognostic relevance. Clin Transl Oncol 2021; 23:1731-1742. [PMID: 33861431 DOI: 10.1007/s12094-021-02585-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/06/2021] [Indexed: 12/19/2022]
Abstract
Chronic myelomonocytic leukemia (CMML) is a hematologic malignancy that overlaps with myeloproliferative neoplasms (MPN) and myelodysplastic syndromes (MDS) and tends to transform into acute myeloid leukemia (AML). Among cases of CMML, > 90% have gene mutations, primarily involving TET2 (~ 60%), ASXL1 (~ 40%), SRSF2 (~ 50%), and the RAS pathways (~ 30%). These gene mutations are associated with both the clinical phenotypes and the prognosis of CMML, special CMML variants and pre-phases of CMML. Cytogenetic abnormalities and the size of genome are also associated with prognosis. Meanwhile, cases with ASXL1, DNMT3A, NRAS, SETBP1, CBL and RUNX1 mutations may have inferior prognoses, but only ASXL1 mutations were confirmed to be independent predictors of the patient outcome and were included in three prognostic models. Novel treatment targets related to the various gene mutations are emerging. Therefore, this review provides new insights to explore the correlations among gene mutations, clinical phenotypes, prognosis, and novel drugs in CMML.
Collapse
Affiliation(s)
- J Jian
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Y Qiao
- Institute of Hematology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Y Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Y Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - H Ma
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China. .,Department of Hematology, The First Affiliated Hospital, Lanzhou University, 1 Donggangxilu street, Lanzhou, Gansu, China.
| | - B Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China. .,Department of Hematology, The First Affiliated Hospital, Lanzhou University, 1 Donggangxilu street, Lanzhou, Gansu, China.
| |
Collapse
|
71
|
Bewersdorf JP, Zeidan AM. Risk-Adapted, Individualized Treatment Strategies of Myelodysplastic Syndromes (MDS) and Chronic Myelomonocytic Leukemia (CMML). Cancers (Basel) 2021; 13:1610. [PMID: 33807279 PMCID: PMC8036734 DOI: 10.3390/cancers13071610] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) are two distinct blood cancers with a variable clinical symptom burden and risk of progression to acute myeloid leukemia. Management decisions should be guided by individual patient and disease characteristics and based on validated risk stratification tools. While supportive care with red blood cell transfusions, erythropoiesis-stimulating agents, and iron chelation remains the mainstay of therapy for lower-risk (LR)-MDS patients, luspatercept has recently been approved for transfusion-dependent anemic LR-MDS patients ending a decade without any new drug approvals for MDS. For higher-risk patients, allogeneic hematopoietic cell transplant (allo-HCT) remains the only curative therapy for both MDS and CMML but most patients are not eligible for allo-HCT. For those patients, the hypomethylating agents (HMA) azacitidine and decitabine remain standard of care with azacitidine being the only agent that has shown an overall survival benefit in randomized trials. Although early results from novel molecularly driven agents such as IDH1/2 inhibitors, venetoclax, magrolimab, and APR-246 for MDS as well as tagraxofusp, tipifarnib, and lenzilumab for CMML appear encouraging, confirmatory randomized trials must be completed to fully assess their safety and efficacy prior to routine clinical use. Herein, we review the current management of MDS and CMML and conclude with a critical appraisal of novel therapies and general trends in this field.
Collapse
Affiliation(s)
| | - Amer M. Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, 333 Cedar Street, P.O. Box 208028, New Haven, CT 06520-8028, USA;
| |
Collapse
|
72
|
Geissler K, Jäger E, Barna A, Graf T, Graf E, Öhler L, Hoermann G, Valent P. Myelomonocytic skewing in chronic myelomonocytic leukemia: phenotypic, molecular and biologic features and impact on survival. Eur J Haematol 2021; 106:627-633. [PMID: 33432601 PMCID: PMC8554855 DOI: 10.1111/ejh.13577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/07/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Myelomonocytic skewing is considered as a key pathophysiologic phenomenon in chronic myelomonocytic leukemia (CMML), but its prevalence and potential correlation with phenotypic, genotypic, and clinical features are poorly defined. METHODS Skewed differentiation toward the myelomonocytic over erythroid commitment as indicated by an inverse ratio of myelomonocytic/erythroid colonies was investigated in 146 patients with CMML by semisolid in vitro cultures. RESULTS There was a high prevalence of myelomonocytic skewing in patients with CMML (120/146, 82%); whereas, this phenomenon was rare in normal individuals (1/98, 1%). Patients with CMML with myelomonocytic skewing had higher white blood cell and peripheral blast cell counts, and lower platelet values. The number of mutations in genes of the epigenetic and/or splicing category was higher in CMML patients with as compared with patients without skewing. Patients with myelomonocytic skewing had more frequently mutations in RASopathy genes and higher growth factor independent myeloid colony formation. Interestingly, the lack of myelomonocytic skewing discriminated patients with CMML with a particularly favorable prognosis (60 vs 19 months, P = .003) and a minimal risk of transformation. CONCLUSION Myelomonocytic skewing as determined by semisolid cultures can discriminate subgroups of patients with CMML with a different phenotype, a different genotype, and a different prognosis.
Collapse
Affiliation(s)
- Klaus Geissler
- Medical School, Sigmund Freud University, Vienna, Austria.,Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, Vienna, Austria
| | - Eva Jäger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Agnes Barna
- Blood Transfusion Service, Blood Transfusion Service for Upper Austria, Austrian Red Cross, Linz, Austria
| | - Temeida Graf
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, Vienna, Austria
| | - Elmir Graf
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, Vienna, Austria
| | - Leopold Öhler
- Department of Internal Medicine/Oncology, St. Josef Hospital, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.,Central Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Innsbruck, Innsbruck, Austria.,Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
73
|
Genomic Landscape and Risk Stratification in Chronic Myelomonocytic Leukemia. Curr Hematol Malig Rep 2021; 16:247-255. [PMID: 33660195 DOI: 10.1007/s11899-021-00613-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE The advent of next-generation sequencing has allowed for the annotation of a vast array of recurrent somatic mutations across human malignancies, ushering in a new era of precision oncology. Chronic myelomonocytic leukemia is recognized as a myelodysplastic/myeloproliferative neoplasm and displays heterogenous clinical and genetic features. Herein, we review what is currently understood regarding the genomic landscape of this disease and discuss how somatic mutations have impacted current risk stratification methods. RECENT FINDINGS Genomic studies in chronic myelomonocytic leukemia have identified a characteristic spectrum of cytogenetic and molecular abnormalities. Chromosomal abnormalities are detected in ~30% of patients and somatic gene mutations in up to 90% of patients, most commonly in TET2, SRSF2, and ASXL1. While cytogenetic abnormalities have long been known to impact the prognosis of myeloid neoplasms, recent studies have identified that somatic mutations impact prognosis independent of cytogenetic and clinical variables. This is best exemplified by mutations in ASXL1, which have been uniformly associated with inferior survival. These findings have led to the development of three molecularly inspired prognostic models, in an attempt to more accurately prognosticate in the disease. Our understanding of the genomic landscape of chronic myelomonocytic leukemia continues to evolve, with somatic mutations demonstrating an expanding role in diagnosis, risk stratification, and therapeutic decision-making. Given these findings, molecular profiling by next-generation sequencing should be considered standard of care in all patients.
Collapse
|
74
|
Kuykendall AT, Tokumori FC, Komrokji RS. Traipsing Through Muddy Waters: A Critical Review of the Myelodysplastic Syndrome/Myeloproliferative Neoplasm (MDS/MPN) Overlap Syndromes. Hematol Oncol Clin North Am 2021; 35:337-352. [PMID: 33641873 DOI: 10.1016/j.hoc.2020.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Myelodysplastic syndrome/Myeloproliferative neoplasms (MDS/MPNs) are molecularly complex, clinically heterogeneous diseases that exhibit proliferative and dysplastic features. Diagnostic criteria use clinical, pathologic, and genomic features to distinguish between disease entities, though considerable clinical and genetic overlap persists. MDS/MPNs are associated with a poor prognosis, save for MDS/MPN with ring sideroblasts and thrombocytosis, which can behave more indolently. The current treatment approach is risk-adapted and symptom-directed and largely extrapolated from experience in MDS or MPN. Gene sequencing has demonstrated frequent mutations involving signaling, epigenetic, and splicing pathways, which present numerous therapeutic opportunities for clinical investigation.
Collapse
Affiliation(s)
- Andrew T Kuykendall
- Moffitt Cancer Center, 12902 USF Magnolia Drive, CSB 7th Floor, Tampa, FL 33612, USA.
| | - Franco Castillo Tokumori
- University of South Florida, 17 Davis Boulevard, Suite 308, Tampa, FL 33606, USA. https://twitter.com/CTFrancoMD
| | - Rami S Komrokji
- Moffitt Cancer Center, 12902 USF Magnolia Drive, CSB 7th Floor, Tampa, FL 33612, USA. https://twitter.com/Ramikomrokji
| |
Collapse
|
75
|
Hasserjian RP, Buckstein R, Patnaik MM. Navigating Myelodysplastic and Myelodysplastic/Myeloproliferative Overlap Syndromes. Am Soc Clin Oncol Educ Book 2021; 41:328-350. [PMID: 34010050 DOI: 10.1200/edbk_320113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Myelodysplastic syndromes (MDS) and MDS/myeloproliferative neoplasms (MPNs) are clonal diseases that differ in morphologic diagnostic criteria but share some common disease phenotypes that include cytopenias, propensity to acute myeloid leukemia evolution, and a substantially shortened patient survival. MDS/MPNs share many clinical and molecular features with MDS, including frequent mutations involving epigenetic modifier and/or spliceosome genes. Although the current 2016 World Health Organization classification incorporates some genetic features in its diagnostic criteria for MDS and MDS/MPNs, recent accumulation of data has underscored the importance of the mutation profiles on both disease classification and prognosis. Machine-learning algorithms have identified distinct molecular genetic signatures that help refine prognosis and notable associations of these genetic signatures with morphologic and clinical features. Combined geno-clinical models that incorporate mutation data seem to surpass the current prognostic schemes. Future MDS classification and prognostication schema will be based on the portfolio of genetic aberrations and traditional features, such as blast count and clinical factors. Arriving at these systems will require studies on large patient cohorts that incorporate advanced computational analysis. The current treatment algorithm in MDS is based on patient risk as derived from existing prognostic and disease classes. Luspatercept is newly approved for patients with MDS and ring sideroblasts who are transfusion dependent after erythropoietic-stimulating agent failure. Other agents that address red blood cell transfusion dependence in patients with lower-risk MDS and the failure of hypomethylating agents in higher-risk disease are in advanced testing. Finally, a plethora of novel targeted agents and immune checkpoint inhibitors are being evaluated in combination with a hypomethylating agent backbone to augment the depth and duration of response and, we hope, improve overall survival.
Collapse
Affiliation(s)
| | - Rena Buckstein
- Division of Hematology/Oncology, Sunnybrook Odette Cancer Center, Toronto, Ontario, Canada
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN
| |
Collapse
|
76
|
Molina JC, Asare JM, Tuschong L, West RR, Calvo KR, Persky R, Boyce AM, Hammoud DA, Holland SM, Hickstein D, Shah NN. Venetoclax/decitabine for a pediatric patient with chronic myelomonocytic leukemia. Pediatr Blood Cancer 2021; 68:e28865. [PMID: 33369023 PMCID: PMC9357463 DOI: 10.1002/pbc.28865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/07/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022]
Abstract
Chronic myelomonocytic leukemia (CMML) is a myelodysplastic syndrome (MDS)/myeloproliferative disorder most commonly seen in the elderly. We describe an adolescent with monosomy 7 CMML presenting as central diabetes insipidus (DI), who was treated with venetoclax and decitabine as a bridge to hematopoietic stem cell transplantation (HSCT). Central DI is a rare manifestation of monosomy 7-associated MDS including CMML, itself a rare manifestation of GATA2 deficiency, particularly in children. Venetoclax/decitabine was effective for treatment of CMML as a bridge to HSCT.
Collapse
Affiliation(s)
- John C. Molina
- Pediatric Oncology Branch, Center for Cancer Research (CCR)National Cancer Institute (NCI)NIH, Bethesda, Maryland,Department of Pediatric Oncology, Johns Hopkins Hospital, Baltimore, Maryland
| | - Julie M. Asare
- Department of Pediatric Oncology, Johns Hopkins Hospital, Baltimore, Maryland
| | - Laura Tuschong
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Robert R. West
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Katherine R. Calvo
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda, Maryland
| | - Rebecca Persky
- National Institute of Child Health and Human Development, NIH, Bethesda, Maryland,Department of Pediatric Endocrinology, Children’s National Medical Center, Washington, District of Columbia
| | - Alison M. Boyce
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial ResearchNIH, Bethesda, Maryland
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, NIH Clinical Center, Bethesda, Maryland
| | - Steven M. Holland
- Immunopathogenesis Section, National Institute of Allergy and Infectious DiseasesNIH, Bethesda, Maryland
| | - Dennis Hickstein
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR)National Cancer Institute (NCI)NIH, Bethesda, Maryland
| |
Collapse
|
77
|
[Guideline of the diagnosis and treatment of chronic myelomonocytic leukemia (2021)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:5-9. [PMID: 33677861 PMCID: PMC7957249 DOI: 10.3760/cma.j.issn.0253-2727.2021.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Indexed: 11/29/2022]
|
78
|
Ronaghy A, Yang RK, Khoury JD, Kanagal-Shamanna R. Clinical Applications of Chromosomal Microarray Testing in Myeloid Malignancies. Curr Hematol Malig Rep 2020; 15:194-202. [PMID: 32382988 DOI: 10.1007/s11899-020-00578-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Knowledge of both somatic mutations and copy number aberrations are important for the understanding of cancer pathogenesis and management of myeloid neoplasms. The currently available standard of care technologies for copy number assessment such as conventional karyotype and FISH are either limited by low resolution or restriction to targeted assessment. RECENT FINDINGS Chromosomal microarray (CMA) is effective in characterization of chromosomal and gene aberrations of diagnostic, prognostic, and therapeutic significance at a higher resolution than conventional karyotyping. These results are complementary to NGS mutation studies. Copy-neutral loss of heterozygosity (CN-LOH), which is prognostic in AML, is currently only identified by CMA. Yet, despite the widespread availability, CMA testing is not routinely performed in diagnostic laboratories due to lack of knowledge on best-testing practices for clinical work-up of myeloid neoplasms. In this review, we provide an overview of the clinical significance of CMA in acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and myelodysplastic/myeloproliferative neoplasms (MDS/MPN). We will also elaborate the specific clinical scenarios where CMA can provide additional information essential for management and could potentially alter treatment. Chromosomal microarray (CMA) is an effective technology for characterizing chromosomal copy number changes and copy-neutral loss of heterozygosity of diagnostic, prognostic, and therapeutic significance at a high resolution in myeloid malignancies.
Collapse
MESH Headings
- Chromosome Aberrations
- Chromosomes, Human
- Comparative Genomic Hybridization
- DNA Copy Number Variations
- Genetic Predisposition to Disease
- High-Throughput Nucleotide Sequencing
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myelomonocytic, Chronic/diagnosis
- Leukemia, Myelomonocytic, Chronic/genetics
- Loss of Heterozygosity
- Microarray Analysis
- Myelodysplastic Syndromes/diagnosis
- Myelodysplastic Syndromes/genetics
- Polymorphism, Single Nucleotide
- Predictive Value of Tests
- Prognosis
- Reproducibility of Results
Collapse
Affiliation(s)
- Arash Ronaghy
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 072, Houston, TX, 77030, USA
| | - Richard K Yang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 072, Houston, TX, 77030, USA
| | - Joseph D Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 072, Houston, TX, 77030, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 072, Houston, TX, 77030, USA.
| |
Collapse
|
79
|
Abstract
In recent years CMML has received increased attention as the most commonly observed MDS/MPN overlap syndrome. Renewed interest has occurred in part due to widespread adoption of next-generation sequencing panels that help render the diagnosis in the absence of morphologic dysplasia. Although most CMML patients exhibit somatic mutations in epigenetic modifiers, spliceosome components, transcription factors and signal transduction genes, it is increasingly clear that a small subset harbors an inherited predisposition to CMML and other myeloid neoplasms. More intriguing is the fact that the mutational spectrum observed in CMML is found in other types of myeloid leukemias, begging the question of how similar genetic backgrounds can lead to such divergent clinical phenotypes. In this review we present a contemporary snapshot of the genetic complexity inherent to CMML, explore the relationship between genotype-phenotype and present a stepwise model of CMML pathogenesis and progression.
Collapse
Affiliation(s)
- Ami B Patel
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael W Deininger
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
80
|
Thomopoulos TP, Bouhla A, Papageorgiou SG, Pappa V. Chronic myelomonocytic leukemia - a review. Expert Rev Hematol 2020; 14:59-77. [PMID: 33275852 DOI: 10.1080/17474086.2021.1860004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Chronic myelomonocytic leukemia (CMML) is a clonal myeloid neoplasm, denoted by overlapping myelodysplastic and myeloproliferative features, with poor overall survival and high transformation rate to acute myeloid leukemia. AREAS COVERED This review, following a thorough Medline search of pertinent published literature, discusses the diagnostic criteria, the pathogenesis, and the complex genetic landscape of the disease. Prognostication, response criteria, therapeutic management of patients, efficacy of established and novel treatment modalities are thoroughly reviewed. EXPERT OPINION Cytogenetic abnormalities and mutations in genes involved in epigenetic and transcriptional regulation, and cell-signaling are abundant in CMML and implicated in its complex pathogenesis. As presence of these mutations carry a prognostic impact, they are increasingly incorporated in risk-stratification schemes. Novel response criteria have been proposed, considering the unique features of the disease. Although allogeneic hematopoietic stem cell transplantation remains the only treatment with curative intent, it is reserved for a minority of patients; therefore, there is an unmet need for optimizing treatment modalities, such as hypomethylating agents, and introducing novel agents, which could substantially improve survival and quality of life of CMML patients. Clinical trials dedicated specifically to CMML are needed to explore the efficacy and safety of novel treatment modalities.
Collapse
Affiliation(s)
- Thomas P Thomopoulos
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| | - Anthi Bouhla
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| | - Sotirios G Papageorgiou
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| | - Vasiliki Pappa
- 2 Department of Internal Medicine - Propaedeutic and Research Unit, National and Kapodistrian University of Athens, Medical School, University General Hospital "Attikon" , Athens, Greece
| |
Collapse
|
81
|
Pastorello EA, Borgonovo L, Preziosi D, Schroeder JW, Pravettoni V, Aversano MG, Pastori S, Bilò MB, Piantanida M, Losappio LM, Nichelatti M, Rossi CM, Farioli L. Basal Tryptase High Levels Associated with a History of Arterial Hypertension and Hypercholesterolemia Represent Risk Factors for Severe Anaphylaxis in Hymenoptera Venom-Allergic Subjects over 50 Years Old. Int Arch Allergy Immunol 2020; 182:146-152. [PMID: 33264768 DOI: 10.1159/000510527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/28/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Allergy to Hymenoptera venom (HV) may lead to life-threatening anaphylaxis. Some of the factors influencing the symptom's severity are still undetermined. The aim of this study was to identify the clinical aspects associated with the most severe reactions in a population with HV allergy, by comparing clinical and immunochemical biomarkers between patients with previous local large reactions (LLRs) and systemic reactions (SRs). METHODS We selected adult patients with a history of HV allergy, with positive diagnostic tests and a correlation with one single Hymenoptera species. Age, gender, atopy, serum basal tryptase (sBT) value, total IgE, venom-specific IgE, history of hypertension, cardiovascular diseases, and hypercholesterolemia were compared between patients with previous LLRs and SRs. RESULTS 460 adult patients (381 SRs, 79 LLRs) were included. Age (p = 0.0097), male gender (p < 0.0001), arterial hypertension (p = 0.046), hypercholesterolemia (p = 0.009), and higher sBT levels (p = 0.0004) were significantly associated with severe reactions as independent variables. Moreover, considering the previous variables as risk factors, there was a significant and progressive increase in the odds of being Mueller III + IV as the number of positive variables increased. Patients with sBT ≥6.4 ng/mL adjusted for any of the positive variables had increased the risk of Mueller grade IV reaction (p < 0.0001). CONCLUSION According to our results, older age, male gender, arterial hypertension, hypercholesterolemia, and increased levels of sBT ≥6.4 ng/mL are risk factors for severe anaphylaxis to HV in adults. Atopy and allergic asthma do not increase the risk of HV-induced SRs.
Collapse
Affiliation(s)
- Elide Anna Pastorello
- Unit of Allergology and Immunology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy,
| | - Linda Borgonovo
- Unit of Allergology and Immunology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Donatella Preziosi
- Unit of Allergology and Immunology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Jan Walter Schroeder
- Unit of Allergology and Immunology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Valerio Pravettoni
- Unit of General Medicine, Immunology and Allergology, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Gloria Aversano
- Unit of Internal Medicine, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Stefano Pastori
- Unit of Laboratory Medicine, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Maria Beatrice Bilò
- Allergy Unit, Internal Medicine, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Marta Piantanida
- Unit of Allergology and Immunology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Michele Nichelatti
- Service of Biostatistics, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Carlo Maria Rossi
- Unit of Allergology and Immunology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Farioli
- Unit of Laboratory Medicine, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
82
|
Jiang Z, Sun X, Wu Z, Alhatem A, Zheng R, Liu D, Wang Y, Kumar D, Xia C, You B, Wang H, Liu C, Jiang JG. Cytogenetic and molecular landscape and its potential clinical significance in Hispanic CMML patients from Puerto Rico. Oncotarget 2020; 11:4411-4420. [PMID: 33315966 PMCID: PMC7720771 DOI: 10.18632/oncotarget.27824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic neoplasm that exhibits myelodysplastic and myeloproliferative characteristics with heterogeneous clinical and pathological features. There are limited publications on the ethnic and racial disparity of cytogenetics and genomics in CMML patients. This study aims to define the cytogenetic and molecular landscape in Hispanic CMML patients from Puerto Rico and explore its possible clinical significance. One hundred and eleven (111) Hispanic CMML patients from Puerto Rico were diagnosed in our institute from 2009 to 2018. Karyotypes were available in one hundred and seven (107) patients. Seventeen (17) patients had abnormal karyotypes (17/107, 16%). Compared to previously published data, Hispanic CMML patients in this study had significantly lower rates of overall cytogenetic abnormalities (16% vs 27-28%, p < 0.05) and trisomy 8 (2% vs 7%, p < 0.05). Among one hundred and eleven (111) Hispanic CMML patients, 40-gene myeloid molecular profile tests were performed in fifty-six (56) CMML patients. Gene mutations were identified in fifty-four (54) patients (96%). The most frequent mutated genes were: TET2, SRSF2, ASXL1, ZRSR2, DNMT3A, NRAS, CBL, and RUNX1. Twenty-nine (29) out of fifty-six (56) patients (29/56, 52%) had mutated TET2/wild type ASXL1 (muTET2/wtASXL1). Previous studies indicated that mutated ASXL1, DNMT3A, NRAS, RUNX1, and SETBP1 may associate with an unfavorable prognosis and muTET2/wtASXL1 may associate with a favorable prognosis in CMML patients. Compared to previously published data, Hispanic CMML patients from Puerto Rico in this study had significantly lower mutation rates in ASXL1 and SETBP1, and a higher rate of muTET2/wtASXL1. The findings raise the possibility of a favorable prognosis in Hispanic CMML patients.
Collapse
Affiliation(s)
- Zeju Jiang
- Department of Pathology, Immunology & Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.,Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Xinlai Sun
- Department of Pathology, Immunology & Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Zhao Wu
- Neogenomics, Carlsbad, CA 92008, USA
| | - Albert Alhatem
- Department of Pathology, Immunology & Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Ruifang Zheng
- Department of Pathology, Immunology & Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Dongfang Liu
- Department of Pathology, Immunology & Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Yaqun Wang
- Department of Biostatistics, Rutgers School of Public Health and Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Dibyendu Kumar
- Institute of Genomics Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Changqing Xia
- Institute of Genomics Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Bei You
- Department of Pathology, Immunology & Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - He Wang
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | - Chen Liu
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jie-Gen Jiang
- Department of Pathology, Immunology & Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA.,Institute of Genomics Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| |
Collapse
|
83
|
Hebeda K, Boudova L, Beham-Schmid C, Orazi A, Kvasnicka HM, Gianelli U, Tzankov A. Progression, transformation, and unusual manifestations of myelodysplastic syndromes and myelodysplastic-myeloproliferative neoplasms: lessons learned from the XIV European Bone Marrow Working Group Course 2019. Ann Hematol 2020; 100:117-133. [PMID: 33128619 DOI: 10.1007/s00277-020-04307-9] [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] [Received: 06/13/2020] [Accepted: 10/15/2020] [Indexed: 11/30/2022]
Abstract
Disease progression in myelodysplastic syndromes (MDS) and myelodysplastic-myeloproliferative neoplasms (MDS/MPN) is a major source of mortality. The European Bone Marrow Working Group organized a dedicated workshop to address MDS and MDS/MPN progression, and myeloid neoplasms with histiocytic and lymphoblastic outgrowths in 2019 in Frankfurt, Germany. In this report, we summarize clinical, histopathological, and molecular features of 28 cases. Most cases illustrate that prognostic mutational profiles change during follow-up due to accumulation of high-risk mutations in the trunk clone, and that results from repeated molecular testing can often explain the clinical progression, suggesting that regular genetic testing may predict transformation by early detection of aggressive clones. Importantly, identical mutations can be linked to different clinical behaviors or risks of fibrotic progression and/or transformation in a context-dependent manner, i.e., MDS or MDS/MPN. Moreover, the order of mutational acquisition and the involved cell lineages matter. Several cases exemplify that histiocytic outgrowths in myeloid neoplasms are usually accompanied by a more aggressive clinical course and may be considered harbinger of disease progression. Exceptionally, lymphoblastic transformations can be seen. As best estimable, the histiocytic and lymphoblastic compounds in all occasions were clonally related to the myeloid compound and-where studied-displayed genomic alterations of, e.g., transcription factor genes or genes involved in MAPK signaling that might be mechanistically linked to the respective type of non-myeloid outgrowth.
Collapse
Affiliation(s)
- Konnie Hebeda
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | | | - Attilio Orazi
- Department of Pathology, Texas Tech Health Sciences Center El Paso, El Paso, TX, USA
| | | | - Umberto Gianelli
- Pathology Unit, Department of Pathophysiology and Transplantation, University of Milan and Fondazione IRCCS, Ca' Granda-Maggiore Policlinico, Milan, Italy
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital of Basel, Schoenbeinstrasse 40, CH-4031, Basel, Switzerland.
| |
Collapse
|
84
|
Hunter AM, Padron E. Molecular genetics of MDS/MPN overlap syndromes. Best Pract Res Clin Haematol 2020; 33:101195. [DOI: 10.1016/j.beha.2020.101195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/27/2020] [Indexed: 01/05/2023]
|
85
|
Foucar K, Hsi ED, Wang SA, Rogers HJ, Hasserjian RP, Bagg A, George TI, Bassett RL, Peterson LC, Morice WG, Arber DA, Orazi A, Bueso-Ramos CE. Concordance among hematopathologists in classifying blasts plus promonocytes: A bone marrow pathology group study. Int J Lab Hematol 2020; 42:418-422. [PMID: 32297416 DOI: 10.1111/ijlh.13212] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 02/03/2023]
Abstract
Enumeration of blasts and promonocytes is essential for World Health Organization (WHO) classification of myelomonocytic neoplasms. The accuracy of distinguishing blasts, promonocytes and monocytes, including normal vs abnormal monocytes, remains controversial. The objective of this analysis is to assess concordances between experienced hematopathologists in classifying cells as blasts, promonocytes, and monocytes according to WHO criteria. Each of 11 hematopathologists assessed glass slides from 20 patients [12 with chronic myelomonocytic leukemia (CMML) and 8 with acute myeloid leukemia (AML)] including blood and BM aspirate smears, and limited nonspecific esterase (NSE) stains. All cases were blindly reviewed. Fleiss' extension of Cohen's kappa for multiple raters was used on these variables, separately for peripheral blood (PB) and bone marrow (BM). Spearman's rank correlation was used to assess correlations between each pair of hematopathologists for each measurement. For the classification based on the sum of blasts and promonocytes in the BM, Fleiss' kappa was estimated as 0.744. For PB, categorizing patients according to the sum of blasts and promonocytes, Fleiss' kappa was estimated as 0.949. Distinction of abnormal monocytes from normal monocytes in PB did not achieve a good concordance and showed strong evidence of differences between hematopathologists (P < .0001). The hematopathologists achieved a good concordance rate of 74% in CMML vs AML classification and a high k rate, confirming that criteria for defining the blasts equivalents (blasts plus promonocytes) could be applied consistently. Identification of monocyte subtypes (abnormal vs normal) was not concordant. Our results support the practice of combining blasts/promonocytes into a single category.
Collapse
Affiliation(s)
- Kathryn Foucar
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico
| | - Eric D Hsi
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Sa A Wang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heesun J Rogers
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Robert P Hasserjian
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tracy I George
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico
| | - Roland L Bassett
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - LoAnn C Peterson
- Department of Pathology, Northwestern University, Chicago, Illinois
| | - William G Morice
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Attilio Orazi
- Department of Pathology, Texas Tech University, El Paso, Texas
| | - Carlos E Bueso-Ramos
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
86
|
Clinical, Hematologic, Biologic and Molecular Characteristics of Patients with Myeloproliferative Neoplasms and a Chronic Myelomonocytic Leukemia-Like Phenotype. Cancers (Basel) 2020; 12:cancers12071891. [PMID: 32674283 PMCID: PMC7409251 DOI: 10.3390/cancers12071891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/07/2020] [Accepted: 07/12/2020] [Indexed: 11/17/2022] Open
Abstract
Patients with a myeloproliferative neoplasm (MPN) sometimes show a chronic myelomonocytic leukemia (CMML)-like phenotype but, according to the 2016 WHO classification, a documented history of an MPN excludes the diagnosis of CMML. Forty-one patients with an MPN (35 polycythemia vera (PV), 5 primary myelofibrosis, 1 essential thrombocythemia) and a CMML-like phenotype (MPN/CMML) were comprehensively characterized regarding clinical, hematologic, biologic and molecular features. The white blood cell counts in MPN/CMML patients were not different from CMML patients and PV patients. The hemoglobin values and platelet counts of these patients were higher than in CMML but lower than in PV, respectively. MPN/CMML patients showed myelomonocytic skewing, a typical in vitro feature of CMML but not of PV. The mutational landscape of MPN/CMML was not different from JAK2-mutated CMML. In two MPN/CMML patients, development of a CMML-like phenotype was associated with a decrease in the JAK2 V617F allelic burden. Finally, the prognosis of MPN/CMML (median overall survival (OS) 27 months) was more similar to CMML (JAK2-mutated, 28 months; JAK2-nonmutated 29 months) than to PV (186 months). In conclusion, we show that patients with MPN and a CMML-like phenotype share more characteristics with CMML than with PV, which may be relevant for their classification and clinical management.
Collapse
|
87
|
Njue L, Porret N, Fux M, Bacher U, Banz Y, Rovó A. Systemic mastocytosis with an associated hematological neoplasms: One or two entities? EJHAEM 2020; 1:353-355. [PMID: 35847724 PMCID: PMC9175919 DOI: 10.1002/jha2.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 11/17/2022]
Affiliation(s)
- Linet Njue
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University HospitalUniversity of Bern Bern Switzerland
| | - Naomi Porret
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University HospitalUniversity of Bern Bern Switzerland
| | - Michaela Fux
- University Institute of Clinical Chemistry, Inselspital, Bern University HospitalUniversity of Bern Bern Switzerland
| | - Ulrike Bacher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University HospitalUniversity of Bern Bern Switzerland
| | - Yara Banz
- Institute of PathologyUniversity of Bern Bern Switzerland
| | - Alicia Rovó
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University HospitalUniversity of Bern Bern Switzerland
| |
Collapse
|
88
|
Moreno Berggren D, Kjellander M, Backlund E, Engvall M, Garelius H, Lorenz F, Nilsson L, Rasmussen B, Lehmann S, Hellström-Lindberg E, Jädersten M, Ungerstedt J, Ejerblad E. Prognostic scoring systems and comorbidities in chronic myelomonocytic leukaemia: a nationwide population-based study. Br J Haematol 2020; 192:474-483. [PMID: 32501529 DOI: 10.1111/bjh.16790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/04/2020] [Indexed: 01/07/2023]
Abstract
Outcomes in chronic myelomonocytic leukaemia (CMML) are highly variable and may be affected by comorbidity. Therefore, prognostic models and comorbidity indices are important tools to estimate survival and to guide clinicians in individualising treatment. In this nationwide population-based study, we assess comorbidities and for the first time validate comorbidity indices in CMML. We also compare the prognostic power of: the revised International Prognostic Scoring System (IPSS-R), CMML-specific prognostic scoring system (CPSS), MD Anderson Prognostic Scoring System (MDAPS) and Mayo score. In this cohort of 337 patients with CMML, diagnosed between 2009 and 2015, the median overall survival was 21·3 months. Autoimmune conditions were present in 25% of the patients, with polymyalgia rheumatica and Hashimoto's thyroiditis being most common. Of the tested comorbidity indices: the Charlson Comorbidity Index (CCI), Haematopoietic cell transplantation-specific Comorbidity Index (HCT-CI) and Myelodysplastic Syndrome-Specific Comorbidity Index (MDS-CI), CCI had the highest C-index (0·62) and was the only comorbidity index independently associated with survival in multivariable analyses. When comparing the prognostic power of the scoring systems, the CPSS had the highest C-index (0·69). In conclusion, using 'real-world' data we found that the CCI and CPSS have the best prognostic power and that autoimmune conditions are overrepresented in CMML.
Collapse
Affiliation(s)
- Daniel Moreno Berggren
- Department of Medical Science, Section of Hematology, Uppsala University, Uppsala, Sweden
| | - Matilda Kjellander
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital and PO Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Ellen Backlund
- Department of Medical Science, Section of Hematology, Uppsala University, Uppsala, Sweden
| | - Marie Engvall
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hege Garelius
- Section for Haematology and Coagulation, Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Fryderyk Lorenz
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Lars Nilsson
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Bengt Rasmussen
- School of Medical Sciences, Örebro University Hospital, Örebro, Sweden
| | - Sören Lehmann
- Department of Medical Science, Section of Hematology, Uppsala University, Uppsala, Sweden
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital and PO Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Jädersten
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital and PO Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Johanna Ungerstedt
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital and PO Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Elisabeth Ejerblad
- Department of Medical Science, Section of Hematology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
89
|
Geissler K, Jäger E, Barna A, Gurbisz M, Graf T, Graf E, Nösslinger T, Pfeilstöcker M, Tüchler H, Sliwa T, Keil F, Geissler C, Heibl S, Thaler J, Machherndl-Spandl S, Zach O, Weltermann A, Bettelheim P, Stauder R, Zebisch A, Sill H, Schwarzinger I, Schneeweiss B, Öhler L, Ulsperger E, Kusec R, Germing U, Sperr WR, Knöbl P, Jäger U, Hörmann G, Valent P. Correlation of RAS-Pathway Mutations and Spontaneous Myeloid Colony Growth with Progression and Transformation in Chronic Myelomonocytic Leukemia-A Retrospective Analysis in 337 Patients. Int J Mol Sci 2020; 21:3025. [PMID: 32344757 PMCID: PMC7215883 DOI: 10.3390/ijms21083025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 01/07/2023] Open
Abstract
Although the RAS-pathway has been implicated as an important driver in the pathogenesis of chronic myelomonocytic leukemia (CMML) a comprehensive study including molecular and functional analyses in patients with progression and transformation has not been performed. A close correlation between RASopathy gene mutations and spontaneous in vitro myeloid colony (CFU-GM) growth in CMML has been described. Molecular and/or functional analyses were performed in three cohorts of 337 CMML patients: in patients without (A, n = 236) and with (B, n = 61) progression/transformation during follow-up, and in patients already transformed at the time of sampling (C, n = 40 + 26 who were before in B). The frequencies of RAS-pathway mutations (variant allele frequency ≥ 20%) in cohorts A, B, and C were 30%, 47%, and 71% (p < 0.0001), and of high colony growth (≥20/105 peripheral blood mononuclear cells) 31%, 44%, and 80% (p < 0.0001), respectively. Increases in allele burden of RAS-pathway mutations and in numbers of spontaneously formed CFU-GM before and after transformation could be shown in individual patients. Finally, the presence of mutations in RASopathy genes as well as the presence of high colony growth prior to transformation was significantly associated with an increased risk of acute myeloid leukemia (AML) development. Together, RAS-pathway mutations in CMML correlate with an augmented autonomous expansion of neoplastic precursor cells and indicate an increased risk of AML development which may be relevant for targeted treatment strategies.
Collapse
MESH Headings
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cytogenetic Analysis
- Disease Progression
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/metabolism
- Leukemia, Myelomonocytic, Chronic/mortality
- Leukemia, Myelomonocytic, Chronic/pathology
- Mutation
- Neoplasm Staging
- Neoplastic Stem Cells/metabolism
- Prognosis
- Retrospective Studies
- Signal Transduction
- ras Proteins/genetics
- ras Proteins/metabolism
Collapse
Affiliation(s)
- Klaus Geissler
- Medical School, Sigmund Freud University, 1020 Vienna, Austria
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, 1130 Vienna, Austria; (T.G.); (E.G.)
| | - Eva Jäger
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; (E.J.); (M.G.); (I.S.)
| | - Agnes Barna
- Blood Transfusion Service, Blood Transfusion Service for Upper Austria, Austrian Red Cross, 4020 Linz, Austria;
| | - Michael Gurbisz
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; (E.J.); (M.G.); (I.S.)
| | - Temeida Graf
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, 1130 Vienna, Austria; (T.G.); (E.G.)
| | - Elmir Graf
- Department of Internal Medicine V with Hematology, Oncology and Palliative Medicine, Hospital Hietzing, 1130 Vienna, Austria; (T.G.); (E.G.)
| | - Thomas Nösslinger
- Department of Internal Medicine III, Hanusch Hospital, 1140 Vienna, Austria; (T.N.); (M.P.); (H.T.); (T.S.); (F.K.)
| | - Michael Pfeilstöcker
- Department of Internal Medicine III, Hanusch Hospital, 1140 Vienna, Austria; (T.N.); (M.P.); (H.T.); (T.S.); (F.K.)
| | - Heinz Tüchler
- Department of Internal Medicine III, Hanusch Hospital, 1140 Vienna, Austria; (T.N.); (M.P.); (H.T.); (T.S.); (F.K.)
| | - Thamer Sliwa
- Department of Internal Medicine III, Hanusch Hospital, 1140 Vienna, Austria; (T.N.); (M.P.); (H.T.); (T.S.); (F.K.)
| | - Felix Keil
- Department of Internal Medicine III, Hanusch Hospital, 1140 Vienna, Austria; (T.N.); (M.P.); (H.T.); (T.S.); (F.K.)
| | - Christoph Geissler
- Department of Laboratory Medicine, Hospital Hietzing, 1130 Vienna, Austria;
| | - Sonja Heibl
- Department of Internal Medicine IV, Hospital Wels-Grieskirchen, 4600 Wels, Austria; (S.H.); (J.T.)
| | - Josef Thaler
- Department of Internal Medicine IV, Hospital Wels-Grieskirchen, 4600 Wels, Austria; (S.H.); (J.T.)
| | - Sigrid Machherndl-Spandl
- Department of Internal Medicine I with Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz Barmherzige Schwestern - Elisabethinen, 4020 Linz, Austria; (S.M.-S.); (O.Z.); (A.W.); (P.B.)
| | - Otto Zach
- Department of Internal Medicine I with Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz Barmherzige Schwestern - Elisabethinen, 4020 Linz, Austria; (S.M.-S.); (O.Z.); (A.W.); (P.B.)
| | - Ansgar Weltermann
- Department of Internal Medicine I with Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz Barmherzige Schwestern - Elisabethinen, 4020 Linz, Austria; (S.M.-S.); (O.Z.); (A.W.); (P.B.)
| | - Peter Bettelheim
- Department of Internal Medicine I with Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz Barmherzige Schwestern - Elisabethinen, 4020 Linz, Austria; (S.M.-S.); (O.Z.); (A.W.); (P.B.)
| | - Reinhard Stauder
- Internal Medicine V with Hematology and Oncology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Armin Zebisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, 8036 Graz, Austria; (A.Z.); (H.S.)
- Otto-Loewi-Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8036 Graz, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, 8036 Graz, Austria; (A.Z.); (H.S.)
| | - Ilse Schwarzinger
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; (E.J.); (M.G.); (I.S.)
| | - Bruno Schneeweiss
- Department of Internal Medicine, Hospital Kirchdorf, 4560 Kirchdorf, Austria;
| | - Leopold Öhler
- Department of Internal Medicine/Oncology, St. Josef Hospital, 1130 Vienna, Austria;
| | - Ernst Ulsperger
- Department of Internal Medicine, Hospital Horn, 3580 Horn, Austria;
| | - Rajko Kusec
- School of Medicine, University of Zagreb, University Hospital Dubrava, 10000 Zagreb, Croatia;
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.K.); (U.J.); (P.V.)
| | - Paul Knöbl
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.K.); (U.J.); (P.V.)
| | - Ulrich Jäger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.K.); (U.J.); (P.V.)
| | - Gregor Hörmann
- Central Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.K.); (U.J.); (P.V.)
- Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
90
|
Patnaik MM, Tefferi A. Chronic Myelomonocytic leukemia: 2020 update on diagnosis, risk stratification and management. Am J Hematol 2020; 95:97-115. [PMID: 31736132 DOI: 10.1002/ajh.25684] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
Abstract
DISEASE OVERVIEW Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder with overlapping features of myelodysplastic syndromes and myeloproliferative neoplasms, with an inherent risk for leukemic transformation (~15% over 3-5 years). DIAGNOSIS Diagnosis is based on the presence of sustained (>3 months) peripheral blood monocytosis (≥1 × 109 /L; monocytes ≥10%), along with bone marrow dysplasia. Clonal cytogenetic abnormalities occur in ~ 30% of patients, while >90% have gene mutations. Mutations involving TET2 (~60%), SRSF2 (~50%), ASXL1 (~40%) and the oncogenic RAS pathway (~30%) are frequent; while the presence of ASXL1 and DNMT3A mutations and the absence of TET2 mutations negatively impact over-all survival. RISK STRATIFICATION Molecularly integrated prognostic models include; the Groupe Français des Myélodysplasies (GFM), Mayo Molecular Model (MMM) and the CMML specific prognostic model (CPSS-Mol). Risk factors incorporated into the MMM include presence of nonsense or frameshift ASXL1 mutations, absolute monocyte count>10 × 109 /L, hemoglobin <10 g/dL, platelet count <100 × 109 /L and the presence of circulating immature myeloid cells. The MMM stratifies CMML patients into four groups; high (≥3 risk factors), intermediate-2 (2 risk factors), intermediate-1 (1 risk factor) and low (no risk factors), with median survivals of 16, 31, 59 and 97 months, respectively. RISK-ADAPTED THERAPY Hypomethylating agents such as 5-azacitidine and decitabine are commonly used, with overall response rates of ~40%-50% and complete remission rates of ~7%-17%; with no impact on mutational allele burdens. Allogeneic stem cell transplant is the only potentially curative option, but is associated with significant morbidity and mortality.
Collapse
Affiliation(s)
- Mrinal M. Patnaik
- Division of Hematology, Department of MedicineMayo Clinic Rochester Minnesota
| | - Ayalew Tefferi
- Division of Hematology, Department of MedicineMayo Clinic Rochester Minnesota
| |
Collapse
|
91
|
Sangiorgio VFI, Orazi A, Arber DA. Myelodysplastic/myeloproliferative neoplasms: are morphology and immunophenotyping still relevant? Best Pract Res Clin Haematol 2019; 33:101139. [PMID: 32460987 DOI: 10.1016/j.beha.2019.101139] [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] [Received: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 01/16/2023]
Abstract
The term myelodysplastic/myeloproliferative neoplasm (MDS/MPN) refers to a group of clonal hematopoietic neoplasms with overlapping clinical, morphologic and genetic myelodysplastic and myeloproliferative features observed at the time of first presentation. Impaired hematopoiesis morphologically associated with evidence of myelodysplasia manifests clinically with cytopenia/s. Simultaneously, myeloproliferation is seen within the bone marrow and leads to cytosis in the peripheral blood. The diagnostic category of MDS/MPN encompasses a heterogeneous group of diseases which share similarities among them, but at the same time have distinct clinical and pathologic features and eventually diverse prognosis; such differences justify their separation in a classification scheme. In the era of genetic and genomic tests, their distinction from conventional myelodysplastic syndromes or myeloproliferative neoplasms still relies on close clinocopathological correlation, with evaluation of both peripheral blood and bone marrow samples being essential in this sense. A multiparametric integration of clinicopathologic data and cytogenetics and molecular genetics results is the preferred diagnostic approach.
Collapse
Affiliation(s)
- V F I Sangiorgio
- Department of Cellular Pathology, The Royal London Hospital, London, UK
| | - A Orazi
- Department of Pathology, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - D A Arber
- Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
92
|
Genotypic and phenotypic evolution in a patient with chronic myelomonocytic leukemia. Leuk Res Rep 2019; 12:100185. [PMID: 31867203 PMCID: PMC6904770 DOI: 10.1016/j.lrr.2019.100185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/21/2019] [Accepted: 10/01/2019] [Indexed: 12/04/2022] Open
Abstract
The correlation of molecular and phenotypic evolution in individual patients with chronic myelomonocytic leukemia (CMML) is poorly investigated. The longitudinal follow up of a CMML patient for more than 10 years illustrates that the emergence of clones harboring mutations in TET2, SRSF2, RUNX1, MPL, NRAS, and finally in multiple genes, respectively, was mirrored by thrombocytopenia, thrombocytosis, myeloproliferation and transformation into acute myeloid leukemia. Moreover, molecular aberrations of the RAS genes were associated with markedly increased spontaneous in vitro myeloid colony formation which has been shown to be a functional indicator of RAS pathway hyperactivation.
Collapse
|
93
|
Sangiorgio VFI, Arber DA, Orazi A. How I investigate chronic myelomonocytic leukemia. Int J Lab Hematol 2019; 42:101-108. [PMID: 31841277 DOI: 10.1111/ijlh.13145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022]
Abstract
The 2016 revised 4th edition of the World Health Organization classification of hematopoietic neoplasms updated the diagnostic criteria for chronic myelomonocytic leukemia (CMML). Persistent peripheral blood monocytosis of at least 1 × 109 /L and a percentage of monocytes ≥10% of the circulating white blood cell count (WBC) are both prerequisite criteria for this diagnosis. CMML represents the prototype of "overlapping" myeloid neoplasms with concurrent myeloproliferative and myelodysplastic features. However, clinical presentation is heterogeneous, with cases showing prevailing "dysplastic" features and others a predominant "proliferative" phenotype. Accounting for this diversity, two variants of CMML are recognized: "dysplastic" CMML defined by WBC < 13 × 109 /L and "proliferative" CMML with WBC ≥ 13 × 109 /L often showing features mimicking a myeloproliferative neoplasm. Although not an official WHO category, the "oligomonocytic" variant of CMML is defined by relative monocytosis with an absolute monocyte count of 0.5-0.9 × 109 /L. It can be considered a "pre-phase," as it frequently anticipates the development of an overt, classic CMML. In an attempt at improving disease prognostication, the blast count based grading system for CMML of the WHO 2008 Classification has been expanded in 2016 to include a new "CMML-0" category. Lastly, the large body of knowledge on the molecular events occurring in CMML has been used to assist diagnosis and assess prognosis. Despite the step forwards, diagnosis of CMML still remains one of exclusion as no clinical, pathologic or molecular findings are specific for this disease. The current review brings insight into the spectrum of CMML and provides practical advice to approach suspected cases of CMML.
Collapse
Affiliation(s)
| | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Attilio Orazi
- Department of Pathology, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| |
Collapse
|
94
|
Valent P. Oligo-monocytic CMML and other pre-CMML states: Clinical impact, prognostication and management. Best Pract Res Clin Haematol 2019; 33:101137. [PMID: 32460976 DOI: 10.1016/j.beha.2019.101137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 11/18/2022]
Abstract
Chronic myelomonocytic leukemia (CMML) is defined by myelodysplasia, pathologic accumulation of monocytes and a substantial risk to transform to secondary acute myeloid leukemia (sAML). In recent years, minimal diagnostic criteria for classical CMML and CMML-variants were proposed. Moreover, potential pre-stages of CMML and interface conditions have been postulated. Oligomonocytic CMML is a condition where the absolute peripheral blood monocyte count does not reach a diagnostic level but all other criteria for CMML are fulfilled. Among potential pre-stages of CMML, clonal and non-clonal conditions have been described, including idiopathic monocytosis (IMUS) and clonal monocytosis of unknown significance (CMUS). Patients with myelodysplastic syndromes (MDS), clonal cytopenia of unknown significance (CCUS), clonal hematopoiesis of indeterminate potential (CHIP) and idiopathic cytopenia of undetermined significance (ICUS) may also progress to CMML. The current article provides an overview of pre-CMML conditions and oligomonocytic CMML, with special reference to diagnostic criteria, differential diagnoses, clinical outcomes and management.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Vienna, Austria.
| |
Collapse
|
95
|
Solary E, Wagner-Ballon O, Selimoglu-Buet D. Incorporating flow cytometry and next-generation sequencing in the diagnosis of CMML. Are we ready for prime? Best Pract Res Clin Haematol 2019; 33:101134. [PMID: 32460985 DOI: 10.1016/j.beha.2019.101134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
In the last version of the WHO classification of myeloid malignancies, flow cytometry and molecular investigation are listed as potentially useful, yet non-essential diagnostic tools in hard-to-recognize chronic myelomonocytic leukemias (CMML). Flow recognition of CMML was initially based on an increase in the fraction of peripheral blood, CD14+,CD16- classical monocytes ≥94% of total monocytes. An associated inflammatory disease can preclude the detection of classical monocyte fraction increase by inducing accumulation of CD14+,CD16+ intermediate monocytes. In such a situation, decrease in the Slan+,CD14low,CD16+ non-classical monocyte fraction below 1.7% still supports CMML diagnosis. This robust, two-step flow cytometry assay identifies CMML with a very high sensitivity. Otherwise, detection of one or several acquired gene mutations with high variant allele frequency supports the diagnosis of CMML, oligomonocytic CMML or clonal monocytosis of clinical significance. Together, recent investigations support integration of flow cytometry analysis of peripheral blood monocyte subsets and new generation sequencing of a panel of 20-30 recurrently mutated genes in the diagnostic work-up of CMML.
Collapse
Affiliation(s)
- Eric Solary
- INSERM U1170, Gustave Roussy Cancer Center, Villejuif, France; Faculté de Médicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France; Département D'Hématologie, Gustave Roussy Cancer Center, Villejuif, France.
| | - Orianne Wagner-Ballon
- INSERM U1170, Gustave Roussy Cancer Center, Villejuif, France; Département D'Hématologie et Immunologie Biologiques, Hôpitaux Universitaires Henri Mondor, APHP, Créteil, France
| | - Dorothée Selimoglu-Buet
- INSERM U1170, Gustave Roussy Cancer Center, Villejuif, France; Faculté de Médicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
96
|
Hofer TP, van de Loosdrecht AA, Stahl-Hennig C, Cassatella MA, Ziegler-Heitbrock L. 6-Sulfo LacNAc (Slan) as a Marker for Non-classical Monocytes. Front Immunol 2019; 10:2052. [PMID: 31572354 PMCID: PMC6753898 DOI: 10.3389/fimmu.2019.02052] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Monocytes are subdivided into three subsets, which have different phenotypic and functional characteristics and different roles in inflammation and malignancy. When in man CD14 and CD16 monoclonal antibodies are used to define these subsets, then the distinction of non-classical CD14low and intermediate CD14high monocytes requires setting a gate in what is a gradually changing level of CD14 expression. In the search for an additional marker to better dissect the two subsets we have explored the marker 6-sulfo LacNAc (slan). Slan is a carbohydrate residue originally described to be expressed on the cell surface of a type of dendritic cell in human blood. We elaborate herein that the features of slan+ cells are congruent with the features of CD16+ non-classical monocytes and that slan is a candidate marker for definition of non-classical monocytes. The use of this marker may help in studying the role of non-classical monocytes in health and in diagnosis and monitoring of disease.
Collapse
Affiliation(s)
- Thomas P Hofer
- Immunoanalytics Core Facility and RG Tissue Control of Immunocytes, Helmholtz Centre Munich, Munich, Germany
| | | | | | - Marco A Cassatella
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | | |
Collapse
|