51
|
Kayano H. Histopathology in the diagnosis of high-risk myelodysplastic syndromes. J Clin Exp Hematop 2018; 58:51-60. [PMID: 29998976 DOI: 10.3960/jslrt.18009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
52
|
Hebeda KM, Tzankov A, Boudova L, Saft L, Hasserjian RP, de Boer M, Fend F, Orazi A, Leguit R. Challenges in Diagnosing Myelodysplastic Syndromes in the Era of Genetic Testing: Proceedings of the 13th Workshop of the European Bone Marrow Working Group. Pathobiology 2018; 86:62-75. [PMID: 29982244 PMCID: PMC6482987 DOI: 10.1159/000489678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/30/2018] [Indexed: 11/19/2022] Open
Abstract
The 13th workshop of the European Bone Marrow Working Group in Utrecht, The Netherlands, was devoted to studying myelodysplastic syndromes (MDS) and their boundaries. The panel received 44 cases submitted to the 3 invited categories, which included: reactive cytopenias with dysplasia, idiopathic cytopenia of undetermined significance, clonal haematopoiesis of indeterminate potential, idiopathic dysplasia of uncertain significance and overt MDS. For this summary, we have selected 17 cases that highlight difficulties in separating true MDS from other causes of cytopenia and the intricate relationship between clonal haematopoiesis and true MDS. In addition, cases of overt MDS with challenging features were also selected. All cases were stained for p53 expression. Using instructive submitted cases we discuss the following: (1) cytopenia with clonal haematopoiesis not fulfilling MDS criteria, (2) cytopenia and/or dysplasia with germline mutations and/or familial history suggesting an underlying gene defect, (3) MDS based on a recurrent chromosomal abnormality and (4) overt MDS with diagnostic difficulties due to concurrent treatment or disease. The lively discussion in the open forum of the workshop illustrated the need for better integrative understanding of the evolution of acquired genetic abnormalities in haematopoiesis, and the challenge of diagnosing true MDS in cytopenic patients with genetic abnormalities, either germline or acquired.
Collapse
Affiliation(s)
- Konnie M Hebeda
- Department of Pathology, Radboud University Medical Center, Nijmegen, The
| | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Ludmila Boudova
- Department of Pathology, Charles University Hospital, Pilsen, Czech Republic
| | - Leonie Saft
- Department of Pathology and Cytology, Karolinska University Hospital and Institute, Stockholm, Sweden
| | - Rob P Hasserjian
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mirthe de Boer
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Falko Fend
- Department of Pathology, University of Tuebingen, Tuebingen, Germany
| | - Attilio Orazi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Roos Leguit
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
53
|
Kuykendall A, Duployez N, Boissel N, Lancet JE, Welch JS. Acute Myeloid Leukemia: The Good, the Bad, and the Ugly. Am Soc Clin Oncol Educ Book 2018; 38:555-573. [PMID: 30231330 DOI: 10.1200/edbk_199519] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Acute myeloid leukemia (AML) was initially subdivided according to morphology (the French-American-British system), which proved helpful in pathologic categorization. Subsequently, clinical and genomic factors were found to correlate with response to chemotherapy and with overall survival. These included a history of antecedent hematologic disease, a history of chemotherapy or radiation therapy, the presence of various recurrent cytogenetic abnormalities, and, more recently, the presence of specific point mutations. This article reviews the biology and responses of one AML subgroup with consistent response and good outcomes following chemotherapy (core-binding factor leukemia), and two subgroups with persistently bad, and even ugly, outcomes (secondary AML and TP53-mutated AML).
Collapse
MESH Headings
- Alleles
- Biomarkers, Tumor
- Chromosome Aberrations
- Combined Modality Therapy
- Core Binding Factors/genetics
- Core Binding Factors/metabolism
- Gene Frequency
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/mortality
- Mutation
- Neoplasm, Residual/diagnosis
- Neoplasms, Second Primary/diagnosis
- Neoplasms, Second Primary/epidemiology
- Neoplasms, Second Primary/etiology
- Neoplasms, Second Primary/therapy
- Signal Transduction
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Andrew Kuykendall
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Nicolas Duployez
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Nicolas Boissel
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Jeffrey E Lancet
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - John S Welch
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
54
|
Early detection and evolution of preleukemic clones in therapy-related myeloid neoplasms following autologous SCT. Blood 2018; 131:1846-1857. [DOI: 10.1182/blood-2017-09-805879] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 12/27/2017] [Indexed: 12/28/2022] Open
Abstract
Key Points
tMNs after ASCT originate from HSCs bearing (pre-)tMN mutations that are present years before disease onset. Post-ASCT treatment can influence selection and outgrowth of (pre)leukemic clones.
Collapse
|
55
|
Yao CY, Hou HA, Lin TY, Lin CC, Chou WC, Tseng MH, Chiang YC, Liu MC, Liu CW, Kuo YY, Wu SJ, Liao XW, Lin CT, Ko BS, Chen CY, Hsu SC, Li CC, Huang SY, Yao M, Tang JL, Tsay W, Liu CY, Tien HF. Distinct mutation profile and prognostic relevance in patients with hypoplastic myelodysplastic syndromes (h-MDS). Oncotarget 2018; 7:63177-63188. [PMID: 27527853 PMCID: PMC5325355 DOI: 10.18632/oncotarget.11050] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/10/2016] [Indexed: 11/25/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of hematologic malignancies. Although most MDS patients have normal or increased BM cellularity (NH-MDS), some have hypocellular BM (h-MDS). The reports concerning the differences in genetic alterations between h-MDS and NH-MDS patients are limited. In this study, 369 MDS patients diagnosed according to the WHO 2008 criteria were recruited. h-MDS patients had lower PB white blood cell and blast counts, and lower BM blast percentages, than those with NH-MDS. h-MDS was closely associated with lower-risk MDS, defined by the International Prognostic Scoring System (IPSS) and revised IPSS (IPSS-R). IPSS-R could properly predict the prognosis in h-MDS (P<0.001) as in NH-MDS patients. The h-MDS patients had lower incidences of RUNX1, ASXL1, DNMT3A, EZH2 and TP53 mutations than NH-MDS patients. The cumulated incidence of acute leukemic transformation at 5 years was 19.3% for h-MDS and 40.4% for NH-MDS patients (P= 0.001). Further, the patients with h-MDS had longer overall survival (OS) than those with NH-MDS (P= 0.001), and BM hypocellularity remains an independent favorable prognostic factor for OS irrespective of age, IPSS-R, and gene mutations. Our findings provide evidence that h-MDS indeed represent a distinct clinico-biological subgroup of MDS and can predict better leukemia-free survival and OS.
Collapse
Affiliation(s)
- Chi-Yuan Yao
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzung-Yi Lin
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Chin Lin
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Hsuan Tseng
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Chieh Chiang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Chih Liu
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Wen Liu
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yuan-Yeh Kuo
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shang-Ju Wu
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Xiu-Wen Liao
- Tai-Cheng Stem Cell Therapy Center, National Taiwan University, Taipei, Taiwan
| | - Chien-Ting Lin
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Tai-Cheng Stem Cell Therapy Center, National Taiwan University, Taipei, Taiwan
| | - Bor-Shen Ko
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Yuan Chen
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-Chun Hsu
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Cheng Li
- Tai-Cheng Stem Cell Therapy Center, National Taiwan University, Taipei, Taiwan
| | - Shang-Yi Huang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming Yao
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jih-Luh Tang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Tai-Cheng Stem Cell Therapy Center, National Taiwan University, Taipei, Taiwan
| | - Woei Tsay
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chieh-Yu Liu
- Biostatistics Consulting Laboratory, Department of Nursing, National Taipei College of Nursing, Taipei, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
56
|
Bustelo XR, Dosil M. Ribosome biogenesis and cancer: basic and translational challenges. Curr Opin Genet Dev 2018; 48:22-29. [DOI: 10.1016/j.gde.2017.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/13/2017] [Accepted: 10/02/2017] [Indexed: 01/08/2023]
|
57
|
A decade of progress in myelodysplastic syndrome with chromosome 5q deletion. Leukemia 2018; 32:1493-1499. [DOI: 10.1038/s41375-018-0029-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/28/2017] [Accepted: 01/05/2018] [Indexed: 12/26/2022]
|
58
|
Scharenberg C, Jansson M, Saft L, Hellström-Lindberg E. Megakaryocytes harbour the del(5q) abnormality despite complete clinical and cytogenetic remission induced by lenalidomide treatment. Br J Haematol 2018; 180:526-533. [DOI: 10.1111/bjh.15094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/07/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Christian Scharenberg
- Department of Medicine; Centre for Haematology and Regenerative Medicine; Karolinska University Hospital Huddinge; Karolinska Institutet; Stockholm Sweden
- Department of Medicine; Division of Haematology; Skaraborgs Hospital Skövde; Stockholm Sweden
| | - Monika Jansson
- Department of Medicine; Centre for Haematology and Regenerative Medicine; Karolinska University Hospital Huddinge; Karolinska Institutet; Stockholm Sweden
| | - Leonie Saft
- Department of Pathology; Karolinska University Hospital, Solna, and Karolinska Institutet; Stockholm Sweden
| | - Eva Hellström-Lindberg
- Department of Medicine; Centre for Haematology and Regenerative Medicine; Karolinska University Hospital Huddinge; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
59
|
Ramos F, Robledo C, Izquierdo-García FM, Suárez-Vilela D, Benito R, Fuertes M, Insunza A, Barragán E, Del Rey M, García-Ruiz de Morales JM, Tormo M, Salido E, Zamora L, Pedro C, Sánchez-Del-Real J, Díez-Campelo M, Del Cañizo C, Sanz GF, Hernández-Rivas JM. Bone marrow fibrosis in myelodysplastic syndromes: a prospective evaluation including mutational analysis. Oncotarget 2017; 7:30492-503. [PMID: 27127180 PMCID: PMC5058695 DOI: 10.18632/oncotarget.9026] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/17/2016] [Indexed: 02/07/2023] Open
Abstract
The biological and molecular events that underlie bone marrow fibrosis in patients with myelodysplastic syndromes are poorly understood, and its prognostic role in the era of the Revised International Prognostic Scoring System (IPSS-R) is not yet fully determined. We have evaluated the clinical and biological events that underlie bone marrow fibrotic changes, as well as its prognostic role, in a well-characterized prospective patient cohort (n=77) of primary MDS patients. The degree of marrow fibrosis was linked to parameters of erythropoietic failure, marrow cellularity, p53 protein accumulation, WT1 gene expression, and serum levels of CXCL9 and CXCL10, but not to other covariates including the IPSS-R score. The presence of bone marrow fibrosis grade 2 or higher was associated with the presence of mutations in cohesin complex genes (31.5% vs. 5.4%, p=0.006). By contrast, mutations in CALR, JAK2, PDGFRA, PDGFRB,and TP53 were very rare. Survival analysis showed that marrow fibrosis grade 2 or higher was a relevant significant predictor for of overall survival, and independent of age, performance status, and IPSS-R score in multivariate analysis.
Collapse
Affiliation(s)
- Fernando Ramos
- Department of Hematology, Hospital Universitario de León, León, Spain.,Instituto de Biomedicina (IBIOMED), Universidad de León, León, Spain
| | - Cristina Robledo
- Unidad de Diagnóstico Molecular y Celular del Cáncer, IBSAL, IBMCC-Centro de Investigación del Cáncer (USAL-CSIC), Salamanca, Spain
| | | | | | - Rocío Benito
- Unidad de Diagnóstico Molecular y Celular del Cáncer, IBSAL, IBMCC-Centro de Investigación del Cáncer (USAL-CSIC), Salamanca, Spain
| | - Marta Fuertes
- Department of Hematology, Hospital Universitario de León, León, Spain
| | - Andrés Insunza
- Department of Hematology, Hospital Universitario U. Marqués de Valdecilla, Santander, Spain
| | - Eva Barragán
- Department of Molecular Pathology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Mónica Del Rey
- Unidad de Diagnóstico Molecular y Celular del Cáncer, IBSAL, IBMCC-Centro de Investigación del Cáncer (USAL-CSIC), Salamanca, Spain
| | | | - Mar Tormo
- Department of Hematology-Oncology, Hospital Clínico Universitario, Valencia, Spain
| | - Eduardo Salido
- Department of Hematology, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Lurdes Zamora
- Unit of Molecular Genetics, ICO-Hospital Germans Trias i Pujol, Institut de Recerca contra la Leucèmia Josep Carreras, Badalona, Spain
| | - Carmen Pedro
- Department of Hematology, Hospital del Mar, Barcelona, Spain
| | | | | | | | - Guillermo F Sanz
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Jesús María Hernández-Rivas
- Unidad de Diagnóstico Molecular y Celular del Cáncer, IBSAL, IBMCC-Centro de Investigación del Cáncer (USAL-CSIC), Salamanca, Spain.,Department of Hematology, Hospital Universitario de Salamanca, Spain
| | | |
Collapse
|
60
|
Giagounidis A. Current treatment algorithm for the management of lower-risk MDS. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:453-459. [PMID: 29222293 PMCID: PMC6142548 DOI: 10.1182/asheducation-2017.1.453] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Lower risk myelodysplastic syndromes (MDS), defined as MDS with a Revised International Prognostic Scoring System score ≤3.5 points, will remain a challenging entity in 2018. Supportive care continues to be the linchpin of treatment, although the options to reduce transfusion needs are broadening. To achieve red blood cell transfusion independence in non-del(5q) patients, erythropoiesis-stimulating agents remain a mainstay of therapy as long as endogenous erythropoietin levels are <500 U/L (and preferably <200 U/L). Experimental strategies for patients ineligible for erythropoiesis-stimulating agents or relapsing after gaining transfusion independence include immunosuppressive agents, transforming growth factor β inhibitors, and lenalidomide. All these alternatives have shown reasonable response rates in selected patient populations with lower risk MDS. Patients with del(5q) disease can derive long-term benefit from lenalidomide, and some patients remain transfusion free for extended periods even after discontinuation of the drug. In rare cases in which thrombocytopenia is the main clinical problem leading to clinically significant bleeding events, thrombopoietin receptor analogues may alleviate bleeding, increase platelet counts, and rarely lead to trilineage responses. It seems prudent to use these drugs only in patients with confirmed bone marrow blast counts <5%. Allogeneic stem cell transplantation is reasonable for patients with high molecular risk of progression and those failing several lines of treatment with signs of progression toward higher-risk MDS.
Collapse
Affiliation(s)
- Aristoteles Giagounidis
- Department of Hematology, Oncology and Palliative Care, Marien Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
61
|
Li X, Wei J, Liu Y, Li P, Fan L, Wang Y, Li M, Zhao D, Yu Z, Ye J, Guo Y, Yan Q, Guo S, Wang Z. Primary Astrocytic Tumours and Paired Recurrences have Similar Biological Features in IDH1, TP53 and TERTp Mutation and MGMT, ATRX Loss. Sci Rep 2017; 7:13038. [PMID: 29026176 PMCID: PMC5638900 DOI: 10.1038/s41598-017-13272-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/21/2017] [Indexed: 11/09/2022] Open
Abstract
Astrocytic tumours are the most common type of primary malignant brain tumour. Most astrocytic tumours will recur at some point after surgery. Currently, the combination of radiotherapy and chemotherapy does not prevent the recurrence of astrocytic tumours. In this study, we investigated the consistency in isocitrate dehydrogenase 1 (IDH1), tumour protein p53 (TP53) and telomerase reverse transcriptase promoter (TERTp) mutations during astrocytic tumour recurrence. We also evaluated the protein loss of O-6-methylguanine-DNA methyltransferase (MGMT) and alpha-thalassemia/mental retardation, X-linked (ATRX) during disease recurrence. We then determined the prognostic significance of these findings in terms of progression-free survival (PFS) using Kaplan-Meier analysis and Cox regression models. Our results showed that in most cases, IDH1, TP53 and TERTp mutation status and MGMT and ATRX protein expression levels were stable during recurrence, which may indicate that these alterations occurred early in astrocytic tumour development. Furthermore, in IDH1 wild type group, the patients who were negative for MGMT and had a low Ki67 index showed a longer PFS. Therefore, we suggest that IDH1 mutation combined with MGMT expression level and Ki67 index might be an effective biomarker panel for evaluating the PFS of patients with astrocytic tumours.
Collapse
Affiliation(s)
- Xia Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Jie Wei
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Yixiong Liu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Peifeng Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Linni Fan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Yingmei Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Danhui Zhao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Zhou Yu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Jing Ye
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Ying Guo
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Qingguo Yan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Shuangping Guo
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China
| | - Zhe Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital; and School of Basic Medicine, The Fourth Military Medical University, Xi'an, Shaan Xi Province, China.
| |
Collapse
|
62
|
Hong M, He G. The 2016 Revision to the World Health Organization Classification of Myelodysplastic Syndromes. J Transl Int Med 2017; 5:139-143. [PMID: 29085786 DOI: 10.1515/jtim-2017-0002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Ming Hong
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu Province, China
| | - Guangsheng He
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
63
|
Tosato V, West N, Zrimec J, Nikitin DV, Del Sal G, Marano R, Breitenbach M, Bruschi CV. Bridge-Induced Translocation between NUP145 and TOP2 Yeast Genes Models the Genetic Fusion between the Human Orthologs Associated With Acute Myeloid Leukemia. Front Oncol 2017; 7:231. [PMID: 29034209 PMCID: PMC5626878 DOI: 10.3389/fonc.2017.00231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 09/07/2017] [Indexed: 01/03/2023] Open
Abstract
In mammalian organisms liquid tumors such as acute myeloid leukemia (AML) are related to spontaneous chromosomal translocations ensuing in gene fusions. We previously developed a system named bridge-induced translocation (BIT) that allows linking together two different chromosomes exploiting the strong endogenous homologous recombination system of the yeast Saccharomyces cerevisiae. The BIT system generates a heterogeneous population of cells with different aneuploidies and severe aberrant phenotypes reminiscent of a cancerogenic transformation. In this work, thanks to a complex pop-out methodology of the marker used for the selection of translocants, we succeeded by BIT technology to precisely reproduce in yeast the peculiar chromosome translocation that has been associated with AML, characterized by the fusion between the human genes NUP98 and TOP2B. To shed light on the origin of the DNA fragility within NUP98, an extensive analysis of the curvature, bending, thermostability, and B-Z transition aptitude of the breakpoint region of NUP98 and of its yeast ortholog NUP145 has been performed. On this basis, a DNA cassette carrying homologous tails to the two genes was amplified by PCR and allowed the targeted fusion between NUP145 and TOP2, leading to reproduce the chimeric transcript in a diploid strain of S. cerevisiae. The resulting translocated yeast obtained through BIT appears characterized by abnormal spherical bodies of nearly 500 nm of diameter, absence of external membrane and defined cytoplasmic localization. Since Nup98 is a well-known regulator of the post-transcriptional modification of P53 target genes, and P53 mutations are occasionally reported in AML, this translocant yeast strain can be used as a model to test the constitutive expression of human P53. Although the abnormal phenotype of the translocant yeast was never rescued by its expression, an exogenous P53 was recognized to confer increased vitality to the translocants, in spite of its usual and well-documented toxicity to wild-type yeast strains. These results obtained in yeast could provide new grounds for the interpretation of past observations made in leukemic patients indicating a possible involvement of P53 in cell transformation toward AML.
Collapse
Affiliation(s)
- Valentina Tosato
- Ulisse Biomed S.r.l., AREA Science Park, Trieste, Italy.,Faculty of Health Sciences, University of Primorska, Izola, Slovenia.,Yeast Molecular Genetics, ICGEB, AREA Science Park, Trieste, Italy
| | - Nicole West
- Clinical Pathology, Hospital Maggiore, Trieste, Italy
| | - Jan Zrimec
- Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Dmitri V Nikitin
- Biology Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Roberto Marano
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michael Breitenbach
- Genetics Division, Department of Cell Biology, University of Salzburg, Salzburg, Austria
| | - Carlo V Bruschi
- Yeast Molecular Genetics, ICGEB, AREA Science Park, Trieste, Italy.,Genetics Division, Department of Cell Biology, University of Salzburg, Salzburg, Austria
| |
Collapse
|
64
|
Sallman DA, Tanaka TN, List A, Bejar R. SOHO State of the Art Update and Next Questions: Biology and Treatment of Myelodysplastic Syndromes. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:613-620. [PMID: 29025689 DOI: 10.1016/j.clml.2017.09.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/19/2017] [Indexed: 11/17/2022]
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of myeloid neoplasms characterized by clonal hematopoiesis leading to bone marrow dysplasia and cytopenias. Recently, significant advancements have been made in understanding the pathogenic mechanisms of this disease. In particular, how a wide array of somatic mutations can induce a common clinical phenotype has been investigated. Specifically, activation of innate immune signaling (i.e. myeloid derived suppressor cells) and the NLRP3 inflammasome in hematopoietic stem/progenitor cells play a central role in the biology of MDS, leading to pyroptotic cell death and clonal expansion. Additionally, deciphering the molecular drivers of MDS using next-generation sequencing has rapidly expanded our understanding of MDS with profound implications for prognosis, treatment decisions, and future clinical investigations. Together, unraveling of the role of innate immunity/pyroptosis in the clinical phenotype of MDS patients and comprehensive molecular characterization has identified novel therapeutic strategies that offer significant promise.
Collapse
Affiliation(s)
- David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Tiffany N Tanaka
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Alan List
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL.
| | - Rafael Bejar
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|
65
|
Santini V. First-line Therapeutic Strategies for Myelodysplastic Syndromes. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17S:S31-S36. [DOI: 10.1016/j.clml.2017.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 01/27/2023]
|
66
|
Progress in Myelodysplastic Syndromes: Clinicopathologic Correlations and Immune Checkpoints. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17S:S16-S25. [DOI: 10.1016/j.clml.2017.02.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/28/2017] [Indexed: 12/22/2022]
|
67
|
Fenaux P, Giagounidis A, Selleslag D, Beyne-Rauzy O, Mittelman M, Muus P, Nimer SD, Hellström-Lindberg E, Powell BL, Guerci-Bresler A, Sekeres MA, Deeg HJ, Del Cañizo C, Greenberg PL, Shammo JM, Skikne B, Yu X, List AF. Clinical characteristics and outcomes according to age in lenalidomide-treated patients with RBC transfusion-dependent lower-risk MDS and del(5q). J Hematol Oncol 2017. [PMID: 28651604 PMCID: PMC5485496 DOI: 10.1186/s13045-017-0491-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Particularly since the advent of lenalidomide, lower-risk myelodysplastic syndromes (MDS) patients with del(5q) have been the focus of many studies; however, the impact of age on disease characteristics and response to lenalidomide has not been analyzed. Methods We assessed the effect of age on clinical characteristics and outcomes in 286 lenalidomide-treated MDS patients with del(5q) from two multicenter trials. Results A total of 33.9, 34.3, and 31.8% patients were aged <65 years, ≥65 to <75 years, and ≥75 years, respectively. Age <65 years was associated with less favorable International Prognostic Scoring System (IPSS) risk and additional cytopenias at baseline versus older age groups, significantly lower cytogenetic response rates (p = 0.022 vs. ≥65 to <75 years; p = 0.047 vs. ≥75 years), and higher rates of acute myeloid leukemia (AML) progression (Gray’s test, p = 0.013). Lenalidomide was equally well tolerated across age groups, producing consistently high rates of red blood cell transfusion independence ≥26 weeks. Conclusions Baseline disease characteristics and AML progression appear to be more severe in younger lower-risk MDS patients with del(5q), whereas older age does not seem to compromise the response to lenalidomide. Trial registration ClinicalTrials.gov NCT00065156 and NCT00179621 Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0491-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pierre Fenaux
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Université Paris 7, 1 Avenue Claude Vellefaux, 75475, Paris, France.
| | | | | | - Odile Beyne-Rauzy
- Purpan Pavillion de Medecines, Centre Hospitalier Universitaire, Toulouse, France
| | | | - Petra Muus
- Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - Bayard L Powell
- Comprehensive Cancer Center of Wake Forest University, Winston-Salem, NC, USA
| | | | | | - H Joachim Deeg
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | - Xujie Yu
- Celgene Corporation, Summit, NJ, USA
| | - Alan F List
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
68
|
Prebet T, Cluzeau T, Park S, Sekeres MA, Germing U, Ades L, Platzbecker U, Gotze K, Vey N, Oliva E, Sugrue MM, Bally C, Kelaidi C, Al Ali N, Fenaux P, Gore SD, Komrokji R. Outcome of patients treated for myelodysplastic syndromes with 5q deletion after failure of lenalidomide therapy. Oncotarget 2017; 8:81926-81935. [PMID: 29137233 PMCID: PMC5669859 DOI: 10.18632/oncotarget.18477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 12/16/2022] Open
Abstract
While lenalidomide (LEN) is the standard of care for the lower-risk myelodysplastic syndromes (MDS) patients with deletion 5q, 35% will not respond to or do not tolerate the drug. Moreover, most of the patients will lose their response after a few years. Defining the outcome of patients with LEN failure and determining the impact of subsequent therapies is therefore important to develop alternative strategies. Based on an international collaboration, we were able to compile a total of 392 patient cases of lower-risk MDS patients with 5q deletion and to analyze their outcome after failure of lenalidomide. The median survival following LEN failure was 23 months. We observed a negative impact on survival of advanced age, higher bone marrow blast count at LEN initiation, progression after LEN failure, and unfavorable cytogenetics. Among the treatment strategies, we observed a relatively prolonged survival of patients treated subsequently with hypomethylating agents and only a limited impact on survival of allogeneic transplantation. In conclusion, our work stresses the relatively short survival of this group of patient and defines the expected baseline for the needed future investigations in this group of patients.
Collapse
Affiliation(s)
| | - Thomas Cluzeau
- Cote d'Azur University, Nice Sophia Antipolis University, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Sophie Park
- U1065, Mediterranean Center of Molecular Medecine, Nice, France.,Groupe Francophone des Myelodysplasies, Hopital Saint Louis, Paris, France
| | | | - Ulrich Germing
- Department Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Dusseldorf, Germany
| | - Lionel Ades
- Groupe Francophone des Myelodysplasies, Hopital Saint Louis, Paris, France.,Service Hematologie Senior, Hopital Saint Louis, Paris, France
| | | | - Katharina Gotze
- Klinikum Rechts der Isar, Technische Universität München, München, Germany
| | - Norbert Vey
- Groupe Francophone des Myelodysplasies, Hopital Saint Louis, Paris, France.,Departement d'Hematologie, Institut Paoli-Calmettes, Marseille, France
| | - Esther Oliva
- Hematology Unit, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | | | - Cecile Bally
- Groupe Francophone des Myelodysplasies, Hopital Saint Louis, Paris, France
| | - Charikleia Kelaidi
- Groupe Francophone des Myelodysplasies, Hopital Saint Louis, Paris, France
| | - Najla Al Ali
- Cote d'Azur University, Nice Sophia Antipolis University, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Pierre Fenaux
- Groupe Francophone des Myelodysplasies, Hopital Saint Louis, Paris, France.,Service Hematologie Senior, Hopital Saint Louis, Paris, France
| | | | - Rami Komrokji
- Cote d'Azur University, Nice Sophia Antipolis University, Centre Hospitalier Universitaire de Nice, Nice, France
| |
Collapse
|
69
|
Successful treatment with azacitidine for the simultaneous occurrence of multiple myeloma and acute myeloid leukemia with concomitant del(5q) and the JAK2 V617F mutation. Ann Hematol 2017; 96:1411-1413. [PMID: 28577044 PMCID: PMC5486789 DOI: 10.1007/s00277-017-3032-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 05/23/2017] [Indexed: 01/28/2023]
|
70
|
Pich A, Godio L, Davico Bonino L. p53 protein expression in patients with myelodysplasia treated with allogeneic bone marrow transplantation. Mol Clin Oncol 2017; 6:876-880. [PMID: 28588781 DOI: 10.3892/mco.2017.1225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/14/2017] [Indexed: 12/12/2022] Open
Abstract
Tumor protein 53 mutations adversely affect the prognosis of myelodysplastic syndromes (MDS); however, few studies have reported on the prognostic significance of the expression of p53 protein in MDS. The current study investigated p53 immunoreactivity (p53-IR) in bone marrow biopsies (BMBs) obtained at diagnosis from 18 patients (6 females and 12 males; mean age, 50.5 years) with MDS that underwent bone marrow transplantation (BMT) to determine the associations between clinical and histopathological data and outcome. There were 5 refractory cytopenia with multilineage dysplasia (RCMD) and 13 refractory anemia with excess blasts, type 2 (RAEB-2) cases. p53-IR was assessed as the percentage of hematopoietic cells exhibiting intense nuclear staining. The cut off for positivity was 5% of stained cells. A positive p53-IR was detected in 7 patients (38.9%) and was associated with age (P=0.005) and pattern of BM fibrosis (P=0.03). A positive p53-IR was more frequent in females, in highly cellular BMBs and in RAEB-2 cases. Overall survival (OS) was associated with patients' age (P=0.01), hemoglobin level (P=0.04), type of MDS (P=0.05), degree of BM fibrosis (P=0.006) and number of BM blasts (P=0.05). The OS of patients with negative p53-IR tended to be longer compared with that of patients with positive p53-IR, although this difference was not statistically significant (P=0.1). Despite the limitation of the low number of cases, the present results indicate that a positive p53-IR at diagnosis is associated with clinically more aggressive MDS subtypes and adverse histological prognostic factors, such as BM fibrosis. Therefore, the evaluation of p53 expression of BMBs of patients with MDS may be introduced in the histopathological work-up of the disease.
Collapse
Affiliation(s)
- Achille Pich
- Department of Molecular Biotechnology and Health Sciences, Section of Pathology, University of Turin, I-10126 Turin, Italy
| | - Laura Godio
- Azienda Ospedaliera Città della Salute e della Scienza, Anatomia e Istologia Patologica 1U, I-10126 Turin, Italy
| | - Laura Davico Bonino
- Azienda Ospedaliera Città della Salute e della Scienza, Anatomia e Istologia Patologica 1U, I-10126 Turin, Italy
| |
Collapse
|
71
|
da Silva-Coelho P, Kroeze LI, Yoshida K, Koorenhof-Scheele TN, Knops R, van de Locht LT, de Graaf AO, Massop M, Sandmann S, Dugas M, Stevens-Kroef MJ, Cermak J, Shiraishi Y, Chiba K, Tanaka H, Miyano S, de Witte T, Blijlevens NMA, Muus P, Huls G, van der Reijden BA, Ogawa S, Jansen JH. Clonal evolution in myelodysplastic syndromes. Nat Commun 2017; 8:15099. [PMID: 28429724 PMCID: PMC5530598 DOI: 10.1038/ncomms15099] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 02/24/2017] [Indexed: 02/08/2023] Open
Abstract
Cancer development is a dynamic process during which the successive accumulation of mutations results in cells with increasingly malignant characteristics. Here, we show the clonal evolution pattern in myelodysplastic syndrome (MDS) patients receiving supportive care, with or without lenalidomide (follow-up 2.5–11 years). Whole-exome and targeted deep sequencing at multiple time points during the disease course reveals that both linear and branched evolutionary patterns occur with and without disease-modifying treatment. The application of disease-modifying therapy may create an evolutionary bottleneck after which more complex MDS, but also unrelated clones of haematopoietic cells, may emerge. In addition, subclones that acquired an additional mutation associated with treatment resistance (TP53) or disease progression (NRAS, KRAS) may be detected months before clinical changes become apparent. Monitoring the genetic landscape during the disease may help to guide treatment decisions. Myelodysplastic syndromes are a broad group of haematopoietic malignancies that often progress to acute myeloid leukaemia. Here, the authors show that linear and branched evolution occurs within myelodysplastic syndrome and these patterns can be impacted by treatment.
Collapse
Affiliation(s)
- Pedro da Silva-Coelho
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands.,Department of Haematology, Centro Hospitalar de São João and Faculdade de Medicina da Universidade do Porto, Alameda Professor Hernâni Monteiro, Porto 4200-319, Portugal
| | - Leonie I Kroeze
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto-shi, Kyoto 606-8501, Japan
| | - Theresia N Koorenhof-Scheele
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Ruth Knops
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Louis T van de Locht
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Aniek O de Graaf
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Marion Massop
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Marian J Stevens-Kroef
- Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Jaroslav Cermak
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| | - Yuichi Shiraishi
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639 Japan
| | - Kenichi Chiba
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639 Japan
| | - Hiroko Tanaka
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639 Japan
| | - Satoru Miyano
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639 Japan
| | - Theo de Witte
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Nicole M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Petra Muus
- Department of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Gerwin Huls
- Department of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands.,Department of Hematology, University Medical Centre Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Bert A van der Reijden
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto-shi, Kyoto 606-8501, Japan
| | - Joop H Jansen
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
72
|
Duarte FB, Barbosa MC, Jesus Dos Santos TE, Lemes RPG, Vasconcelos JP, de Vasconcelos PRL, Rocha FD, Zalcberg I, Coutinho DF. Bone marrow fibrosis at diagnosis is associated with TP53 overexpression and adverse prognosis in low-risk myelodysplastic syndrome. Br J Haematol 2017; 181:547-549. [PMID: 28318026 DOI: 10.1111/bjh.14656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Fernando B Duarte
- Department of Surgery, University Hospital Water Cantídio, Federal University of Ceará, Fortaleza, Brazil
| | - Maritza C Barbosa
- Research Laboratory in Haemoglobinopathies and Genetics of Haematological Diseases, Federal University of Ceará, Fortaleza, Brazil
| | - Talyta E Jesus Dos Santos
- Research Laboratory in Haemoglobinopathies and Genetics of Haematological Diseases, Federal University of Ceará, Fortaleza, Brazil
| | - Romélia P G Lemes
- Research Laboratory in Haemoglobinopathies and Genetics of Haematological Diseases, Federal University of Ceará, Fortaleza, Brazil
| | - João P Vasconcelos
- Haematology Service of Walter Cantídio University Hospital, Fortaleza, Brazil
| | | | - Francisco D Rocha
- Department of Surgery, Federal University of Ceará, Fortaleza, Brazil
| | - Ilana Zalcberg
- Cellular Therapy Center of Porto Alegre, Center for Experimental Research, Porto Alegre, Rio Grande do Sul, Brazil
| | - Diego F Coutinho
- Cellular Therapy Center of Porto Alegre, Center for Experimental Research, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
73
|
Immunohistochemistry for p53 is a useful tool to identify cases of acute myeloid leukemia with myelodysplasia-related changes that are TP53 mutated, have complex karyotype, and have poor prognosis. Mod Pathol 2017; 30:382-392. [PMID: 27934876 DOI: 10.1038/modpathol.2016.206] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 12/17/2022]
Abstract
In this study, we evaluate the expression of p53 in core biopsies with acute myeloid leukemia and correlate the level of expression with acute myeloid leukemia subtype, TP53 mutation status, karyotype, and survival. Of the 143 cases evaluated, 71 fulfilled the WHO 2016 criteria for acute myeloid leukemia with myelodysplasia-related changes, 40 were acute myeloid leukemia-not otherwise specified, 25 were acute myeloid leukemia with recurrent genetic abnormalities, and 7 were therapy-related acute myeloid leukemia. By immunohistochemistry, 17% showed p53 expression in >5% of the cells. Of the 24 cases with >5% p53-positive cells, 17 were acute myeloid leukemia with myelodysplasia-related changes, 5 were acute myeloid leukemia-not otherwise specified, 1 was acute myeloid leukemia with recurrent genetic abormalities, and 1 was therapy-related acute myeloid leukemia. In cases for which data was available, expression of >5% p53-positive cells was significantly associated with genotype (n=67) and/or karyotype (n=130). Among the 115 cases for which clinical follow up was available, the overall survival of cases with p53 expression >15% (Median=102 days) was significantly shorter compared with cases with p53 expression ≤15% (Median=435 days). Within the acute myeloid leukemia with myelodysplasia-related changes group, this association remained significant, with cases with ≤15% p53-positive cells having a median overall survival of 405 days versus 102 days for cases with >15% p53-positive cells. Among acute myeloid leukemia with myelodysplasia-related changes cases with a complex karyotype, the finding of >15% p53-positive cells was significantly associated with worse overall survival. The poor prognosis associated with more than 15% p53-positive cells was independent of age and karyotype. In acute myeloid leukemia with myelodysplasia-related changes, p53 expression may be useful to infer TP53 mutation status, complex karyotype, and/or poor prognosis in situations where other modalities are not readily available.
Collapse
|
74
|
Oka S, Ono K, Nohgawa M. Myelodysplastic syndrome with concomitant del(5q) and JAK2 V617F mutation transformed to acute myeloid leukemia with an additional chromosomal abnormality after a long-term treatment with lenalidomide. Leuk Lymphoma 2017; 58:2511-2513. [DOI: 10.1080/10428194.2017.1295146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Satoko Oka
- Division of Hematology, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Kazuo Ono
- Division of Pathology, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Masaharu Nohgawa
- Division of Hematology, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| |
Collapse
|
75
|
Duarte FB, Santos TEDJD, Barbosa MC, Kaufman J, Vasconcelos JPD, Lemes RPG, Rocha FD, Coutinho DF, Zalcberg I, Vasconcelos PRLD. Relevance of prognostic factors in the decision-making of stem cell transplantation in Myelodysplastic Syndromes. Rev Assoc Med Bras (1992) 2016; 62 Suppl 1:25-28. [PMID: 27982317 DOI: 10.1590/1806-9282.62.suppl1.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The hematopoietic stem cell transplantation (HSCT) is the only curative alternative for Myelodysplastic Syndrome (MDS), but many patients are not eligible for this treatment, as there are several limiting factors, especially in the case of patients with low-risk MDS. The aim of this study is to discuss the factors that can guide the decision-making on referring or not a patient to HSCT. Three cases of MDS, two of which were submitted to HSCT are presented. We intend to report the difficulties in referring patients with MDS to transplant and the prognostic factors that contribute to define eligibility.
Collapse
Affiliation(s)
- Fernando Barroso Duarte
- Bone Marrow Transplantation Service, Hospital Universitário Walter Cantídio, Centro de Hematologia e Hemoterapia do Ceará, Brazil
| | | | | | - Jacques Kaufman
- Bone Marrow Transplantation Service, Hospital Universitário Walter Cantídio, Brazil
| | | | | | | | | | | | | |
Collapse
|
76
|
Zhang L, McGraw KL, Sallman DA, List AF. The role of p53 in myelodysplastic syndromes and acute myeloid leukemia: molecular aspects and clinical implications. Leuk Lymphoma 2016; 58:1777-1790. [PMID: 27967292 DOI: 10.1080/10428194.2016.1266625] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
TP53 gene mutations occurring in patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are associated with high-risk karyotypes including 17p abnormalities, monosomal and complex cytogenetics. TP53 mutations in these disorders portend rapid disease progression and resistance to conventional therapeutics. Notably, the size of the TP53 mutant clone as measured by mutation allele burden is directly linked to overall survival (OS) confirming the importance of p53 as a negative prognostic variable. In nucleolar stress-induced ribosomopathies, such as del(5q) MDS, disassociation of MDM2 and p53 results in p53 accumulation in erythroid precursors manifested as erythroid hypoplasia. P53 antagonism by lenalidomide or other therapeutics such as antisense oligonucleotides, repopulates erythroid precursors and enhances effective erythropoiesis. These findings demonstrate that p53 is an intriguing therapeutic target that is currently under investigation in MDS and AML. This study reviews molecular advances in understanding the role of p53 in MDS and AML, and explores potential therapeutic strategies in this era of personalized medicine.
Collapse
Affiliation(s)
- Ling Zhang
- a Department of Hematopathology and Laboratory Medicine , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Kathy L McGraw
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - David A Sallman
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Alan F List
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| |
Collapse
|
77
|
Santini V. Treatment of low-risk myelodysplastic syndromes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:462-469. [PMID: 27913517 PMCID: PMC6142510 DOI: 10.1182/asheducation-2016.1.462] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The majority of myelodysplastic syndrome (MDS) patients belong to the International Prognostic Scoring System (IPSS) and IPSS-revised (IPSS-R) lower-risk categories. Their precise diagnostics and prognostic stratification is often a challenge, but may ensure the optimization of therapy. The availability of diverse treatment options has significantly improved the quality of life and survival of this group of patients. Anemia is the most relevant cytopenia in terms of frequency and symptoms in lower-risk MDS, and may be treated successfully with erythropoietic stimulating agents, provided a careful selection is performed on the basis of IPSS-R, endogenous erythropoietin levels, and transfusion independence. Doses and duration of therapy of erythropoietic-stimulating agents (ESAs) are critical to determine efficacy. In case a patient fails ESA treatment, the available options may include lenalidomide (approved for del5q positive cases), hypomethylating agents, and a rather large number of experimental agents, whose clinical trials should be offered to a larger number of MDS patients. The choice for second-line treatment must take into account biologic, cytogenetic, and molecular-identified characteristics of individual patients, as well as frailty and comorbidities. Other cytopenias are less frequently presenting as isolated. Specific therapy for thrombocytopenia has been proposed in experimental clinical trials with thrombomimetic agents that have shown good efficacy, but raised some safety concern. Although neutropenia is targeted symptomatically with growth factor supportive care, the immunosuppressive treatments are indicated mainly for pancytopenic, hypoplastic lower-risk MDS; they are not widely used because of their toxicity, despite the fact that they may induce responses. Finally, hematopoietic stem cell transplant is the curative option also for lower-risk MDS and timing should be carefully evaluated, balancing toxicity and the possibility of survival advantage. Finally, even when considered suitable for lower-risk MDS, transplant application is limited to the rarer fit and younger MDS patient.
Collapse
Affiliation(s)
- Valeria Santini
- SODc Hematology, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| |
Collapse
|
78
|
Scharenberg C, Giai V, Pellagatti A, Saft L, Dimitriou M, Jansson M, Jädersten M, Grandien A, Douagi I, Neuberg DS, LeBlanc K, Boultwood J, Karimi M, Jacobsen SEW, Woll PS, Hellström-Lindberg E. Progression in patients with low- and intermediate-1-risk del(5q) myelodysplastic syndromes is predicted by a limited subset of mutations. Haematologica 2016; 102:498-508. [PMID: 27884971 PMCID: PMC5394951 DOI: 10.3324/haematol.2016.152025] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/15/2016] [Indexed: 12/24/2022] Open
Abstract
A high proportion of patients with lower-risk del(5q) myelodysplastic syndromes will respond to treatment with lenalidomide. The median duration of transfusion-independence is 2 years with some long-lasting responses, but almost 40% of patients progress to acute leukemia by 5 years after starting treatment. The mechanisms underlying disease progression other than the well-established finding of small TP53-mutated subclones at diagnosis remain unclear. We studied a longitudinal cohort of 35 low- and intermediate-1-risk del(5q) patients treated with lenalidomide (n=22) or not (n=13) by flow cytometric surveillance of hematopoietic stem and progenitor cell subsets, targeted sequencing of mutational patterns, and changes in the bone marrow microenvironment. All 13 patients with disease progression were identified by a limited number of mutations in TP53, RUNX1, and TET2, respectively, with PTPN11 and SF3B1 occurring in one patient each. TP53 mutations were found in seven of nine patients who developed acute leukemia, and were documented to be present in the earliest sample (n=1) and acquired during lenalidomide treatment (n=6). By contrast, analysis of the microenvironment, and of hematopoietic stem and progenitor cells by flow cytometry was of limited prognostic value. Based on our data, we advocate conducting a prospective study aimed at investigating, in a larger number of cases of del(5q) myelodysplastic syndromes, whether the detection of such mutations before and after lenalidomide treatment can guide clinical decision-making.
Collapse
Affiliation(s)
- Christian Scharenberg
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Medicine, Division of Hematology, Skaraborgs Hospital, Skövde, Sweden
| | - Valentina Giai
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and NIHR Biomedical Research Centre, Oxford, UK
| | - Leonie Saft
- Department of Pathology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Marios Dimitriou
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Monika Jansson
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Martin Jädersten
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Alf Grandien
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Iyadh Douagi
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Donna S Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katarina LeBlanc
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and NIHR Biomedical Research Centre, Oxford, UK
| | - Mohsen Karimi
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Sten Eirik W Jacobsen
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Petter S Woll
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Eva Hellström-Lindberg
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
79
|
Canaani J, Nagler A. Established and emerging targeted therapies in the myelodysplastic syndromes. Expert Rev Hematol 2016; 9:997-1005. [PMID: 27615383 DOI: 10.1080/17474086.2016.1233054] [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: 10/21/2022]
Abstract
INTRODUCTION Therapy for the myelodysplastic syndromes (MDS) is an evolving area of research which has made significant use of the increased understanding of the complex biology of these disorders. Novel agents targeting multiple pathogenic pathways are being actively tested in preclinical and clinical settings and hold the potential to be available to clinicians before long. AREAS COVERED Herein we provide an historical framework for understanding the current use of hypomethylating agents in MDS and discuss recent developments in the field of targeted therapy in MDS including data from published and ongoing clinical studies with oral hypomethylating agents, PI3/polo-like kinase inhibitors, TGF-β inhibitor/ligand traps, and immune checkpoint inhibitors. A comprehensive review of recent literature was undertaken using PubMed and Medline. Expert commentary: Management of MDS patients will evolve substantially in the near future with the incorporation of molecular data into patient stratification models and with the introduction of novel targeted agents.
Collapse
Affiliation(s)
- Jonathan Canaani
- a Hematology Division , Chaim Sheba Medical Center , Tel Hashomer , Israel
| | - Arnon Nagler
- a Hematology Division , Chaim Sheba Medical Center , Tel Hashomer , Israel
| |
Collapse
|
80
|
McGraw KL, Cluzeau T, Sallman DA, Basiorka AA, Irvine BA, Zhang L, Epling-Burnette PK, Rollison DE, Mallo M, Sokol L, Solé F, Maciejewski J, List AF. TP53 and MDM2 single nucleotide polymorphisms influence survival in non-del(5q) myelodysplastic syndromes. Oncotarget 2016; 6:34437-45. [PMID: 26416416 PMCID: PMC4741464 DOI: 10.18632/oncotarget.5255] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/14/2015] [Indexed: 11/25/2022] Open
Abstract
P53 is a key regulator of many cellular processes and is negatively regulated by the human homolog of murine double minute-2 (MDM2) E3 ubiquitin ligase. Single nucleotide polymorphisms (SNPs) of either gene alone, and in combination, are linked to cancer susceptibility, disease progression, and therapy response. We analyzed the interaction of TP53 R72P and MDM2 SNP309 SNPs in relationship to outcome in patients with myelodysplastic syndromes (MDS). Sanger sequencing was performed on DNA isolated from 208 MDS cases. Utilizing a novel functional SNP scoring system ranging from +2 to −2 based on predicted p53 activity, we found statistically significant differences in overall survival (OS) (p = 0.02) and progression-free survival (PFS) (p = 0.02) in non-del(5q) MDS patients with low functional scores. In univariate analysis, only IPSS and the functional SNP score predicted OS and PFS in non-del(5q) patients. In multivariate analysis, the functional SNP score was independent of IPSS for OS and PFS. These data underscore the importance of TP53 R72P and MDM2 SNP309 SNPs in MDS, and provide a novel scoring system independent of IPSS that is predictive for disease outcome.
Collapse
Affiliation(s)
- Kathy L McGraw
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Thomas Cluzeau
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Hematology Department, CHU of Nice, Nice, France.,University Nice Sophia Antipolis, Faculty of Medicine, Nice, France.,Mediterranean Center of Molecular Medicine, INSERM U1065, Nice, France.,French Group of Myelodysplasia, France
| | - David A Sallman
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ashley A Basiorka
- Moffitt Cancer Center and Research Institute and The Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
| | - Brittany A Irvine
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ling Zhang
- Department of Pathology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - P K Epling-Burnette
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Dana E Rollison
- Department of Cancer Epidemiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mar Mallo
- Institut de Recerca Contra la Leucèmia Josep Carreras (IJC) Badalona, Barcelona, Spain
| | - Lubomir Sokol
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Francesc Solé
- Institut de Recerca Contra la Leucèmia Josep Carreras (IJC) Badalona, Barcelona, Spain
| | | | - Alan F List
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
81
|
van der Helm LH, Berger G, Diepstra A, Huls G, Vellenga E. Overexpression of TP53 is associated with poor survival, but not with reduced response to hypomethylating agents in older patients with acute myeloid leukaemia. Br J Haematol 2016; 178:810-812. [DOI: 10.1111/bjh.14166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 04/01/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Lieke H. van der Helm
- Department of Haematology; University Medical Centre Groningen; University of Groningen; Groningen The Netherlands
| | - Gerbrig Berger
- Department of Haematology; University Medical Centre Groningen; University of Groningen; Groningen The Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology; University Medical Centre Groningen; University of Groningen; Groningen The Netherlands
| | - Gerwin Huls
- Department of Haematology; University Medical Centre Groningen; University of Groningen; Groningen The Netherlands
| | - Edo Vellenga
- Department of Haematology; University Medical Centre Groningen; University of Groningen; Groningen The Netherlands
| |
Collapse
|
82
|
Fang J, Liu X, Bolanos L, Barker B, Rigolino C, Cortelezzi A, Oliva EN, Cuzzola M, Grimes HL, Fontanillo C, Komurov K, MacBeth K, Starczynowski DT. A calcium- and calpain-dependent pathway determines the response to lenalidomide in myelodysplastic syndromes. Nat Med 2016; 22:727-34. [PMID: 27294874 PMCID: PMC5507589 DOI: 10.1038/nm.4127] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/13/2016] [Indexed: 12/15/2022]
Abstract
Despite the high response rates of individuals with myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)) to treatment with lenalidomide (LEN) and the recent identification of cereblon (CRBN) as the molecular target of LEN, the cellular mechanism by which LEN eliminates MDS clones remains elusive. Here we performed an RNA interference screen to delineate gene regulatory networks that mediate LEN responsiveness in an MDS cell line, MDSL. We identified GPR68, which encodes a G-protein-coupled receptor that has been implicated in calcium metabolism, as the top candidate gene for modulating sensitivity to LEN. LEN induced GPR68 expression via IKAROS family zinc finger 1 (IKZF1), resulting in increased cytosolic calcium levels and activation of a calcium-dependent calpain, CAPN1, which were requisite steps for induction of apoptosis in MDS cells and in acute myeloid leukemia (AML) cells. In contrast, deletion of GPR68 or inhibition of calcium and calpain activation suppressed LEN-induced cytotoxicity. Moreover, expression of calpastatin (CAST), an endogenous CAPN1 inhibitor that is encoded by a gene (CAST) deleted in del(5q) MDS, correlated with LEN responsiveness in patients with del(5q) MDS. Depletion of CAST restored responsiveness of LEN-resistant non-del(5q) MDS cells and AML cells, providing an explanation for the superior responses of patients with del(5q) MDS to LEN treatment. Our study describes a cellular mechanism by which LEN, acting through CRBN and IKZF1, has cytotoxic effects in MDS and AML that depend on a calcium- and calpain-dependent pathway.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Apoptosis/drug effects
- Apoptosis/genetics
- Calcium/metabolism
- Calcium-Binding Proteins/genetics
- Calpain/drug effects
- Calpain/genetics
- Calpain/metabolism
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks
- Humans
- Ikaros Transcription Factor/drug effects
- Ikaros Transcription Factor/genetics
- Ikaros Transcription Factor/metabolism
- Immunologic Factors/pharmacology
- Lenalidomide
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/metabolism
- Peptide Hydrolases/metabolism
- RNA Interference
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Thalidomide/analogs & derivatives
- Thalidomide/pharmacology
- Ubiquitin-Protein Ligases
Collapse
Affiliation(s)
- Jing Fang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Xiaona Liu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Lyndsey Bolanos
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Brenden Barker
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Carmela Rigolino
- Bone Marrow Transplant Unit, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Agostino Cortelezzi
- Department of Hematology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Esther N Oliva
- Hematology Unit, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Maria Cuzzola
- Bone Marrow Transplant Unit, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - H Leighton Grimes
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | | | - Kakajan Komurov
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Kyle MacBeth
- Celgene Corporation, San Francisco, California, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
83
|
McGraw KL, Nguyen J, Komrokji RS, Sallman D, Al Ali NH, Padron E, Lancet JE, Moscinski LC, List AF, Zhang L. Immunohistochemical pattern of p53 is a measure of TP53 mutation burden and adverse clinical outcome in myelodysplastic syndromes and secondary acute myeloid leukemia. Haematologica 2016; 101:e320-3. [PMID: 27081179 DOI: 10.3324/haematol.2016.143214] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Kathy L McGraw
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Johnny Nguyen
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Rami S Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - David Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Najla H Al Ali
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jeffrey E Lancet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Lynn C Moscinski
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Alan F List
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Ling Zhang
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
84
|
The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016; 127:2391-405. [PMID: 27069254 DOI: 10.1182/blood-2016-03-643544] [Citation(s) in RCA: 6649] [Impact Index Per Article: 738.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/06/2016] [Indexed: 02/06/2023] Open
Abstract
The World Health Organization (WHO) classification of tumors of the hematopoietic and lymphoid tissues was last updated in 2008. Since then, there have been numerous advances in the identification of unique biomarkers associated with some myeloid neoplasms and acute leukemias, largely derived from gene expression analysis and next-generation sequencing that can significantly improve the diagnostic criteria as well as the prognostic relevance of entities currently included in the WHO classification and that also suggest new entities that should be added. Therefore, there is a clear need for a revision to the current classification. The revisions to the categories of myeloid neoplasms and acute leukemia will be published in a monograph in 2016 and reflect a consensus of opinion of hematopathologists, hematologists, oncologists, and geneticists. The 2016 edition represents a revision of the prior classification rather than an entirely new classification and attempts to incorporate new clinical, prognostic, morphologic, immunophenotypic, and genetic data that have emerged since the last edition. The major changes in the classification and their rationale are presented here.
Collapse
|
85
|
Jain P, Tang G, Konoplev SN, Kanagal-Shamanna R, Wang SA, Pemmaraju N, Daver N, Estrov Z. Synchronous del5q myelodysplastic syndrome (del5qMDS) and adult B-cell acute lymphoblastic leukemia (B-ALL) with TET2 and TP53 mutations. Am J Hematol 2016; 91:354-5. [PMID: 26437693 DOI: 10.1002/ajh.24208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Preetesh Jain
- M.D. Anderson Cancer Center; Department of Leukemia; the University of Texas; Houston Texas
| | - Guilin Tang
- M.D. Anderson Cancer Center; Department of Hematopathology; the University of Texas; Houston Texas
| | - Sergej N. Konoplev
- M.D. Anderson Cancer Center; Department of Hematopathology; the University of Texas; Houston Texas
| | - Rashmi Kanagal-Shamanna
- M.D. Anderson Cancer Center; Department of Hematopathology; the University of Texas; Houston Texas
| | - Sa A Wang
- M.D. Anderson Cancer Center; Department of Hematopathology; the University of Texas; Houston Texas
| | - Naveen Pemmaraju
- M.D. Anderson Cancer Center; Department of Leukemia; the University of Texas; Houston Texas
| | - Naval Daver
- M.D. Anderson Cancer Center; Department of Leukemia; the University of Texas; Houston Texas
| | - Zeev Estrov
- M.D. Anderson Cancer Center; Department of Leukemia; the University of Texas; Houston Texas
| |
Collapse
|
86
|
Christopeit M, Badbaran A, Alawi M, Zabelina T, Zeck G, Wolschke C, Ayuk F, Kröger N. Correlation of somatic mutations with outcome after FLAMSA-busulfan sequential conditioning and allogeneic stem cell transplantation in patients with myelodysplastic syndromes. Eur J Haematol 2016; 97:288-96. [PMID: 26680262 DOI: 10.1111/ejh.12724] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2015] [Indexed: 01/08/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative treatment option for myelodysplastic syndromes (MDS). Little is known about the prognostic impact of mutations, for example, in TP53 specifically after allo-HSCT. We here describe the prognostic impact of mutations in a panel of 19 genes analyzed by amplicon-based next-generation-sequencing in a uniformly treated patient cohort. Sixty-two patients with a median age of 61 yr suffered from MDS with 0-20% bone marrow blasts. International Prognostic Score was intermediate 1 (15%) and higher (79%). Conditioning uniformly was performed using a sequential approach in which FLAMSA chemotherapy was followed by Busulfan-based conditioning. Patients mostly were transplanted from an unrelated donor (77%), and 36% of patients received a graft from a mismatched donor. Median number of mutations was 2 (range 0-6). RUNX1, GATA2, TET2, and CEBPA were the genes most frequently found mutated. TP53, a factor previously reported to confer adverse prognostic impact after allogeneic stem cell transplantation, was mutated in samples from eight patients, one of which showed a silent mutation. With an estimated 5-yr overall/disease-free survival of 48 ± 7%/41 ± 7%, none of the mutations analyzed showed a prognostic impact in this analysis of the largest uniformly treated cohort thus far. This especially holds true for patients with a mutation in TP53.
Collapse
Affiliation(s)
- Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Badbaran
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Service Facility, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatjana Zabelina
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gaby Zeck
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Wolschke
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
87
|
Goudarzi KM, Lindström MS. Role of ribosomal protein mutations in tumor development (Review). Int J Oncol 2016; 48:1313-24. [PMID: 26892688 PMCID: PMC4777597 DOI: 10.3892/ijo.2016.3387] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/07/2016] [Indexed: 12/16/2022] Open
Abstract
Ribosomes are cellular machines essential for protein synthesis. The biogenesis of ribosomes is a highly complex and energy consuming process that initiates in the nucleolus. Recently, a series of studies applying whole-exome or whole-genome sequencing techniques have led to the discovery of ribosomal protein gene mutations in different cancer types. Mutations in ribosomal protein genes have for example been found in endometrial cancer (RPL22), T-cell acute lymphoblastic leukemia (RPL10, RPL5 and RPL11), chronic lymphocytic leukemia (RPS15), colorectal cancer (RPS20), and glioma (RPL5). Moreover, patients suffering from Diamond-Blackfan anemia, a bone marrow failure syndrome caused by mutant ribosomal proteins are also at higher risk for developing leukemia, or solid tumors. Different experimental models indicate potential mechanisms whereby ribosomal proteins may initiate cancer development. In particular, deregulation of the p53 tumor suppressor network and altered mRNA translation are mechanisms likely to be involved. We envisage that changes in expression and the occurrence of ribosomal protein gene mutations play important roles in cancer development. Ribosome biology constitutes a re-emerging vital area of basic and translational cancer research.
Collapse
Affiliation(s)
- Kaveh M Goudarzi
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska, CCK R8:05, Karolinska University Hospital in Solna, Stockholm, Sweden
| | - Mikael S Lindström
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
88
|
Nishiwaki S, Ito M, Watarai R, Okuno S, Harada Y, Yamamoto S, Suzuki K, Kurahashi S, Iwasaki T, Sugiura I. A new prognostic index to make short-term prognoses in MDS patients treated with azacitidine: A combination of p53 expression and cytogenetics. Leuk Res 2016; 41:21-6. [DOI: 10.1016/j.leukres.2015.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 11/11/2015] [Accepted: 11/22/2015] [Indexed: 01/13/2023]
|
89
|
Novel therapeutic strategies in myelodysplastic syndromes: do molecular genetics help? Curr Opin Hematol 2016; 23:79-87. [PMID: 26825694 DOI: 10.1097/moh.0000000000000211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Many studies over the past decade have together identified genes that are recurrently mutated in the myelodysplastic syndromes (MDS). We will summarize how this information has informed our understanding of disease pathogenesis and behavior, with an emphasis on how this information may inform therapeutic strategies. RECENT FINDINGS Genomic sequencing techniques have allowed for the identification of many recurrently mutated genes in MDS, with the most common mutations being found in epigenetic modifiers and components of the splicing machinery. Although many mutations are associated with clinical outcomes and disease phenotypes, at the current time they add relatively little to already robust clinical prognostic algorithms. However, as molecular genetic data are accumulated in larger numbers of patients, it is likely that the clinical significance of co-occurring mutations and less common mutations will come to light. Finally, mutated genes may identify biologically distinct subgroups of MDS that may benefit from novel therapies, and a subset of these genes may themselves serve as therapeutic targets. SUMMARY Advances in our knowledge of the molecular genetics of MDS have significantly improved our understanding of disease biology and promise to improve tools for clinical decision-making and identify new therapies for patients.
Collapse
|
90
|
Myelodysplastic syndrome with del (5q) and JAK2(V617F) mutation transformed to acute myeloid leukaemia with complex karyotype. Ann Hematol 2016; 95:525-7. [PMID: 26750119 DOI: 10.1007/s00277-015-2584-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/23/2015] [Indexed: 10/22/2022]
|
91
|
Mattsson K, Honkaniemi E, Barbany G, Gustafsson B. Increased p53 protein expression as a potential predictor of early relapse after hematopoietic stem cell transplantation in children with acute myelogenous leukemia. Pediatr Transplant 2015; 19:767-75. [PMID: 26432780 DOI: 10.1111/petr.12585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2015] [Indexed: 01/17/2023]
Abstract
Dysregulation of genes involved in the cell cycle such as TP53, P21, P16, and PTEN plays a key role in oncogenesis. We have earlier reported increased expression of the TP53 encoded protein p53, in bone marrow samples from pediatric patients with more aggressive, rare chronic myeloid malignancies. The aim of this study was to investigate protein expression of p53, p21, p16, and PTEN before and after HSCT in pediatric patients with AML and evaluate whether any potential alterations could predict relapse after HSCT. Paraffin-embedded bone marrow samples from 34 pediatric patients with AML were collected retrospectively from time of diagnosis as well as pre- and post-HSCT. IHC was performed on tissue microarrays with antibodies against p53, p21, p16, and PTEN. Study material was analyzed by independent t-tests and nonlinear regression. t-Tests showed a statistical significant difference in p53 (p = 0.010) with overexpression in the group of patients who relapsed compared to the relapse-free patients at >3-6 months post-HSCT. Analysis of p53 protein expression by IHC may be a potential predictor for relapse after HSCT in children with AML.
Collapse
Affiliation(s)
- Kristin Mattsson
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Emma Honkaniemi
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatrics, Södertälje Hospital, Stockholm, Sweden
| | - Gisela Barbany
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Britt Gustafsson
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
92
|
Komrokji RS, List AF. Short- and long-term benefits of lenalidomide treatment in patients with lower-risk del(5q) myelodysplastic syndromes. Ann Oncol 2015; 27:62-8. [PMID: 26504152 PMCID: PMC4684154 DOI: 10.1093/annonc/mdv488] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/28/2015] [Indexed: 11/17/2022] Open
Abstract
In patients with transfusion-dependent, lower-risk, del(5q) myelodysplastic syndromes, lenalidomide effectively reduces red blood cell transfusion burden and acts as a disease-modifying agent, altering the natural history of the disease by suppressing the del(5q) clone. Hematologic and/or cytogenetic responses correlate with delayed acute myeloid leukemia progression and better overall survival. The treatment of patients with myelodysplastic syndromes (MDS) begins with assessment of karyotype and risk. Lenalidomide is approved for the treatment of patients who have transfusion-dependent anemia due to lower-risk MDS with chromosome 5q deletion (del(5q)) with or without additional cytogenetic abnormalities, and isolated del(5q) only in the European Union. Mounting evidence suggests that lenalidomide is effective not only in reducing red blood cell (RBC) transfusion burden, but also in modifying the disease natural history by suppressing the malignant clone. Data discussed here from the pivotal phase 2 (MDS-003) and phase 3 (MDS-004) studies of lenalidomide demonstrate that lenalidomide treatment was associated with both short- and long-term benefits. These clinical benefits included high rates of RBC-transfusion independence (TI) with prolonged durations of response, high rates of cytogenetic response (CyR) associated with achievement of durable RBC-TI, no significant difference in rates of progression to acute myeloid leukemia (AML), and improvements in health-related quality of life (HRQOL). Achievement of RBC-TI and CyR with lenalidomide treatment was associated with extended survival and time to AML progression. Achievement of RBC-TI and hemoglobin response was additionally associated with HRQOL benefits. Recent data describing the impact of TP53 mutations and p53 expression on the prognosis of patients with del(5q) and the impact on response to lenalidomide are also discussed. The authors provide practical recommendations for the use of lenalidomide in patients with lower-risk del(5q) MDS.
Collapse
Affiliation(s)
- R S Komrokji
- Malignant Hematology Department, Moffitt Cancer Center, Tampa, USA
| | - A F List
- Malignant Hematology Department, Moffitt Cancer Center, Tampa, USA
| |
Collapse
|
93
|
Loghavi S, Al-Ibraheemi A, Zuo Z, Garcia-Manero G, Yabe M, Wang SA, Kantarjian HM, Yin CC, Miranda RN, Luthra R, Medeiros LJ, Bueso-Ramos CE, Khoury JD. TP53 overexpression is an independent adverse prognostic factor in de novo myelodysplastic syndromes with fibrosis. Br J Haematol 2015; 171:91-9. [PMID: 26123119 DOI: 10.1111/bjh.13529] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/10/2015] [Indexed: 12/20/2022]
Abstract
Bone marrow (BM) fibrosis is associated with poor prognosis in patients with de novo myelodysplastic syndromes (MDS). TP53 mutations and TP53 (p53) overexpression in MDS are also associated with poor patient outcomes. The prevalence and significance of TP53 mutations and TP53 overexpression in MDS with fibrosis are unknown. We studied 67 patients with de novo MDS demonstrating moderate to severe reticulin fibrosis (MDS-F). Expression of TP53 was evaluated in BM core biopsy specimens using dual-colour CD34/TP53 immunohistochemistry with computer-assisted image analysis. Mutation analysis was performed using next-generation sequencing, or Sanger sequencing methods. TP53 mutations were present in 47·1% of cases. TP53 mutation was significantly associated with TP53 expression (P = 0·0294). High levels of TP53 expression (3 + in ≥10% cells) were associated with higher BM blast counts (P = 0·0149); alterations of chromosomes 5 (P = 0·0009) or 7 (P = 0·0141); complex karyotype (P = 0·0002); high- and very-high risk IPSS-R groups (P = 0·009); and TP53 mutations (P = 0·0003). High TP53 expression independently predicted shorter overall survival (OS) by multivariate analysis (P = <0·001). Expression of TP53 by CD34-positive cells was associated with shorter OS and leukaemia-free survival (P = 0·0428). TP53 overexpression is a predictor of poor outcome in patients with MDS-F.
Collapse
Affiliation(s)
- Sanam Loghavi
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Alyaa Al-Ibraheemi
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Zhuang Zuo
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Mariko Yabe
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Sa A Wang
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Cameron C Yin
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Raja Luthra
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos E Bueso-Ramos
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph D Khoury
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
94
|
Loiseau C, Ali A, Itzykson R. New therapeutic approaches in myelodysplastic syndromes: Hypomethylating agents and lenalidomide. Exp Hematol 2015; 43:661-72. [PMID: 26123365 DOI: 10.1016/j.exphem.2015.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 01/17/2023]
Abstract
Recent advances in the treatment of myelodysplastic syndromes have come from the use of the hypomethylating agents decitabine and azacitidine as well as the immunomodulatory drug lenalidomide. Their clinical benefit has been demonstrated by randomized phase III clinical trials, mostly in high-risk and del(5q) myelodysplastic syndromes, respectively. Neither drug, however, appears to eradicate myelodysplastic stem cells, and thus they currently do not represent curative options. Here, we review data from both clinical and translational research on those drugs to identify their molecular and cellular mechanisms of action and to delineate paths for improved treatment allocation and further therapeutic advances in myelodysplastic syndromes.
Collapse
Affiliation(s)
- Clémence Loiseau
- Department of Hematology, Saint-Louis Hospital, Assistance Publique, Hopitaux de Paris, Paris Diderot University, Paris, France
| | - Ashfaq Ali
- Institut National de la Santé et de la Recherche Médicale, Saint-Louis Institute, Paris, France
| | - Raphael Itzykson
- Department of Hematology, Saint-Louis Hospital, Assistance Publique, Hopitaux de Paris, Paris Diderot University, Paris, France; Institut National de la Santé et de la Recherche Médicale, Saint-Louis Institute, Paris, France.
| |
Collapse
|
95
|
Zhang X, Lancet JE, Zhang L. Molecular pathology of myelodysplastic syndromes: new developments and implications for diagnosis and treatment. Leuk Lymphoma 2015; 56:3022-30. [DOI: 10.3109/10428194.2015.1037756] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
96
|
Recent Advances in the 5q- Syndrome. Mediterr J Hematol Infect Dis 2015; 7:e2015037. [PMID: 26075044 PMCID: PMC4450650 DOI: 10.4084/mjhid.2015.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/28/2015] [Indexed: 12/12/2022] Open
Abstract
The 5q- syndrome is the most distinct of the myelodysplastic syndromes (MDS) and patients with this disorder have a deletion of chromosome 5q [del(5q)] as the sole karyotypic abnormality. Several genes mapping to the commonly deleted region of the 5q- syndrome have been implicated in disease pathogenesis in recent years. Haploinsufficiency of the ribosomal gene RPS14 has been shown to cause the erythroid defect in the 5q- syndrome. Loss of the microRNA genes miR-145 and miR-146a has been associated with the thrombocytosis observed in 5q- syndrome patients. Haploinsufficiency of CSNK1A1 leads to hematopoietic stem cell expansion in mice and may play a role in the initial clonal expansion in patients with 5q- syndrome. Moreover, a subset of patients harbor mutation of the remaining CSNK1A1 allele. Mouse models of the 5q- syndrome, which recapitulate the key features of the human disease, indicate that a p53-dependent mechanism underlies the pathophysiology of this disorder. Importantly, activation of p53 has been demonstrated in the human 5q- syndrome. Recurrent TP53 mutations have been associated with an increased risk of disease evolution and with decreased response to the drug lenalidomide in del(5q) MDS patients. Potential new therapeutic agents for del(5q) MDS include the translation enhancer L-leucine.
Collapse
|
97
|
Smith AE, Kulasekararaj AG, Jiang J, Mian S, Mohamedali A, Gaken J, Ireland R, Czepulkowski B, Best S, Mufti GJ. CSNK1A1 mutations and isolated del(5q) abnormality in myelodysplastic syndrome: a retrospective mutational analysis. LANCET HAEMATOLOGY 2015; 2:e212-21. [PMID: 26688096 DOI: 10.1016/s2352-3026(15)00050-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/20/2015] [Accepted: 03/20/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND A mechanism for clonal growth advantage in isolated del(5q) disease remains elusive. CSNK1A1 resides on the critically deleted region, and deletion of this gene has been shown in mouse knockout and transplantation studies to produce some characteristics of bone marrow failure, including a proliferative advantage. We aimed to establish the frequency, nature, and clinical association of CSNK1A1 mutations in patients with myelodysplastic syndrome and associated myeloid neoplasms. METHODS Between June 1, 2004, and May 31, 2014, in King's College (London, UK), we did whole-exome sequencing of five patients with isolated del(5q) followed by targeted screening for CSNK1A1 mutations and 20 myelodysplastic syndrome-associated mutations in 245 additional patients with myeloid neoplasms. All patients met present WHO diagnostic criteria for myelodysplastic syndrome and other related myeloid neoplasms. FINDINGS 39 (16%) of 250 patients with myeloid neoplasms had isolated del(5q), of whom seven (18%) had CSNK1A1 mutations. All these mutations were missense and presented in a highly conserved region that is implicated in ATP catalysis. Serial sampling and response to lenalidomide treatment showed that CSNK1A1 mutations were highly associated with the del(5q) clone. Only one patient with a CSNK1A1 mutation showed complete cytogenetic response to lenalidomide. Four (57%) of the seven patients carrying a CSNK1A1 mutation showed disease progression coupled with an increase in mutant allele burden (all four were on lenalidomide). We detected coexisting myelodysplastic syndrome-related gene mutations in patients with CSNK1A1 mutations, including TP53. INTERPRETATION Similar to the effect of TP53 mutations on progression of del(5q) abnormality, mutant CSNK1A1 also gives rise to a poor prognosis in del(5q) abnormality, for which a coupled increase in P53 activation is suggested. CSNK1A1 mutations in del(5q) disease are important in the context of therapeutic manipulation and need incorporation into future prospective studies. FUNDING Leukaemia and Lymphoma Research.
Collapse
Affiliation(s)
- Alexander E Smith
- Department of Haematological Medicine, King's College London School of Medicine, Rayne Institute, King's College London, London, UK; Department of Haematology, King's College Hospital, King's College London, London, UK
| | - Austin G Kulasekararaj
- Department of Haematological Medicine, King's College London School of Medicine, Rayne Institute, King's College London, London, UK; Department of Haematology, King's College Hospital, King's College London, London, UK
| | - Jie Jiang
- Department of Haematological Medicine, King's College London School of Medicine, Rayne Institute, King's College London, London, UK; Department of Haematology, King's College Hospital, King's College London, London, UK
| | - Syed Mian
- Department of Haematological Medicine, King's College London School of Medicine, Rayne Institute, King's College London, London, UK
| | - Azim Mohamedali
- Department of Haematological Medicine, King's College London School of Medicine, Rayne Institute, King's College London, London, UK; Department of Haematology, King's College Hospital, King's College London, London, UK
| | - Joop Gaken
- Department of Haematological Medicine, King's College London School of Medicine, Rayne Institute, King's College London, London, UK
| | - Robin Ireland
- Department of Haematology, King's College Hospital, King's College London, London, UK
| | - Barbara Czepulkowski
- Department of Haematology, King's College Hospital, King's College London, London, UK
| | - Steven Best
- Department of Haematology, King's College Hospital, King's College London, London, UK
| | - Ghulam J Mufti
- Department of Haematological Medicine, King's College London School of Medicine, Rayne Institute, King's College London, London, UK; Department of Haematology, King's College Hospital, King's College London, London, UK.
| |
Collapse
|
98
|
High p53 protein expression in therapy-related myeloid neoplasms is associated with adverse karyotype and poor outcome. Mod Pathol 2015; 28:552-63. [PMID: 25412846 DOI: 10.1038/modpathol.2014.153] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/26/2014] [Accepted: 09/16/2014] [Indexed: 11/09/2022]
Abstract
Identification of p53-positive cells by immunohistochemistry in bone marrow from primary myelodysplastic syndrome patients correlates with the presence of TP53 mutations and poor prognosis. Mutations in the tumor suppressor gene TP53 are more frequent in therapy-related acute myeloid leukemia and myelodysplastic syndrome than in de novo disease, but the role of p53 immunohistochemistry in the therapy-related setting has not been specifically investigated. We studied p53 protein immunoreactivity in bone marrow biopsies of therapy-related myeloid neoplasms and correlated protein expression with TP53 mutation status, clinicopathologic features and outcome. We first studied 32 patients with therapy-related acute myeloid leukemia and 63 patients with therapy-related myelodysplastic syndrome/chronic myelomonocytic leukemia from one institution and then validated our results in a separate group of 32 patients with therapy-related acute myeloid leukemia and 56 patients with therapy-related myelodysplastic syndrome from a different institution. Strong p53 immunostaining in ≥1% of bone marrow cells was highly predictive of a TP53 gene mutation (P<0.0001) and was strongly associated with a high-risk karyotype (P<0.0001). The presence of ≥1% p53 strongly positive cells was associated with poorer overall and disease-specific survival, particularly in the subset of patients treated with stem-cell transplantation. In a multivariable Cox regression model, the presence of ≥1% p53 strongly expressing cells was an independent prognostic marker for overall survival in both cohorts, with hazard ratios of 3.434 (CI: 1.751-6.735, P<0.0001) and 3.156 (CI: 1.502-6.628, P=0.002). Our data indicate that p53 protein expression, evaluated in bone marrow biopsies by a widely available immunohistochemical method, prognostically stratifies patients with therapy-related myeloid neoplasms independent of other risk factors. p53 immunostaining thus represents an easily applicable method to assess risk in therapy-related acute myeloid leukemia/myelodysplastic syndrome patients.
Collapse
|
99
|
Gustafsson BM. Different aspects of stem cell procedures in children with poor responding AML: when is HSCT the best answer? Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Acute myeloid leukemia in children is a heterogeneous disease with different morphological and cytogenetic features. New diagnostic tools and treatments, improved supportive care and the use of genomic tissue typing in selecting donors for hematopoietic stem cell transplantation (HSCT) adds to increased survival rates. Candidates to HSCT in first complete remission are patients with cytogenetic or molecular unfavorable prognostic markers, or blasts >15% after first induction. The use of minimal residual disease can also identify children benefiting from HSCT in first complete remission and the patients post HSCT with signs of relapse. The outcome and cure rate of acute myeloid leukemia, still remains poor and new diagnostic tools and treatments strategies need to be evaluated. In this management perspective, future management of novel minimal residual disease tools are discussed, conditioning therapies, as well as different transplantation procedures including haplo-transplantation and haplo-identical natural killer cell transplantation, but also altered graft-versus-host-disease treatments.
Collapse
Affiliation(s)
- Britt M Gustafsson
- Department of Clinical Science, Intervention & Technology, CLINTEC, Karolinska Institutet, SE141 86 Stockholm, Sweden
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
100
|
Cerqui E, Pelizzari A, Schieppati F, Borlenghi E, Pagani C, Bellotti D, Lamorgese C, Boiocchi L, Sottini A, Imberti L, Rossi G. Lenalidomide in patients with red blood cell transfusion-dependent myelodysplastic syndrome and del(5q): a single-centre "real-world" experience. Leuk Lymphoma 2015; 56:3129-34. [PMID: 25811676 DOI: 10.3109/10428194.2015.1034703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
"Real life" data are needed to complement published trials on the efficacy of lenalidomide in patients with myelodysplastic syndrome (MDS) and del(5q) and on the risk of inducing acute myeloid leukemia (AML) progression. Here, we present results of lenalidomide treatment in a consecutive, population-based series of 21 red blood cell (RBC) transfusion-dependent elderly patients with multiple comorbidities. Of 18 evaluable patients (median follow-up: 22 months), 17 achieved an erythroid hematologic response (HI-E) and 16 an RBC transfusion independence. Cytogenetic response (CyR) rate was 80%, median overall survival was 48 months (range 3-164), and 5-year leukemia-free survival was 84%. Three patients progressed to AML; one, with baseline TP53 mutation, achieved HI-E, partial CyR, and did not progress to AML. Eighteen patients experienced hematological adverse events. Overall, lenalidomide was very effective and well tolerated even in unselected elderly patients with multiple comorbidities and did not appear to increase the risk of AML.
Collapse
Affiliation(s)
- Elisa Cerqui
- a Department of Hematology , A.O. Spedali Civili , Brescia , Italy
| | | | | | - Erika Borlenghi
- a Department of Hematology , A.O. Spedali Civili , Brescia , Italy
| | - Chiara Pagani
- a Department of Hematology , A.O. Spedali Civili , Brescia , Italy
| | - Daniela Bellotti
- b Cytogenetics and Genetics Laboratory, Department of Genetic and Molecular Medicine , University of Brescia , Brescia , Italy
| | - Cinzia Lamorgese
- a Department of Hematology , A.O. Spedali Civili , Brescia , Italy
| | - Leonardo Boiocchi
- c Pathology Section, Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| | - Alessandra Sottini
- d Centro Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, A.O. Spedali Civili , Brescia , Italy
| | - Luisa Imberti
- d Centro Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, A.O. Spedali Civili , Brescia , Italy
| | - Giuseppe Rossi
- a Department of Hematology , A.O. Spedali Civili , Brescia , Italy
| |
Collapse
|