551
|
West AH, Churpek JE. Old and new tools in the clinical diagnosis of inherited bone marrow failure syndromes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:79-87. [PMID: 29222240 PMCID: PMC6142587 DOI: 10.1182/asheducation-2017.1.79] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Patients with inherited bone marrow failure syndromes (IBMFSs) classically present with specific patterns of cytopenias along with congenital anomalies and/or other physical features that are often recognizable early in life. However, increasing application of genomic sequencing and clinical awareness of subtle disease presentations have led to the recognition of IBMFS in pediatric and adult populations more frequently than previously realized, such as those with early onset myelodysplastic syndrome (MDS). Given the well-defined differences in clinical management needs and outcomes for aplastic anemia, acute myeloid leukemia, and MDS in patients with an IBMFS vs those occurring sporadically, as well as nonhematologic comorbidities in patients with IBMFSs, it is critical for hematologists to understand how to approach screening for the currently known IBMFSs. This review presents a practical approach for the clinical hematologist that outlines when to suspect an IBMFS and how to use various diagnostic tools, from physical examination to screening laboratory tests and genomics, for the diagnosis of the most frequent IBMFSs: Fanconi anemia, telomere biology disorders, Diamond-Blackfan anemia, GATA2 deficiency syndrome, Shwachman-Diamond syndrome, and severe congenital neutropenia.
Collapse
Affiliation(s)
- Allison H. West
- Section of Hematology/Oncology, The University of Chicago Comprehensive Cancer Center, Chicago, IL; and
| | - Jane E. Churpek
- Section of Hematology/Oncology, The University of Chicago Comprehensive Cancer Center, Chicago, IL; and
- Center for Clinical Cancer Genetics, The University of Chicago, Chicago, IL
| |
Collapse
|
552
|
Lindsley RC. Uncoding the genetic heterogeneity of myelodysplastic syndrome. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:447-452. [PMID: 29222292 PMCID: PMC6142603 DOI: 10.1182/asheducation-2017.1.447] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Myelodysplastic syndrome (MDS) is a clinically heterogeneous disease characterized by functional impairment of hematopoiesis and abnormal bone marrow morphology. The type and severity of hematopoietic dysfunction in MDS are highly variable, and the kinetics of disease progression are difficult to predict. Genomic studies have shown that MDS is typically driven by a multistep somatic genetic process affecting a core set of genes. By definition, recurrent MDS driver mutations all drive clonal dominance, although they can have stereotyped positions in the clonal hierarchy or patterns of comutation association and exclusivity. Furthermore, environmental context, such as exposures to cytotoxic chemotherapy or the presence of germ-line predisposition, can influence disease pathogenesis and clinical outcomes. This review will address how an enhanced understanding of MDS genetics may enable refinement of current diagnostic schema, improve understanding of the pathogenesis of therapy-related MDS, and identify germ-line predispositions to development of MDS that are more common than recognized by standard clinical evaluation.
Collapse
Affiliation(s)
- R Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
553
|
Soiffer RJ, Chen YB. Pharmacologic agents to prevent and treat relapse after allogeneic hematopoietic cell transplantation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:699-707. [PMID: 29222324 PMCID: PMC6142540 DOI: 10.1182/asheducation-2017.1.699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Affiliation(s)
- Robert J. Soiffer
- Dana-Farber Cancer Institute, Boston, MA
- Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Yi-Bin Chen
- Harvard Medical School, Boston, MA; and
- Massachusetts General Hospital, Boston, MA
| |
Collapse
|
554
|
Singh N, Loren AW. Overview of Hematopoietic Cell Transplantation for the Treatment of Hematologic Malignancies. Clin Chest Med 2017; 38:575-593. [DOI: 10.1016/j.ccm.2017.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
555
|
Tefferi A, Lasho TL, Patnaik MM, Saeed L, Mudireddy M, Idossa D, Finke C, Ketterling RP, Pardanani A, Gangat N. Targeted next-generation sequencing in myelodysplastic syndromes and prognostic interaction between mutations and IPSS-R. Am J Hematol 2017; 92:1311-1317. [PMID: 28875545 DOI: 10.1002/ajh.24901] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 08/31/2017] [Indexed: 12/22/2022]
Abstract
A 27-gene panel was used for next-generation sequencing (NGS) in 179 patients (median age 73 years) with primary myelodysplastic syndromes (MDS); risk distribution according to the revised International Prognostic Scoring System (IPSS-R) was 11% very high, 18% high, 17% intermediate, 38% low and 16% very low. At least one mutation/variant was detected in 147 (82%) patients; 23% harbored three or more mutations/variants. The most frequent mutations/variants included ASXL1 (30%), TET2 (25%), SF3B1 (20%), U2AF1 (16%), SRSF2 (16%), TP53 (13%), RUNX1 (11%), and DNMT3A (10%). At a median follow up of 30 months, 148 (83%) deaths and 26 (15%) leukemic transformations were recorded. Multivariable analysis of mutations/variants identified ASXL1 (HR 1.7, 95% CI 1.2-2.5), SETBP1 (HR 4.1, 95% CI 1.6-10.2) and TP53 (HR 2.2, 95% CI 1.3-3.4) as risk factors for overall and SRSF2 (HR 3.9, 95% CI 1.5-10.2), IDH2 (HR 3.7, 95% CI 1.2-11.4), and CSF3R (HR 6.0, 95% CI 1.6-22.6) for leukemia-free survival. Addition of age to the multivariable model did not affect these results while accounting for IPSS-R weakened the significance of TP53 mutations/variants (P = .1). An apparently favorable survival impact of SF3B1 mutations was no longer evident after adjustment for IPSS-R. Approximately 41% and 20% of patients harbored at least one adverse mutation/variant for overall and leukemia-free survival, respectively. Number of mutations/variants did not provide additional prognostic value. The survival impact of adverse mutations was most evident in IPSS-R very low/low risk patients. These observations suggest that targeted NGS might assist in treatment decision-making in lower risk MDS.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Terra L. Lasho
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Mrinal M. Patnaik
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Lyla Saeed
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Mythri Mudireddy
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Dame Idossa
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Christy Finke
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Rhett P. Ketterling
- Division of Cytogenetics, Department of Laboratory Medicine; Mayo Clinic; Rochester Minnesota
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Naseema Gangat
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
556
|
Cooper JN, Young NS. Clonality in context: hematopoietic clones in their marrow environment. Blood 2017; 130:2363-2372. [PMID: 29046282 PMCID: PMC5709788 DOI: 10.1182/blood-2017-07-794362] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/04/2017] [Indexed: 11/20/2022] Open
Abstract
Clonal hematopoiesis occurs normally, especially with aging, and in the setting of disease, not only in myeloid cancers but in bone marrow failure as well. In cancer, malignant clones are characterized by recurrent somatic mutations in specific sets of genes, but the direct relationship of such mutations to leukemogenesis, when they occur in cells of an apparently healthy older individual or after recovery from immune aplastic anemia, is uncertain. Here we emphasize a view of clonal evolution that stresses natural selection over deterministic ontogeny, and we stress the selective role of the environment of the marrow and organism. Clonal hematopoieses after chemotherapy, in marrow failure, and with aging serve as models. We caution against the overinterpretation of clinical results of genomic testing in the absence of a better understanding of clonal selection and evolution.
Collapse
Affiliation(s)
- James N Cooper
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
557
|
Pharmacologic agents to prevent and treat relapse after allogeneic hematopoietic cell transplantation. Blood Adv 2017; 1:2473-2482. [PMID: 29296897 DOI: 10.1182/bloodadvances.2017009894] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/28/2017] [Indexed: 01/01/2023] Open
|
558
|
Alter BP. Inherited bone marrow failure syndromes: considerations pre- and posttransplant. Blood 2017; 130:2257-2264. [PMID: 29167174 PMCID: PMC5714231 DOI: 10.1182/blood-2017-05-781799] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/28/2017] [Indexed: 11/20/2022] Open
Abstract
Patients with inherited bone marrow failure syndromes are usually identified when they develop hematologic complications such as severe bone marrow failure, myelodysplastic syndrome, or acute myeloid leukemia. They often have specific birth defects or other physical abnormalities that suggest a syndrome, and sequencing of specific genes or next-generation sequencing can determine or confirm the particular syndrome. The 4 most frequent syndromes are Fanconi anemia, dyskeratosis congenita, Diamond Blackfan anemia, and Shwachman Diamond syndrome. This review discusses the major complications that develop as the patients with these syndromes age, as well as additional late effects following hematopoietic stem cell transplantation. The most common complications are iron overload in transfused patients and syndrome-specific malignancies in untransplanted patients, which may occur earlier and with higher risks in those who have received transplants.
Collapse
Affiliation(s)
- Blanche P Alter
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| |
Collapse
|
559
|
Abstract
Over the past decade, precision medicine (PM) approaches have received significant investment to create new therapies, learn more about disease processes, and potentially prevent diseases before they arise. However, in many ways, PM investments may come at the expense of existing public health measures that could have a greater impact on population health. As we tackle burgeoning public health concerns, such as obesity, and chronic diseases, such as cancer, it is not clear whether PM is aligned with public health or in conflict with its goals. We summarize the areas of promise demonstrated by PM, discuss the limitations of each of these areas from a population health perspective, and discuss how we can approach PM in a manner that is congruent with the core aims of public health.
Collapse
Affiliation(s)
- Ramya Ramaswami
- Imperial College NHS Healthcare Trust, Hammersmith Hospital, London W12 0HS, United Kingdom;
| | - Ronald Bayer
- Mailman School of Public Health, Columbia University, New York, NY 10032, USA;
| | - Sandro Galea
- Boston University School of Public Health, Boston, Massachusetts 02118, USA;
| |
Collapse
|
560
|
The genomic landscape of pediatric myelodysplastic syndromes. Nat Commun 2017; 8:1557. [PMID: 29146900 PMCID: PMC5691144 DOI: 10.1038/s41467-017-01590-5] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/29/2017] [Indexed: 01/19/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are uncommon in children and have a poor prognosis. In contrast to adult MDS, little is known about the genomic landscape of pediatric MDS. Here, we describe the somatic and germline changes of pediatric MDS using whole exome sequencing, targeted amplicon sequencing, and/or RNA-sequencing of 46 pediatric primary MDS patients. Our data show that, in contrast to adult MDS, Ras/MAPK pathway mutations are common in pediatric MDS (45% of primary cohort), while mutations in RNA splicing genes are rare (2% of primary cohort). Surprisingly, germline variants in SAMD9 or SAMD9L were present in 17% of primary MDS patients, and these variants were routinely lost in the tumor cells by chromosomal deletions (e.g., monosomy 7) or copy number neutral loss of heterozygosity (CN-LOH). Our data confirm that adult and pediatric MDS are separate diseases with disparate mechanisms, and that SAMD9/SAMD9L mutations represent a new class of MDS predisposition. Myelodysplastic syndromes (MDS) are uncommon in children and have poor prognosis. Here, the authors interrogate the genomic landscape of MDS, confirming adult and paediatric MDS are separate diseases with disparate mechanisms, and highlighting that SAMD9/SAMD9L mutations represent a new class of MDS predisposition.
Collapse
|
561
|
Chen S, Gao R, Yao C, Kobayashi M, Liu SZ, Yoder MC, Broxmeyer H, Kapur R, Boswell HS, Mayo LD, Liu Y. Genotoxic stresses promote clonal expansion of hematopoietic stem cells expressing mutant p53. Leukemia 2017; 32:850-854. [PMID: 29263439 DOI: 10.1038/leu.2017.325] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- S Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Gao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - C Yao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Rheumatism, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - M Kobayashi
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Z Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - M C Yoder
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H S Boswell
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - L D Mayo
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Y Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
562
|
Hypomethylating agents for treatment and prevention of relapse after allogeneic blood stem cell transplantation. Int J Hematol 2017; 107:138-150. [DOI: 10.1007/s12185-017-2364-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 11/08/2017] [Indexed: 12/14/2022]
|
563
|
Abstract
Age-related alterations in the human blood system occur in B cells, T cells, cells of the innate system, as well as hematopoietic stem and progenitor cells (HSPCs). Interestingly, age-related, reduced genetic diversity can be identified at the stem cell level and also independently in B cells and T cells. This reduced diversity is most probably related to somatic mutations or to changes in the microenvironmental niche. Either process can select for specific clones or cause repeated evolutionary bottlenecks. This review discusses the age-related clonal expansions in the human HSPC pool, which was termed in the past age-related clonal hematopoiesis (ARCH). ARCH is defined as the gradual, clonal expansion of HSPCs carrying specific, disruptive, and recurrent genetic variants, in individuals without clear diagnosis of hematological malignancies. ARCH is associated not just with chronological aging but also with several other, age-related pathological conditions, including inflammation, vascular diseases, cancer mortality, and high risk for hematological malignancies. Although it remains unclear whether ARCH is a marker of aging or plays an active role in these various pathophysiologies, it is suggested here that treating or even preventing ARCH may prove to be beneficial for human health. This review also describes a decision tree for the diagnosis and follow-up for ARCH in a research setting.
Collapse
|
564
|
Somatic mutations and clonal hematopoiesis in congenital neutropenia. Blood 2017; 131:408-416. [PMID: 29092827 DOI: 10.1182/blood-2017-08-801985] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/21/2017] [Indexed: 12/28/2022] Open
Abstract
Severe congenital neutropenia (SCN) and Shwachman-Diamond syndrome (SDS) are congenital neutropenia syndromes with a high rate of leukemic transformation. Hematopoietic stressors may contribute to leukemic transformation by increasing the mutation rate in hematopoietic stem/progenitor cells (HSPCs) and/or by promoting clonal hematopoiesis. We sequenced the exome of individual hematopoietic colonies derived from 13 patients with congenital neutropenia to measure total mutation burden and performed error-corrected sequencing on a panel of 46 genes on 80 patients with congenital neutropenia to assess for clonal hematopoiesis. An average of 3.6 ± 1.2 somatic mutations per exome was identified in HSPCs from patients with SCN compared with 3.9 ± 0.4 for healthy controls (P = NS). Clonal hematopoiesis due to mutations in TP53 was present in 48% (13/27) of patients with SDS but was not seen in healthy controls (0/17, P < .001) or patients with SCN (0/40, P < .001). Our SDS cohort was young (median age 6.3 years), and many of the patients had multiple TP53 mutations. Conversely, clonal hematopoiesis due to mutations of CSF3R was present in patients with SCN but was not detected in healthy controls or patients with SDS. These data show that hematopoietic stress, including granulocyte colony-stimulating factor, do not increase the mutation burden in HSPCs in congenital neutropenia. Rather, distinct hematopoietic stressors result in the selective expansion of HSPCs carrying specific gene mutations. In particular, in SDS there is enormous selective pressure to expand TP53-mutated HSPCs, suggesting that acquisition of TP53 mutations is an early, likely initiating event, in the transformation to myelodysplastic syndrome/acute myeloid leukemia in patients with SDS.
Collapse
|
565
|
Weber C, von Hundelshausen P. CANTOS Trial Validates the Inflammatory Pathogenesis of Atherosclerosis: Setting the Stage for a New Chapter in Therapeutic Targeting. Circ Res 2017; 121:1119-1121. [PMID: 29074528 DOI: 10.1161/circresaha.117.311984] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
566
|
Lodé L, Ameur A, Coste T, Ménard A, Richebourg S, Gaillard JB, Le Bris Y, Béné MC, Lavabre-Bertrand T, Soussi T. Single-molecule DNA sequencing of acute myeloid leukemia and myelodysplastic syndromes with multiple TP53 alterations. Haematologica 2017; 103:e13-e16. [PMID: 29079597 DOI: 10.3324/haematol.2017.176719] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Laurence Lodé
- Hematology Laboratory, University Hospital, Montpellier, F-34000, France.,Hematology Laboratory, University Hospital, Nantes, F-44093, France
| | - Adam Ameur
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Thibault Coste
- Hematology Laboratory, University Hospital, Montpellier, F-34000, France.,Department of Clinical Cytology and Cytogenetics, Nîmes University Hospital, France
| | - Audrey Ménard
- Hematology Laboratory, University Hospital, Nantes, F-44093, France
| | - Steven Richebourg
- Hematology Laboratory, University Hospital, Nantes, F-44093, France.,Cytogenetics, Hôpital du Saint Sacrement, CHU de Québec, Université Laval, Department of molecular medicine, Canada
| | | | - Yannick Le Bris
- Hematology Laboratory, University Hospital, Nantes, F-44093, France
| | | | | | - Thierry Soussi
- Sorbonne Université, UPMC Univ Paris 06, F-75005 Paris, France .,INSERM, U1138, Centre de Recherche des Cordeliers, Paris, France.,Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK) R8:04, Stockholm SE-171 76, Sweden
| |
Collapse
|
567
|
Mutations in the DNA methylation pathway and number of driver mutations predict response to azacitidine in myelodysplastic syndromes. Oncotarget 2017; 8:106948-106961. [PMID: 29291002 PMCID: PMC5739787 DOI: 10.18632/oncotarget.22157] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 10/03/2017] [Indexed: 11/25/2022] Open
Abstract
We evaluated the association of mutations in 34 candidate genes and response to azacitidine in 84 patients with myelodysplastic syndrome (MDS), with 217 somatic mutations identified by next-generation sequencing. Most patients (93%) had ≥1 mutation (mean=2.6/patient). The overall response rate to azacitidine was 42%. No clinical characteristic was associated with response to azacitidine. However, total number of mutations/patient was negatively associated with overall drug response (odds ratio [OR]: 0.56, 95% confidence interval [CI]: 0.33–0.94; p=0.028), and a positive association was found for having ≥1 mutation in a DNA methylation-related gene: TET2, DNMT3A, IDH1 and/or IDH2 (OR: 4.76, 95%CI: 1.31–17.27; p=0.017). Mutations in TP53 (hazard ratio [HR]: 3.88; 95%CI: 1.94–7.75) and EZH2 (HR: 2.50; 95%CI: 1.23–5.09) were associated with shorter overall survival. Meta-analysis of 6 studies plus present data (n=815 patients) allowed assessment of the association of drug response with mutations in 9 candidate genes: ASXL1, CBL, EZH2, SF3B1, SRSF2, TET2, DNMT3A, IDH1/2 and TP53. TET2 mutations predicted a more favorable drug response compared with ‘wild-type’ peers (pooled OR: 1.67, 95%CI: 1.14–2.44; p=0.01). In conclusion, mutations in the DNA methylation pathway, especially TET2 mutations, and low number of total mutations are associated with a better response to azacitidine.
Collapse
|
568
|
Abstract
Myelodysplastic syndromes are characterized by genetic and clinical heterogeneity. Some mutations are able to drive clonal hematopoiesis without causing clinical consequences, while other mutations may have significant impact, including the transformation to leukemia. This review aims to describe the pathogenesis of myelodysplastic syndromes (MDS) by focusing on 3 aspects: combinatorial genetic events, environmental factors, and inherited genetic conditions.
Collapse
|
569
|
Malcovati L. Introduction. Semin Hematol 2017; 54:129-132. [PMID: 28958285 DOI: 10.1053/j.seminhematol.2017.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Luca Malcovati
- Department of Molecular Medicine, University of Pavia and; Department of Hematology Oncology, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy.
| |
Collapse
|
570
|
Aldoss I, Pham A, Li SM, Gendzekhadze K, Afkhami M, Telatar M, Hong H, Padeganeh A, Bedell V, Cao T, Khaled SK, Malki MMA, Salhotra A, Ali H, Aribi A, Palmer J, Aoun P, Spielberger R, Stein AS, Snyder D, O'Donnell MR, Murata-Collins J, Senitzer D, Weisenburger D, Forman SJ, Pullarkat V, Marcucci G, Pillai R, Nakamura R. Favorable impact of allogeneic stem cell transplantation in patients with therapy-related myelodysplasia regardless of TP53 mutational status. Haematologica 2017; 102:2030-2038. [PMID: 28971906 PMCID: PMC5709102 DOI: 10.3324/haematol.2017.172544] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/20/2017] [Indexed: 01/04/2023] Open
Abstract
Therapy-related myelodysplastic syndrome is a long-term complication of cancer treatment in patients receiving cytotoxic therapy, characterized by high-risk genetics and poor outcomes. Allogeneic hematopoietic cell transplantation is the only potential cure for this disease, but the prognostic impact of pre-transplant genetics and clinical features has not yet been fully characterized. We report here the genetic and clinical characteristics and outcomes of a relatively large cohort of patients with therapy-related myelodysplastic syndrome (n=67) who underwent allogeneic transplantation, comparing these patients to similarly treated patients with de novo disease (n=199). The 5-year overall survival was not different between patients with therapy-related and de novo disease (49.9% versus 53.9%; P=0.61) despite a higher proportion of individuals with an Intermediate-2/High International Prognostic Scoring System classification (59.7% versus 43.7%; P=0.003) and high-risk karyotypes (61.2% versus 30.7%; P<0.01) among the patients with therapy-related disease. In mutational analysis, TP53 alteration was the most common abnormality in patients with therapy-related disease (n=18: 30%). Interestingly, the presence of mutations in TP53 or in any other of the high-risk genes (EZH2, ETV6, RUNX1, ASXL1: n=29: 48%) did not significantly affect either overall survival or relapse-free survival. Allogeneic stem-cell transplantation is, therefore, a curative treatment for patients with therapy-related myelodysplastic syndrome, conferring a similar long-term survival to that of patients with de novo disease despite higher-risk features. While TP53 alteration was the most common mutation in therapy-related myelodysplastic syndrome, the finding was not detrimental in our case-series.
Collapse
Affiliation(s)
- Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Anh Pham
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Sierra Min Li
- Department of Information Sciences, Division of Biostatistics, City of Hope, Duarte, CA, USA
| | | | | | | | - Hao Hong
- Department of Pathology, City of Hope, Duarte, CA, USA
| | | | | | - Thai Cao
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Samer K Khaled
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Amandeep Salhotra
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Haris Ali
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Ahmed Aribi
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Joycelynne Palmer
- Department of Information Sciences, Division of Biostatistics, City of Hope, Duarte, CA, USA
| | - Patricia Aoun
- Department of Pathology, City of Hope, Duarte, CA, USA
| | - Ricardo Spielberger
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Anthony S Stein
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - David Snyder
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Margaret R O'Donnell
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | | | | | | | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Guido Marcucci
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Raju Pillai
- Department of Pathology, City of Hope, Duarte, CA, USA
| | - Ryotaro Nakamura
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| |
Collapse
|
571
|
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
|
572
|
The evolving role of genomic testing in assessing prognosis of patients with myelodysplastic syndromes. Best Pract Res Clin Haematol 2017; 30:295-300. [PMID: 29156198 DOI: 10.1016/j.beha.2017.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The introduction into routine hematology-oncology clinical practice of molecular genetic testing assays based on next-generation sequencing platforms is prompting reassessment of the importance of molecular assay results in comparison to existing disease-specific risk stratification tools based on clinical assessment and light microscopy. For patients with myelodysplastic syndromes (MDS), the most commonly used tools for prognostication currently include the International Prognostic Scoring System (IPSS) and the Revised IPSS (IPSS-R), which are based on marrow blast proportion, number and degree of cytopenias, and the metaphase karyotype. Integration of DNA sequencing data into an existing evidence-based practice approach inclusive of the IPSS or IPSS-R may be challenging, but the additional information provided by molecular genetic testing clearly can influence clinical decisions, such as determining patients' eligibility for clinical trials of novel targeted agents or helping assess which patients should be referred for allogeneic hematopoietic stem cell transplantation. This review discusses the prognostic and predictive value of mutation testing in the context of current clinical care of patients with MDS.
Collapse
|
573
|
Systematic STAT3 sequencing in patients with unexplained cytopenias identifies unsuspected large granular lymphocytic leukemia. Blood Adv 2017; 1:1786-1789. [PMID: 29296824 DOI: 10.1182/bloodadvances.2017011197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 08/28/2017] [Indexed: 01/09/2023] Open
|
574
|
Impact of genomic alterations on outcomes in myelofibrosis patients undergoing JAK1/2 inhibitor therapy. Blood Adv 2017; 1:1729-1738. [PMID: 29296819 DOI: 10.1182/bloodadvances.2017009530] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/24/2017] [Indexed: 11/20/2022] Open
Abstract
In myelofibrosis (MF), driver mutations in JAK2, MPL, or CALR impact survival and progression to blast phase, with the greatest risk conferred by triple-negative status. Subclonal mutations, including mutations in high-molecular risk (HMR) genes, such as ASXL1, EZH2, IDH1/2, and SRSF2 have also been associated with inferior prognosis. However, data evaluating the impact of next-generation sequencing in MF patients treated with JAK1/2 inhibitors are lacking. Using a 54-gene myeloid panel, we performed targeted sequencing on 100 MF patients treated with ruxolitinib (n = 77) or momelotinib (n = 23) and correlated mutational profiles with treatment outcomes. Ninety-nine patients had at least 1 mutation identified, 46 (46%) had 2 mutations, and 34 (34%) patients had ≥3 mutations. Seventy-nine patients carried a mutation in JAK2V617F, 14 patients had mutations in CALR, 6 patients had an MPL mutation, and 2 patients were triple negative. No mutation was significantly associated with spleen or anemia response. A high Dynamic International Prognostic Scoring System score and pretreatment transfusion dependence were associated with a shorter time to treatment failure (TTF), and this association retained significance on multivariable analysis. Patients with ASXL1 (hazard ratio [HR], 1.86; P = .03) and EZH2 mutations (HR, 2.94; P = .009) and an HMR profile (HR, 2.06; P = .01) had shorter TTF. On multivariate analysis, ASXL1 or EZH2 mutations were independently associated with shorter TTF and overall survival. These findings help identify patients unlikely to have a durable response with current JAK1/2 inhibitors and provide a framework for future studies.
Collapse
|
575
|
Molecular Data and the IPSS-R: How Mutational Burden Can Affect Prognostication in MDS. Curr Hematol Malig Rep 2017; 12:461-467. [DOI: 10.1007/s11899-017-0407-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
576
|
Affiliation(s)
- Roberto Valli
- Medical Genetic Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Annalisa Frattini
- UOS Milano, Institute of Genetics and Biomedical Research, National Research Council, Milano, Italy
- Department of Medicine and Surgery, University of Insubria, Milano, Italy
| | - Antonella Minelli
- Medical Genetic Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
577
|
Coombs CC, Zehir A, Devlin SM, Kishtagari A, Syed A, Jonsson P, Hyman DM, Solit DB, Robson ME, Baselga J, Arcila ME, Ladanyi M, Tallman MS, Levine RL, Berger MF. Therapy-Related Clonal Hematopoiesis in Patients with Non-hematologic Cancers Is Common and Associated with Adverse Clinical Outcomes. Cell Stem Cell 2017; 21:374-382.e4. [PMID: 28803919 DOI: 10.1016/j.stem.2017.07.010] [Citation(s) in RCA: 595] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/27/2017] [Accepted: 07/14/2017] [Indexed: 11/26/2022]
Abstract
Clonal hematopoiesis (CH), as evidenced by recurrent somatic mutations in leukemia-associated genes, commonly occurs among aging human hematopoietic stem cells. We analyzed deep-coverage, targeted, next-generation sequencing (NGS) data of paired tumor and blood samples from 8,810 individuals to assess the frequency and clinical relevance of CH in patients with non-hematologic malignancies. We identified CH in 25% of cancer patients, with 4.5% harboring presumptive leukemia driver mutations (CH-PD). CH was associated with increased age, prior radiation therapy, and tobacco use. PPM1D and TP53 mutations were associated with prior exposure to chemotherapy. CH and CH-PD led to an increased incidence of subsequent hematologic cancers, and CH-PD was associated with shorter patient survival. These data suggest that CH occurs in an age-dependent manner and that specific perturbations can enhance fitness of clonal hematopoietic stem cells, which can impact outcome through progression to hematologic malignancies and through cell-non-autonomous effects on solid tumor biology.
Collapse
Affiliation(s)
- Catherine C Coombs
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ashwin Kishtagari
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Aijazuddin Syed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Philip Jonsson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David M Hyman
- Department of Medicine, Developmental Therapeutics Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - David B Solit
- Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mark E Robson
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Hereditary Cancer and Genetics Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - José Baselga
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Martin S Tallman
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Ross L Levine
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Memorial Sloan Kettering Cancer Center, Center for Hematologic Malignancies, New York, NY 10065, USA.
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
578
|
If All You Have Is a Hammer…: Transplantation for Myelodysplastic Syndrome after Hypomethylating Agents Fail. Biol Blood Marrow Transplant 2017; 23:1413-1414. [PMID: 28711726 DOI: 10.1016/j.bbmt.2017.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 11/20/2022]
|
579
|
Kröger N. Maximizing the benefit of allogeneic stem cell transplantation in myelodysplastic syndromes. Semin Hematol 2017; 54:154-158. [DOI: 10.1053/j.seminhematol.2017.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/13/2017] [Indexed: 12/17/2022]
|
580
|
Kharfan-Dabaja MA, Komrokji RS, Zhang Q, Kumar A, Tsalatsanis A, Perkins J, Nishihori T, Field T, Al Ali N, Mishra A, Sallman D, Salem KZ, Zhang L, Moscinski L, Fernandez HF, Lancet J, List A, Anasetti C, Padron E. TP53 and IDH2 Somatic Mutations Are Associated With Inferior Overall Survival After Allogeneic Hematopoietic Cell Transplantation for Myelodysplastic Syndrome. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:753-758. [PMID: 28687222 DOI: 10.1016/j.clml.2017.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/08/2017] [Accepted: 06/09/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Next-generation sequencing has identified somatic mutations that are prognostic of cancer. PATIENTS AND METHODS We evaluated the incidence and prognostic significance of somatic mutations in 89 myelodysplastic syndrome (MDS) patients who received an allogeneic hematopoietic cell transplantation. Next-generation sequencing was performed on paraffin embedded bone marrow, which was obtained at a median of 31 days before initiating the preparative regimen. RESULTS The 3 most common subtypes of MDS were refractory anemia with excess blasts (RAEB)-1 (35%), RAEB-2 (29%), and refractory cytopenia with multilineage dysplasia (18%). Most patients (91%) received a myeloablative regimen of fludarabine with intravenous busulfan. Somatic mutations (> 0) were identified in 39 (44%) of analyzed samples. The 6 most commonly identified gene mutations were ASXL1 (8%), DNMT3A (8%), RUNX1 (7%), KRAS (6%), IDH2 (4%), and TP53 (4%). The low incidence of mutations in our study sample might be explained by tissue source and stringent variant-calling methodology. Moreover, we speculate that the low incidence of mutations might, perhaps, also be explained by previous azacitidine treatment in 82% of cases. Multivariate analysis identified TP53 (hazard ratio [HR], 3.82; 95% confidence interval [CI], 1.12-13.09; P = .03) and IDH2 mutations (HR, 4.74; 95% CI, 1.33-16.91; P = .02) as predictors of inferior 3-year overall survival. CONCLUSION This study furthers implementation of clinical genomics in MDS and identifies TP53 and IDH2 as targets for pre- or post-transplant therapy.
Collapse
Affiliation(s)
- Mohamed A Kharfan-Dabaja
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL.
| | - Rami S Komrokji
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Qing Zhang
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Ambuj Kumar
- University of South Florida Health Program for Comparative Effectiveness Research, Tampa, FL
| | - Athanasios Tsalatsanis
- University of South Florida Health Program for Comparative Effectiveness Research, Tampa, FL
| | | | - Taiga Nishihori
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| | - Teresa Field
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| | - Najla Al Ali
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Asmita Mishra
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| | - David Sallman
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Karma Z Salem
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| | - Ling Zhang
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL
| | - Lynn Moscinski
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL
| | - Hugo F Fernandez
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| | - Jeffrey Lancet
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Alan List
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| | - Eric Padron
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
581
|
Diagnosis and classification of hematologic malignancies on the basis of genetics. Blood 2017; 130:410-423. [PMID: 28600336 DOI: 10.1182/blood-2017-02-734541] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Genomic analysis has greatly influenced the diagnosis and clinical management of patients affected by diverse forms of hematologic malignancies. Here, we review how genetic alterations define subclasses of patients with acute leukemias, myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPNs), non-Hodgkin lymphomas, and classical Hodgkin lymphoma. These include new subtypes of acute myeloid leukemia defined by mutations in RUNX1 or BCR-ABL1 translocations as well as a constellation of somatic structural DNA alterations in acute lymphoblastic leukemia. Among patients with MDS, detection of mutations in SF3B1 define a subgroup of patients with the ring sideroblast form of MDS and a favorable prognosis. For patients with MPNs, detection of the BCR-ABL1 fusion delineates chronic myeloid leukemia from classic BCR-ABL1- MPNs, which are largely defined by mutations in JAK2, CALR, or MPL In the B-cell lymphomas, detection of characteristic rearrangements involving MYC in Burkitt lymphoma, BCL2 in follicular lymphoma, and MYC/BCL2/BCL6 in high-grade B-cell lymphomas are essential for diagnosis. In T-cell lymphomas, anaplastic large-cell lymphoma is defined by mutually exclusive rearrangements of ALK, DUSP22/IRF4, and TP63 Genetic alterations affecting TP53 and the mutational status of the immunoglobulin heavy-chain variable region are important in clinical management of chronic lymphocytic leukemia. Additionally, detection of BRAFV600E mutations is helpful in the diagnosis of classical hairy cell leukemia and a number of histiocytic neoplasms. Numerous additional examples provided here demonstrate how clinical evaluation of genomic alterations have refined classification of myeloid neoplasms and major forms of lymphomas arising from B, T, or natural killer cells.
Collapse
|
582
|
Varaldo R, Raiola AM, Di Grazia C, Aquino S, Beltrami G, Bregante S, Cruciani F, Dominietto A, Ghiso A, Giannoni L, Gualandi F, Ibatici A, Lamparelli T, Marani C, Van Lint MT, Santini V, Bacigalupo A, Angelucci E. Haploidentical bone marrow transplantation in patients with advanced myelodysplastic syndrome. Am J Hematol 2017; 92:E117-E119. [PMID: 28316085 DOI: 10.1002/ajh.24725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/11/2017] [Accepted: 03/15/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Riccardo Varaldo
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Anna Maria Raiola
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Carmen Di Grazia
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Sara Aquino
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Germana Beltrami
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Stefania Bregante
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Fabio Cruciani
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Alida Dominietto
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Anna Ghiso
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Livia Giannoni
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Francesca Gualandi
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Adalberto Ibatici
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Teresa Lamparelli
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Carlo Marani
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Maria Teresa Van Lint
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Valeria Santini
- AOU Careggi, Università degli Studi di Firenze; Firenze Italy
- FISM: Fondazione Italiana Sindromi mielodisplastiche; Alessandria Italy
| | - Andrea Bacigalupo
- Istituto di Ematologia, Università Cattolica del Sacro Cuore, Fondazione, Policlinico Universitario Gemelli Roma; Italy
| | - Emanuele Angelucci
- U.O. Ematologia IRCCS Azienda Ospedaliera Universitaria San Martino-IST. Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
- FISM: Fondazione Italiana Sindromi mielodisplastiche; Alessandria Italy
| |
Collapse
|
583
|
Allogeneic Hematopoietic Cell Transplantation for Older Patients: Prognosis Determined by Disease Risk Index. Biol Blood Marrow Transplant 2017; 23:1485-1490. [PMID: 28522345 DOI: 10.1016/j.bbmt.2017.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/11/2017] [Indexed: 12/28/2022]
Abstract
The treatment of elderly patients with advanced hematological malignancies has expanded to include reduced-intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (alloHCT) as a potentially curative option. We studied the association between Disease Risk Index (DRI) and clinical outcomes of 196 elderly patients (median age, 64.8; range, 60 to 75 years) with hematological malignancies receiving RIC alloHCT (2000 to 2014). Donors were related and unrelated adults (n = 100, 51.1%) or umbilical cord blood (n = 96, 48.9%). DRI classified 12 patients (6.1%) as low risk (LR), 146 patients (74.5%) as intermediate risk (IR), and 38 patients (19.4%) as high risk (HR). Two-year overall survival (OS) was 47% (52% for LR/IR versus 29% for HR, P < .01) and 2-year disease-free survival was 39% (44% for LR/IR versus 21% for HR, P < .01). Relapse incidence was 30% (26% for LR/IR versus 44% for HR, P < .01). Treatment-related mortality was 29% at 2 years; this was similar for all DRI groups. In multiple regression analysis, HR DRI was associated with increased risk of relapse (hazard ratio, 2.07; 95% confidence interval [CI], 1.34 to 3.33; P = .02) and treatment failure (hazard ratio, 2.07; 95% CI, 1.35 to 3.18; P < .01) and decreased OS (hazard ratio, 2.11; 95% CI, 1.34 to 3.33; P < .01). In elderly patients, DRI is a significant prognostic factor for post-transplantation relapse, treatment failure, and mortality. Because of increased risk of relapse leading to poor survival in HR DRI, participation in clinical trials offering relapse prevention strategies after RIC alloHCT should be encouraged when available.
Collapse
|
584
|
Allogeneic hematopoietic stem cell transplantation for MDS and CMML: recommendations from an international expert panel. Blood 2017; 129:1753-1762. [PMID: 28096091 DOI: 10.1182/blood-2016-06-724500] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 01/04/2017] [Indexed: 01/19/2023] Open
Abstract
An international expert panel, active within the European Society for Blood and Marrow Transplantation, European LeukemiaNet, Blood and Marrow Transplant Clinical Trial Group, and the International Myelodysplastic Syndromes Foundation developed recommendations for allogeneic hematopoietic stem cell transplantation (HSCT) in myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML). Disease risks scored according to the revised International Prognostic Scoring System (IPSS-R) and presence of comorbidity graded according to the HCT Comorbidity Index (HCT-CI) were recognized as relevant clinical variables for HSCT eligibility. Fit patients with higher-risk IPSS-R and those with lower-risk IPSS-R with poor-risk genetic features, profound cytopenias, and high transfusion burden are candidates for HSCT. Patients with a very high MDS transplantation risk score, based on combination of advanced age, high HCT-CI, very poor-risk cytogenetic and molecular features, and high IPSS-R score have a low chance of cure with standard HSCT and consideration should be given to treating these patients in investigational studies. Cytoreductive therapy prior to HSCT is advised for patients with ≥10% bone marrow myeloblasts. Evidence from prospective randomized clinical trials does not provide support for specific recommendations on the optimal high intensity conditioning regimen. For patients with contraindications to high-intensity preparative regimens, reduced intensity conditioning should be considered. Optimal timing of HSCT requires careful evaluation of the available effective nontransplant strategies. Prophylactic donor lymphocyte infusion (DLI) strategies are recommended in patients at high risk of relapse after HSCT. Immune modulation by DLI strategies or second HSCT is advised if relapse occurs beyond 6 months after HSCT.
Collapse
|