1
|
Nitschke NJ, Jelsig AM, Lautrup C, Lundsgaard M, Severinsen MT, Cowland JB, Maroun LL, Andersen MK, Grønbæk K. Expanding the understanding of telomere biology disorder with reports from two families harboring variants in ZCCHC8 and TERC. Clin Genet 2024; 106:187-192. [PMID: 38606545 DOI: 10.1111/cge.14534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/04/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
Telomere biology disorder (TBD) can present within a wide spectrum of symptoms ranging from severe congenital malformations to isolated organ dysfunction in adulthood. Diagnosing TBD can be challenging given the substantial variation in symptoms and age of onset across generations. In this report, we present two families, one with a pathogenic variant in ZCCHC8 and another with a novel variant in TERC. In the literature, only one family has previously been reported with a ZCCHC8 variant and TBD symptoms. This family had multiple occurrences of pulmonary fibrosis and one case of bone marrow failure. In this paper, we present a second family with the same ZCCHC8 variant (p.Pro186Leu) and symptoms of TBD including pulmonary fibrosis, hematological disease, and elevated liver enzymes. The suspicion of TBD was confirmed with the measurement of short telomeres in the proband. In another family, we report a novel likely pathogenic variant in TERC. Our comprehensive description encompasses hematological manifestations, as well as pulmonary and hepatic fibrosis. Notably, there are no other reports which associate this variant to disease. The families expand our understanding of the clinical implications and genetic causes of TBD.
Collapse
Affiliation(s)
- Nikolaj Juul Nitschke
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Anne Marie Jelsig
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Lautrup
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Malene Lundsgaard
- Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark
| | - Marianne Tang Severinsen
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jack Bernard Cowland
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lisa Leth Maroun
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mette Klarskov Andersen
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
García-Solorio J, Martínez-Villegas O, Rodríguez-Corona U, Molina-Garay C, Jiménez-Olivares M, Carrillo-Sanchez K, Mendoza-Caamal EC, Muñoz-Rivas A, Villegas-Torres BE, Cervera A, Flores-Lagunes LL, Alaez-Verson C. Case report: A familial B-acute lymphoblastic leukemia associated with a new germline pathogenic variant in PAX5. The first report in Mexico. Front Oncol 2024; 14:1355335. [PMID: 38571503 PMCID: PMC10987763 DOI: 10.3389/fonc.2024.1355335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/06/2024] [Indexed: 04/05/2024] Open
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is one of the most common childhood cancers worldwide. Although most cases are sporadic, some familial forms, inherited as autosomal dominant traits with incomplete penetrance, have been described over the last few years. Germline pathogenic variants in transcription factors such as PAX5, IKZF1, and ETV6 have been identified as causal in familial forms. The proband was a 7-year-old Mexican girl diagnosed with high-risk B-ALL at five years and 11 months of age. Family history showed that the proband's mother had high-risk B-ALL at 16 months of age. She received chemotherapy and was discharged at nine years of age without any evidence of recurrence of leukemia. The proband's father was outside the family nucleus, but no history of leukemia or cancer was present up to the last contact with the mother. We performed exome sequencing on the proband and the proband's mother and identified the PAX5 variant NM_016734.3:c.963del: p.(Ala322LeufsTer11), located in the transactivation domain of the PAX5 protein. The variant was classified as probably pathogenic according to the ACMG criteria. To the best of our knowledge, this is the first Mexican family with an inherited increased risk of childhood B-ALL caused by a novel germline pathogenic variant of PAX5. Identifying individuals with a hereditary predisposition to cancer is essential for modern oncological practice. Individuals at high risk of leukemia would benefit from hematopoietic stem cell transplantation, but family members carrying the pathogenic variant should be excluded as hematopoietic stem cell donors.
Collapse
Affiliation(s)
- Joaquín García-Solorio
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Octavio Martínez-Villegas
- Departamento de hemato-oncología pediátrica, Unidad Médicade Alta Especialidad Hospital de Ginecología Pediatría No 48, Centro Médico del Bajío, León, Guanajuato, Mexico
| | - Ulises Rodríguez-Corona
- Ribonucleo Protein Biochemistry Research Unit, Montreal Clinical Research Institute, Montreal, QC, Canada
| | - Carolina Molina-Garay
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Marco Jiménez-Olivares
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Karol Carrillo-Sanchez
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Elvia C. Mendoza-Caamal
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Anallely Muñoz-Rivas
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Beatriz E. Villegas-Torres
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Alejandra Cervera
- Subdirección de Genómica Poblacional, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Luis L. Flores-Lagunes
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Carmen Alaez-Verson
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
3
|
Kok CH, Yeung DT, Hiwase DK. Special Issue "Advances in Molecular Pathogenesis and Targeted Therapies for Myeloid Neoplasms". Int J Mol Sci 2024; 25:2056. [PMID: 38396733 PMCID: PMC10888731 DOI: 10.3390/ijms25042056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/22/2023] [Indexed: 02/25/2024] Open
Abstract
Myeloid neoplasms (MNs) constitute a diverse group of haematological malignancies that includes myelodysplastic neoplasms (MDS), myeloproliferative neoplasms (MPN), MDS/MPN overlap syndrome, and acute myeloid leukaemia (AML) [...].
Collapse
Affiliation(s)
- Chung Hoow Kok
- Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide 5000, Australia;
- Adelaide Medical School, University of Adelaide, Adelaide 5000, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - David T. Yeung
- Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide 5000, Australia;
- Adelaide Medical School, University of Adelaide, Adelaide 5000, Australia
- Department of Haematology, Royal Adelaide Hospital and SA Pathology, Adelaide 5000, Australia
| | - Devendra K. Hiwase
- Precision Cancer Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide 5000, Australia;
- Adelaide Medical School, University of Adelaide, Adelaide 5000, Australia
- Department of Haematology, Royal Adelaide Hospital and SA Pathology, Adelaide 5000, Australia
| |
Collapse
|
4
|
Arai H, Matsui H, Chi S, Utsu Y, Masuda S, Aotsuka N, Minami Y. Germline Variants and Characteristic Features of Hereditary Hematological Malignancy Syndrome. Int J Mol Sci 2024; 25:652. [PMID: 38203823 PMCID: PMC10779750 DOI: 10.3390/ijms25010652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Due to the proliferation of genetic testing, pathogenic germline variants predisposing to hereditary hematological malignancy syndrome (HHMS) have been identified in an increasing number of genes. Consequently, the field of HHMS is gaining recognition among clinicians and scientists worldwide. Patients with germline genetic abnormalities often have poor outcomes and are candidates for allogeneic hematopoietic stem cell transplantation (HSCT). However, HSCT using blood from a related donor should be carefully considered because of the risk that the patient may inherit a pathogenic variant. At present, we now face the challenge of incorporating these advances into clinical practice for patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) and optimizing the management and surveillance of patients and asymptomatic carriers, with the limitation that evidence-based guidelines are often inadequate. The 2016 revision of the WHO classification added a new section on myeloid malignant neoplasms, including MDS and AML with germline predisposition. The main syndromes can be classified into three groups. Those without pre-existing disease or organ dysfunction; DDX41, TP53, CEBPA, those with pre-existing platelet disorders; ANKRD26, ETV6, RUNX1, and those with other organ dysfunctions; SAMD9/SAMD9L, GATA2, and inherited bone marrow failure syndromes. In this review, we will outline the role of the genes involved in HHMS in order to clarify our understanding of HHMS.
Collapse
Affiliation(s)
- Hironori Arai
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Hirotaka Matsui
- Department of Laboratory Medicine, National Cancer Center Hospital, Tsukiji, Chuoku 104-0045, Japan;
- Department of Medical Oncology and Translational Research, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8665, Japan
| | - SungGi Chi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
| | - Yoshikazu Utsu
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Shinichi Masuda
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Nobuyuki Aotsuka
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
| |
Collapse
|
5
|
Guijarro F, López-Guerra M, Morata J, Bataller A, Paz S, Cornet-Masana JM, Banús-Mulet A, Cuesta-Casanovas L, Carbó JM, Castaño-Díez S, Jiménez-Vicente C, Cortés-Bullich A, Triguero A, Martínez-Roca A, Esteban D, Gómez-Hernando M, Álamo Moreno JR, López-Oreja I, Garrote M, Risueño RM, Tonda R, Gut I, Colomer D, Díaz-Beya M, Esteve J. Germ line variants in patients with acute myeloid leukemia without a suspicion of hereditary hematologic malignancy syndrome. Blood Adv 2023; 7:5799-5811. [PMID: 37450374 PMCID: PMC10561046 DOI: 10.1182/bloodadvances.2023009742] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/04/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Germ line predisposition in acute myeloid leukemia (AML) has gained attention in recent years because of a nonnegligible frequency and an impact on management of patients and their relatives. Risk alleles for AML development may be present in patients without a clinical suspicion of hereditary hematologic malignancy syndrome. In this study we investigated the presence of germ line variants (GVs) in 288 genes related to cancer predisposition in 47 patients with available paired, tumor-normal material, namely bone marrow stroma cells (n = 29), postremission bone marrow (n = 17), and saliva (n = 1). These patients correspond to 2 broad AML categories with heterogeneous genetic background (AML myelodysplasia related and AML defined by differentiation) and none of them had phenotypic abnormalities, previous history of cytopenia, or strong cancer aggregation. We found 11 pathogenic or likely pathogenic variants, 6 affecting genes related to autosomal dominant cancer predisposition syndromes (ATM, DDX41, and CHEK2) and 5 related to autosomal recessive bone marrow failure syndromes (FANCA, FANCM, SBDS, DNAJC21, and CSF3R). We did not find differences in clinical characteristics nor outcome between carriers of GVs vs noncarriers. Further studies in unselected AML cohorts are needed to determine GV incidence and penetrance and, in particular, to clarify the role of ATM nonsense mutations in AML predisposition.
Collapse
Affiliation(s)
- Francesca Guijarro
- Pathology Department, Hematopathology Section, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Monica López-Guerra
- Pathology Department, Hematopathology Section, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Jordi Morata
- Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - Alex Bataller
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Sara Paz
- Pathology Department, Hematopathology Section, Hospital Clínic Barcelona, Barcelona, Spain
| | | | | | | | | | - Sandra Castaño-Díez
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Carlos Jiménez-Vicente
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Albert Cortés-Bullich
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Ana Triguero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Alexandra Martínez-Roca
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Daniel Esteban
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
| | - Marta Gómez-Hernando
- Pathology Department, Hematopathology Section, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Irene López-Oreja
- Pathology Department, Hematopathology Section, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Marta Garrote
- Pathology Department, Hematopathology Section, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Ruth M. Risueño
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Raúl Tonda
- Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - Ivo Gut
- Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - Dolors Colomer
- Pathology Department, Hematopathology Section, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- University of Barcelona, Barcelona, Spain
| | - Marina Díaz-Beya
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Jordi Esteve
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hematology Department, Hospital Clínic Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Chin HL, Lam JCM, Christopher D, Michelle PL, Junrong BY. Challenges associated with the identification of germline variants on myeloid malignancy genomic profiling-a Singaporean experience. Front Oncol 2023; 13:1182639. [PMID: 37860182 PMCID: PMC10582742 DOI: 10.3389/fonc.2023.1182639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023] Open
Abstract
Genomic profiling to identify myeloid-malignancy-related gene mutations is routinely performed for patients with suspected or definite myeloid malignancies. The most common specimen types in our experience are peripheral blood and bone marrow aspirates. Although primarily intended to identify somatic mutations, not infrequently, potentially clinically significant germline variants are also identified. Confirmation of the germline status of these variants is typically performed by hair follicle or skin fibroblast testing. If the germline variant is classified as a pathogenic or likely pathogenic variant and occurs in a gene known to be associated with a disease relevant to the patient's phenotype (for example, the identification of a DDX41 pathogenic variant in an individual with acute myeloid leukemia), the management algorithm is typically quite straightforward. Challenging situations may occur such as when the germline variant is classified as a pathogenic or likely pathogenic variant and occurs in a gene not known to be associated with the patient's phenotype/presenting complaint. We have encountered several such challenging cases in which potentially clinically significant germline variants were identified on the initial genomic profiling of peripheral blood or bone marrow aspirate. In this article, we present these cases and discuss the genetic counseling and management approaches.
Collapse
Affiliation(s)
- Hui-Lin Chin
- Khoo Teck Puat National University Children's Medical Institute, Department of Paediatrics, National University Hospital, Singapore, Singapore
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Joyce Ching Mei Lam
- Children’s Blood and Cancer Centre, KK Women’s and Children’s Hospital, Singapore, Singapore
- Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Dheepa Christopher
- Department of Haematology, Tan Tock Seng Hospital, Singapore, Singapore
- Department of Laboratory Medicine, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Poon Limei Michelle
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
| | - Benedict Yan Junrong
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| |
Collapse
|
7
|
O’Connor TE, Shaw R, Madero-Marroquin R, Roloff GW. Clinical considerations at the intersection of hematopoietic cell transplantation and hereditary hematopoietic malignancy. Front Oncol 2023; 13:1180439. [PMID: 37251919 PMCID: PMC10213438 DOI: 10.3389/fonc.2023.1180439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
In recent years, advances in genetics and the integration of clinical-grade next-generation sequencing (NGS) assays into patient care have facilitated broader recognition of hereditary hematopoietic malignancy (HHM) among clinicians, in addition to the identification and characterization of novel HHM syndromes. Studies on genetic risk distribution within affected families and unique considerations of HHM biology represent exciting areas of translational research. More recently, data are now emerging pertaining to unique aspects of clinical management of malignancies arising in the context of pathogenic germline mutations, with particular emphasis on chemotherapy responsiveness. In this article, we explore considerations surrounding allogeneic transplantation in the context of HHMs. We review pre- and post-transplant patient implications, including genetic testing donor selection and donor-derived malignancies. Additionally, we consider the limited data that exist regarding the use of transplantation in HHMs and safeguards that might be pursued to mitigate transplant-related toxicities.
Collapse
Affiliation(s)
- Timothy E. O’Connor
- Department of Medicine, Loyola University Medical Center, Maywood, IL, United States
| | - Reid Shaw
- Department of Medicine, Loyola University Medical Center, Maywood, IL, United States
| | | | - Gregory W. Roloff
- Section of Hematology/Oncology, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
8
|
Feasibility of Leukemia-Derived Exosome Enrichment and Co-isolated dsDNA Sequencing in Acute Myeloid Leukemia Patients: A Proof of Concept for New Leukemia Biomarkers Detection. Cancers (Basel) 2022; 14:cancers14184504. [PMID: 36139664 PMCID: PMC9497185 DOI: 10.3390/cancers14184504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The present pilot study aimed at investigating the feasibility of a leukemia-derived exosome enrichment approach followed by exosomal dsDNA target re-sequencing for adult Acute Myeloid Leukemias (AML) marker detection. To our knowledge, this is the first time that a proof-of-concept combining a leukemia-derived exosome enrichment strategy based on a commercial CE-IVD kit and next-generation sequencing was applied in a cohort of adult AML patients. The reported approach is easy, quick and user friendly and gives the possibility of obtaining a good quantity of exosomal dsDNA (composed of exosomal cargo and surrounding DNA) suitable for further analysis. The time-effective procedure opens up future effective clinical applications. This pilot study presents the potential of a proof-of-concept based on exosome analysis to be applied in clinical practice, as well as the feasibility of this kind of investigations using a certified kit, avoiding many additional analyses. It may encourage further studies regarding extracellular vesicles in myeloid neoplasia. Abstract Exosomes are extracellular vesicles playing a pivotal role in the intercellular communication. They shuttle different cargoes, including nucleic acids from their cell of origin. For this reason, they have been studied as carriers of tumor markers in different liquid biopsy approaches, in particular for solid tumors. Few data are available concerning exosomes as markers of myeloid neoplasia. To better understand their real potential and the best approach to investigate leukemic exosomes, we present the results of a pilot feasibility study evaluating the application of next-generation sequencing analysis of dsDNA derived from exosomes isolated in 14 adult patients affected by acute myeloid leukemias. In particular, leukemia-derived exosome fractions have been analyzed. The concentration of dsDNA co-extracted with exosomes and the number and types of mutations detected were considered and compared with ones identified in the Bone Marrow (BM) and Peripheral Blood (PB) cells. Exosomal DNA concentration, both considering the cargo and the DNA surrounding the lipid membrane resulted in a linear correlation with leukemic burden. Moreover, exosomal DNA mutation status presented 86.5% of homology with BM and 75% with PB. The results of this pilot study confirmed the feasibility of a leukemia-derived exosome enrichment approach followed by exosomal dsDNA NGS analysis for AML biomarker detection. These data point to the use of liquid biopsy in myeloid neoplasia for the detection of active leukemic cells resident in the BM via a painless procedure.
Collapse
|
9
|
Duployez N, Largeaud L, Duchmann M, Kim R, Rieunier J, Lambert J, Bidet A, Larcher L, Lemoine J, Delhommeau F, Hirsch P, Fenwarth L, Kosmider O, Decroocq J, Bouvier A, Le Bris Y, Ochmann M, Santagostino A, Adès L, Fenaux P, Thomas X, Micol JB, Gardin C, Itzykson R, Soulier J, Clappier E, Recher C, Preudhomme C, Pigneux A, Dombret H, Delabesse E, Sébert M. Prognostic impact of DDX41 germline mutations in intensively treated acute myeloid leukemia patients: an ALFA-FILO study. Blood 2022; 140:756-768. [PMID: 35443031 PMCID: PMC9389637 DOI: 10.1182/blood.2021015328] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/01/2022] [Indexed: 11/20/2022] Open
Abstract
DDX41 germline mutations (DDX41MutGL) are the most common genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia (AML). Recent reports suggest that DDX41MutGL myeloid malignancies could be considered as a distinct entity, even if their specific presentation and outcome remain to be defined. We describe here the clinical and biological features of 191 patients with DDX41MutGL AML. Baseline characteristics and outcome of 86 of these patients, treated with intensive chemotherapy in 5 prospective Acute Leukemia French Association/French Innovative Leukemia Organization trials, were compared with those of 1604 patients with DDX41 wild-type (DDX41WT) AML, representing a prevalence of 5%. Patients with DDX41MutGL AML were mostly male (75%), in their seventh decade, and with low leukocyte count (median, 2 × 109/L), low bone marrow blast infiltration (median, 33%), normal cytogenetics (75%), and few additional somatic mutations (median, 2). A second somatic DDX41 mutation (DDX41MutSom) was found in 82% of patients, and clonal architecture inference suggested that it could be the main driver for AML progression. DDX41MutGL patients displayed higher complete remission rates (94% vs 69%; P < .0001) and longer restricted mean overall survival censored at hematopoietic stem cell transplantation (HSCT) than 2017 European LeukemiaNet intermediate/adverse (Int/Adv) DDX41WT patients (5-year difference in restricted mean survival times, 13.6 months; P < .001). Relapse rates censored at HSCT were lower at 1 year in DDX41MutGL patients (15% vs 44%) but later increased to be similar to Int/Adv DDX41WT patients at 3 years (82% vs 75%). HSCT in first complete remission was associated with prolonged relapse-free survival (hazard ratio, 0.43; 95% confidence interval, 0.21-0.88; P = .02) but not with longer overall survival (hazard ratio, 0.77; 95% confidence interval, 0.35-1.68; P = .5).
Collapse
Affiliation(s)
- Nicolas Duployez
- Hematology Laboratory, Unité 1277-Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), Centre Hospitalier Universitaire (CHU) de Lille, University of Lille, INSERM, Lille, France
| | - Laëtitia Largeaud
- Hematology Laboratory, CHU de Toulouse-Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Matthieu Duchmann
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Rathana Kim
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Julie Rieunier
- Hematology Laboratory, CHU de Toulouse-Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | | | - Audrey Bidet
- Hematology Laboratory, CHU de Bordeaux, Bordeaux, France
| | - Lise Larcher
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean Lemoine
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - François Delhommeau
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Laboratoire d'hématologie biologique, Hôpital Saint-Antoine, Paris, France
| | - Pierre Hirsch
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Laboratoire d'hématologie biologique, Hôpital Saint-Antoine, Paris, France
| | - Laurène Fenwarth
- Hematology Laboratory, Unité 1277-Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), Centre Hospitalier Universitaire (CHU) de Lille, University of Lille, INSERM, Lille, France
| | | | | | - Anne Bouvier
- Hematology Laboratory, CHU Angers, Angers, France
| | - Yannick Le Bris
- Hematology Biology, Nantes University Hospital, Nantes, France
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | | | | | - Lionel Adès
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Pierre Fenaux
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Xavier Thomas
- Hematology Department, Hospices Civils de Lyon, Lyon-Sud Hospital, Lyon, France
| | - Jean-Baptiste Micol
- Hematology Department, Gustave Roussy Institute, University of Paris-Saclay, Villejuif, France
| | - Claude Gardin
- Hematology Department, Avicenne Hospital, AP-HP, Bobigny, France
- Unité 3518, Saint-Louis Institute for Research, Université de Paris, Paris, France
| | - Raphael Itzykson
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Jean Soulier
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Emmanuelle Clappier
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Christian Recher
- Service d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université Toulouse III Paul Sabatier, Toulouse, France; and
| | - Claude Preudhomme
- Hematology Laboratory, Unité 1277-Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), Centre Hospitalier Universitaire (CHU) de Lille, University of Lille, INSERM, Lille, France
| | - Arnaud Pigneux
- Hematology Department, CHU de Bordeaux, Bordeaux, France
| | - Hervé Dombret
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
- Unité 3518, Saint-Louis Institute for Research, Université de Paris, Paris, France
| | - Eric Delabesse
- Hematology Laboratory, CHU de Toulouse-Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Marie Sébert
- Université de Paris, Unité 944/7212-GenCellDi, INSERM and Centre National de la Recherche Scientifique (CNRS), Paris, France
- Hematology Department, Saint Louis Hospital, AP-HP, Paris, France
| |
Collapse
|
10
|
DeRoin L, Cavalcante de Andrade Silva M, Petras K, Arndt K, Phillips N, Wanjari P, Subramanian HP, Montes D, McElherne J, Theissen M, Briese R, Das S, Godley LA, Segal J, del Gaudio D, Fitzpatrick C, Churpek JE. Feasibility and limitations of cultured skin fibroblasts for germline genetic testing in hematologic disorders. Hum Mutat 2022; 43:950-962. [PMID: 35419889 PMCID: PMC9177640 DOI: 10.1002/humu.24374] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/05/2022] [Accepted: 03/26/2022] [Indexed: 12/19/2022]
Abstract
To avoid acquired variants found in the blood, cultured skin fibroblasts are a recommended DNA source for germline genetic testing in patients with hematologic disorders, but data are lacking regarding practicality and limitations. We conducted a retrospective cohort study of 350 subjects with hematologic disorders who underwent skin fibroblast culture for germline genetic testing. We analyzed next-generation sequencing data from the targeted capture of 144 inherited cancer and bonemarrow failure genes to identify variants at heterozygous and subclonal variant allele frequencies. Sixteen (5%) biopsies failed to culture. Culture failure was more likely in samples with delays in culture initiation (OR = 4.3; p < 0.01) or a pathogenic variant in a telomere gene (OR = 42.6; p < 0.01). Median culture time was 28 days (IQR 22-29 days). Culture time was longer for subjects with prior allogeneic stem cell transplantation (+10.7%; p = 0.02) and shorter in subjects with a heterozygous pathogenic variant (-11.9%; p < 0.01), larger biopsy size (-10.6%; p < 0.01), or lymphoid malignancy (-8.4%; p < 0.01). Subclonal variants were identified in 10 (4%) and confirmed in five (56%) of eight with alternate samples available. Subclonal and discordant variants illustrate that germline testing from cultured skin fibroblasts requires phenotypic correlation and, in rare cases, follow-up studies for optimal interpretation.
Collapse
Affiliation(s)
- Lia DeRoin
- Pritzker School of MedicineUniversity of ChicagoChicagoIllinoisUSA
| | | | - Kristin Petras
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Kelly Arndt
- Department of Human GeneticsUniversity of ChicagoChicagoIllinoisUSA
| | | | | | | | - David Montes
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - James McElherne
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Megan Theissen
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Renee Briese
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Soma Das
- Department of Human GeneticsUniversity of ChicagoChicagoIllinoisUSA
| | - Lucy A. Godley
- Section of Hematology/Oncology and Center for Clinical Cancer GeneticsUniversity of ChicagoChicagoIllinoisUSA
| | - Jeremy Segal
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | | | | | - Jane E. Churpek
- Section of Hematology/Oncology and Center for Clinical Cancer GeneticsUniversity of ChicagoChicagoIllinoisUSA
- Present address:
Jane E. Churpek, Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine & Carbone Cancer CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWIUSA
| |
Collapse
|
11
|
Yan X, Lai B, Zhou X, Yang S, Ge Q, Zhou M, Shi C, Xu Z, Ouyang G. The Differential Expression of CD47 may be Related to the Pathogenesis From Myelodysplastic Syndromes to Acute Myeloid Leukemia. Front Oncol 2022; 12:872999. [PMID: 35433462 PMCID: PMC9008711 DOI: 10.3389/fonc.2022.872999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Myelodysplastic syndrome (MDS) can lead to the development of peripheral blood cytopenia and abnormal cell morphology. MDS has the potential to evolve into AML and can lead to reduced survival. CD47, a member of the immunoglobulin family, is one molecule that is overexpressed in a variety of cancer cells and is associated with clinical features and poor prognosis in a variety of malignancies. In this study, we analyzed the expression and function of CD47 in MDS and AML, and further analyzed its role in other tumors. Our analysis revealed significantly low CD47 expression in MDS and significantly high expression in AML. Further analysis of the function or pathway of CD47 from different perspectives identified a relationship to the immune response, cell growth, and other related functions or pathways. The relationship between CD47 and other tumors was analyzed from four aspects: DNA methyltransferase, TMB, MSI, and tumor cell stemness. Changes in gene expression levels have a known association with aberrant DNA methylation, and this methylation is the main mechanism of tumor suppressor gene silencing and clonal variation during the evolution of MDS to AML. Taken together, our findings support the hypothesis that the differential expression of CD47 might be related to the transformation of MDS to AML.
Collapse
Affiliation(s)
- Xiao Yan
- Haematology Department of Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo, China
| | - Binbin Lai
- Haematology Department of Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo, China
| | - Xuyan Zhou
- Haematology Department, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Shujun Yang
- Medical Research Center of Ningbo First Hospital, Ningbo, China
| | - Qunfang Ge
- Haematology Department of Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo, China
| | - Miao Zhou
- Haematology Department of Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo, China
| | - Cong Shi
- Stem Cell Transplantation Laboratory of Ningbo First Hospital, Institute of Hematology of Ningbo First Hospital, Ningbo, China
| | - Zhijuan Xu
- Haematology Department of Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo, China
| | - Guifang Ouyang
- Haematology Department of Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo, China
| |
Collapse
|
12
|
Roloff GW, Drazer MW, Godley LA. Inherited Susceptibility to Hematopoietic Malignancies in the Era of Precision Oncology. JCO Precis Oncol 2022; 5:107-122. [PMID: 34994594 DOI: 10.1200/po.20.00387] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
As germline predisposition to hematopoietic malignancies has gained increased recognition and attention in the field of oncology, it is important for clinicians to use a systematic framework for the identification, management, and surveillance of patients with hereditary hematopoietic malignancies (HHMs). In this article, we discuss strategies for identifying individuals who warrant diagnostic evaluation and describe considerations pertaining to molecular testing. Although a paucity of prospective data is available to guide clinical monitoring of individuals harboring pathogenic variants, we provide recommendations for clinical surveillance based on consensus opinion and highlight current advances regarding the risk of progression to overt malignancy in HHM variant carriers. We also discuss the prognosis of HHMs and considerations surrounding the utility of allogeneic stem-cell transplantation in these individuals. We close with an overview of contemporary issues at the intersection of HHMs and precision oncology.
Collapse
Affiliation(s)
- Gregory W Roloff
- Department of Medicine, Loyola University Medical Center, Maywood, IL
| | - Michael W Drazer
- Section of Hematology/Oncology, Department of Medicine and the Department of Human Genetics, the University of Chicago, Chicago, IL
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine and the Department of Human Genetics, the University of Chicago, Chicago, IL
| |
Collapse
|
13
|
AML with germline DDX41 variants is a clinicopathologically distinct entity with an indolent clinical course and favorable outcome. Leukemia 2021; 36:664-674. [PMID: 34671111 DOI: 10.1038/s41375-021-01404-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 12/17/2022]
Abstract
Germline DDX41 variants in myeloid neoplasms (MNs) are not uncommon, and we explored the prevalence and characterized the clinical and pathologic features in a cohort of 3132 unrelated adult MN patients. By targeted next-generation sequencing, we identified 28 patients (20 men and 8 women) with pathogenic germline DDX41 variants who developed acute myeloid leukemia (AML), in which only 3 (11%) had a family history (FH) of MNs. A subacute clinical course of cytopenia (mean duration of 11.2 months, range 0-72 months) prior to the initial AML diagnosis was accompanied by a low blast count (median at 30%, range 20-70%) in hypocellular marrows (93% of all patients), in vast contrast to the typical proliferative subtypes of AML in the elderly. Most patients had a normal karyotype (75%) and acquired a second DDX41 variant (69%). A favorable overall survival (OS) was observed in comparison to that of common subtypes of AML with wild-type DDX41 in age-matched patients. Our study demonstrated that the frequent germline pathogenic DDX41 variants characterized a clinically distinct AML entity. Features characteristic of DDX41-mutated AML include male predominance, often lack of FH, indolent course, low proliferative potential, frequent somatic DDX41 variants, and a favorable OS.
Collapse
|
14
|
Tawana K, Brown AL, Churpek JE. Integrating germline variant assessment into routine clinical practice for myelodysplastic syndrome and acute myeloid leukaemia: current strategies and challenges. Br J Haematol 2021; 196:1293-1310. [PMID: 34658019 DOI: 10.1111/bjh.17855] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/24/2021] [Accepted: 09/12/2021] [Indexed: 12/28/2022]
Abstract
Over the last decade, the field of hereditary haematological malignancy syndromes (HHMSs) has gained increasing recognition among clinicians and scientists worldwide. Germline mutations now account for almost 10% of adult and paediatric myelodysplasia/acute myeloid leukaemia (MDS/AML). As our ability to diagnose HHMSs has improved, we are now faced with the challenges of integrating these advances into routine clinical practice for patients with MDS/AML and how to optimise management and surveillance of patients and asymptomatic carriers. Discoveries of novel syndromes combined with clinical, genetic and epigenetic profiling of tumour samples, have highlighted unique patterns of disease evolution across HHMSs. Despite these advances, causative lesions are detected in less than half of familial cases and evidence-based guidelines are often lacking, suggesting there is much still to learn. Future research efforts are needed to sustain current momentum within the field, led not only by advancing genetic technology but essential collaboration between clinical and academic communities.
Collapse
Affiliation(s)
- Kiran Tawana
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| | - Anna L Brown
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia.,Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Jane E Churpek
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, School of Medicine and Public Health, The University of Wisconsin, Madison, WI, USA
| |
Collapse
|
15
|
Genetic features and clinical outcomes of patients with isolated and comutated DDX41-mutated myeloid neoplasms. Blood Adv 2021; 6:528-532. [PMID: 34644397 PMCID: PMC8791578 DOI: 10.1182/bloodadvances.2021005738] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/13/2021] [Indexed: 12/04/2022] Open
Abstract
Isolated and comutated DDX41 myeloid neoplasms have different characteristics. DDX41-mutated AML has a relatively favorable outcome comparable to core binding factor AML.
DDX41 mutations (germline and somatic) are associated with late onset myelodysplastic syndromes/acute myeloid leukemia (MDS/AML). Myeloid neoplasms (MN) with germline predisposition was identified as a distinct category in the 2016 WHO classification revision, including MN with germline DDX41 mutation. We retrospectively analyzed the molecular findings and clinical characteristics of thirty-three DDX41-mutated (mDDX41) patients at our institution. We identified 14 distinct pathogenic DDX41 variants in 32 patients and 8 DDX41 variants of unknown significance (VUS) in 9 patients. Five (16%) patients had a second DDX41 somatic mutation p.R525H and 13 (40%) had at least one additional oncogenic co-mutation in other genes. The median age at the time of diagnosis was 66 years, with male predominance (72%) and the majority of patients had normal cytogenetics (91%). Two-year overall survival (OS) was 86% and 6 (21%) MDS/AML patients with relatively preserved hematopoietic function were observed without further intervention. In comparison to AML patients with prognostically more favorable subtypes [t(8;21), n=27 and inv(16), n=40], mDDX41 patients in our cohort showed similarly favorable OS. Our study highlights that mDDX41-MN patients often have an indolent course and mDDX41-AML has comparable OS to favorable-risk AML.
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Recognition of hereditary hematopoietic malignancies impacts patient management as well as health surveillance strategies for the patient and relatives who share the causative DNA variant. In this review, barriers to the diagnosis and management of patients are outlined. RECENT FINDINGS Increasingly, individuals are being recognized as having germline predisposition to hematopoietic malignancies. Clinical testing for these syndromes is difficult for most clinicians given the need to send true germline samples and the lack of standardization in the field with regard to which genes are covered and the types of DNA changes detected. Additional barriers such as insurance coverage, especially for older individuals, and access to clinical experts need to be overcome in the future. SUMMARY New research addressing whether use of hematopoietic stem cells with deleterious variants are permissive to transplantation; effective means of delivering genetic counseling and results disclosure to decrease the psychological impact of these diagnoses; and a comprehensive list of all predisposition genes will advance our ability to provide the best treatment possible for our patients and facilitate strategies to maintain excellent health throughout their lifetimes and for members of younger generations. VIDEO ABSTRACT Submitted, http://links.lww.com/COH/A22.
Collapse
|
17
|
Feurstein S, Drazer M, Godley LA. Germline predisposition to haematopoietic malignancies. Hum Mol Genet 2021; 30:R225-R235. [PMID: 34100074 DOI: 10.1093/hmg/ddab141] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Once thought to be exceedingly rare, the advent of next-generation sequencing has revealed a plethora of germline predisposition disorders that confer risk for haematopoietic malignancies (HMs). These syndromes are now recognized to be much more common than previously thought. The recognition of a germline susceptibility risk allele in an individual impacts the clinical management and health surveillance strategies in the index patient and relatives who share the causative DNA variant. Challenges to accurate clinical testing include a lack of familiarity in many health care providers, the requirement for DNA samples that reasonably approximate the germline state, and a lack of standardization among diagnostic platforms as to which genes are sequenced and their capabilities in detecting the full range of variant types that confer risk. Current knowledge gaps include a comprehensive understanding of all predisposition genes; whether scenarios exist in which an allogeneic stem cell transplant using donor haematopoietic stem cells with deleterious variants is permissive; and effective means of delivering genetic counseling and results disclosure for these conditions. We are hopeful that comprehensive germline genetic testing, universal germline testing for all patients with an HM, universal germline testing for allogeneic haematopoietic stem cell donors, and the development of preventive strategies to delay or even prevent malignancies will be available in the near future. These factors will likely contribute to improved health outcomes for at-risk individuals and their family members.
Collapse
Affiliation(s)
- Simone Feurstein
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Michael Drazer
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL.,Department of Human Genetics, The University of Chicago, Chicago, IL
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL.,Department of Human Genetics, The University of Chicago, Chicago, IL
| |
Collapse
|
18
|
Fenwarth L, Duployez N, Marceau-Renaut A, Chahla WA, Ducassou S, Gandemer V, Pasquet M, Leblanc T, Schneider P, Domenech C, Saultier P, Leverger G, Lapillonne H, Preudhomme C, Petit A. Germline pathogenic variants in transcription factors predisposing to pediatric acute myeloid leukemia: results from the French ELAM02 trial. Haematologica 2021; 106:908-912. [PMID: 32554555 PMCID: PMC7928013 DOI: 10.3324/haematol.2020.248872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Indexed: 12/29/2022] Open
Affiliation(s)
- Laurène Fenwarth
- Laboratory of Hematology, CHU Lille, INSERM UMR-S 1277 - 9020 CNRS, Lille
| | - Nicolas Duployez
- Laboratory of Hematology, CHU Lille, INSERM UMR-S 1277 - 9020 CNRS, Lille
| | | | | | - Stéphane Ducassou
- Pediatric Hematology and Oncology Department, CHU Bordeaux, Bordeaux
| | | | - Marlène Pasquet
- Pediatric Hematology and Immunology Department, CHU Toulouse, Toulouse
| | - Thierry Leblanc
- Pediatric Hematology Department, AP-HP Robert Debré Hospital, Paris
| | | | - Carine Domenech
- Institute of Hematology and Pediatric Oncology, Lyon 1 University, Hospices Civils de Lyon, Lyon
| | - Paul Saultier
- Department of Pediatric Hematology and Oncology, Timone Enfants Hospital, APHM and Aix-Marseille University, Marseille
| | - Guy Leverger
- Pediatric Hematology and Oncology Department, Armand Trousseau Hospital, AP-HP, Sorbonne University, UMRS_938, CONECTAML, Paris
| | - Hélène Lapillonne
- Laboratory of Hematology, Armand Trousseau Hospital, Sorbonne University, UMRS_938, CONECT-AML, Paris, France
| | - Claude Preudhomme
- Laboratory of Hematology, CHU Lille, INSERM UMR-S 1277 - 9020 CNRS, Lille
| | - Arnaud Petit
- Pediatric Hematology and Oncology Department, Armand Trousseau Hospital, AP-HP, Sorbonne University, UMRS_938, CONECTAML, Paris
| |
Collapse
|
19
|
Pegliasco J, Schmaltz-Panneau B, Martin JE, Chraibi S, Khalife-Hachem S, Salviat F, Pasquier F, Willekens C, Lopez M, Ben-Ali A, Bera O, Caron O, Castilla-Llorent C, Cotteret S, Bourdin C, Saada V, Auger N, de Botton S, Vainchenker W, Fuseau P, Helias P, Benabdelali R, Marzac C, Meniane JC, Plo I, Bellanné-Chantelot C, Micol JB. ATG2B/GSKIP in de novo acute myeloid leukemia (AML): high prevalence of germline predisposition in French West Indies. Leuk Lymphoma 2021; 62:1770-1773. [PMID: 33554699 DOI: 10.1080/10428194.2021.1881508] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jean Pegliasco
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | - Jean-Edouard Martin
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Samy Chraibi
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Sabine Khalife-Hachem
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Flore Salviat
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Florence Pasquier
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Christophe Willekens
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Maureen Lopez
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Adel Ben-Ali
- Clinique Le Moulin de Viry, Viry-Châtillon, France
| | - Odile Bera
- Hospital Pierre Zobda-Quitman, Fort-de-France Bay, France
| | - Olivier Caron
- Département de médecine oncologique, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | - Sophie Cotteret
- Département de Biologie et Pathologie Médicales, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Carole Bourdin
- Hospital Pierre Zobda-Quitman, Fort-de-France Bay, France
| | - Veronique Saada
- Département de Biologie et Pathologie Médicales, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Nathalie Auger
- Département de Biologie et Pathologie Médicales, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Stephane de Botton
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | - Pascal Fuseau
- Hospital Pierre Zobda-Quitman, Fort-de-France Bay, France
| | | | | | - Christophe Marzac
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Département de Biologie et Pathologie Médicales, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | - Isabelle Plo
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Christine Bellanné-Chantelot
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Department of Genetics, Pitié-Salpêtrière hospital, Sorbonne University, Paris, France
| | - Jean-Baptiste Micol
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
20
|
Bernardi S, Farina M. Exosomes and Extracellular Vesicles in Myeloid Neoplasia: The Multiple and Complex Roles Played by These " Magic Bullets". BIOLOGY 2021; 10:biology10020105. [PMID: 33540594 PMCID: PMC7912829 DOI: 10.3390/biology10020105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Extracellular vesicles (EVs) are released by the majority of cell types and can be isolated from both cell cultures and body fluids. They are involved in cell-to-cell communication and may shuttle different messages (RNA, DNA, and proteins). These messages are known to influence the microenvironment of cells and their behavior. In recent years, some evidence about the involvement of EVs and exosomes, an EV subgroup, in immunomodulation, the transfer of disease markers, and the treatment of myeloid malignancies have been reported. Little is known about these vesicles in this particular setting of hematologic neoplasia; here, we summarize and critically review the available results, aiming to encourage further investigations. Abstract Extracellular vesicles (exosomes, in particular) are essential in multicellular organisms because they mediate cell-to-cell communication via the transfer of secreted molecules. They are able to shuttle different cargo, from nucleic acids to proteins. The role of exosomes has been widely investigated in solid tumors, which gave us surprising results about their potential involvement in pathogenesis and created an opening for liquid biopsies. Less is known about exosomes in oncohematology, particularly concerning the malignancies deriving from myeloid lineage. In this review, we aim to present an overview of immunomodulation and the microenvironment alteration mediated by exosomes released by malicious myeloid cells. Afterwards, we review the studies reporting the use of exosomes as disease biomarkers and their influence in response to treatment, together with the recent experiences that have focused on the use of exosomes as therapeutic tools. The further development of new technologies and the increased knowledge of biological (exosomes) and clinical (myeloid neoplasia) aspects are expected to change the future approaches to these malignancies.
Collapse
Affiliation(s)
- Simona Bernardi
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- Correspondence: or ; Tel.: +39-0303998464
| | - Mirko Farina
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| |
Collapse
|
21
|
Roloff GW, Godley LA, Drazer MW. Assessment of technical heterogeneity among diagnostic tests to detect germline risk variants for hematopoietic malignancies. Genet Med 2021; 23:211-214. [PMID: 32807974 PMCID: PMC9299956 DOI: 10.1038/s41436-020-0934-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To determine the degree of testing consistency among commercially available diagnostic assays for hereditary hematopoietic malignancies (HHMs). METHODS Next-generation sequencing assays designed for the diagnosis of HHMs were studied to determine which genes were sequenced, their ability to detect variant types relevant for HHMs, and clinical-grade characteristics such as price, turnaround time, and tissue types accepted. RESULTS Commercial assays varied in price (USD 250-4702), number of genes sequenced (12-73), and average turnaround time (14-42 days). A number of nongermline tissue types were accepted despite the tests being designed for germline diagnostic purposes. Multiple genes with well-characterized roles in HHM pathogenesis were omitted from more than one-third of panels intended for the evaluation of HHMs. Only 4 of 82 genes were consistently covered across all HHM diagnostic panels. The assays were highly variable in their sensitivity for structural alterations relevant to HHMs, such as copy-number variants. CONCLUSION A high degree of diagnostic heterogeneity exists among commercially available HHM diagnostic assays. Many of these assays are incapable of detecting the full spectrum of HHM-associated variants, leaving patients vulnerable to the consequences of underdiagnosis, missed opportunities for screening, and the potential for donor-derived malignancies.
Collapse
Affiliation(s)
- Gregory W Roloff
- Department of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Michael W Drazer
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago Comprehensive Cancer Center, Chicago, IL, USA.
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
22
|
Strategy for identification of a potential inherited leukemia predisposition in a 299 patient’s cohort with tumor-only sequencing data. Leuk Res 2020; 95:106386. [DOI: 10.1016/j.leukres.2020.106386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/18/2020] [Accepted: 05/18/2020] [Indexed: 11/19/2022]
|
23
|
Xiao X, He X, Li Q, Zhang W, Zhu H, Yang W, Li Y, Geng L, Liu H, Li L, Wang H, Fu R, Zhao M, Chen Z, Shao Z. Single-Nucleotide Polymorphism Array Technique Generating Valuable Risk-Stratification Information for Patients With Myelodysplastic Syndromes. Front Oncol 2020; 10:962. [PMID: 32733790 PMCID: PMC7358551 DOI: 10.3389/fonc.2020.00962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/15/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Chromosomal abnormalities play an important role in the diagnosis and prognosis of patients with myelodysplastic syndromes (MDSs). The single-nucleotide polymorphism array (SNP-A) technique has gained popularity due to its improved resolution compared to that of metaphase cytogenetic (MC) analysis. Methods: A total of 376 individuals were recruited from two medical centers in China, including 350 patients and 26 healthy individuals. Among these patients, 200 were diagnosed with de novo MDS, 25 with myeloproliferative neoplasm (MPN), 63 with primary acute myeloid leukemia (AML), and 62 with idiopathic cytopenia of undetermined significance (ICUS). We evaluated the significance of abnormal chromosomes detected by SNP-A in the diagnosis and prognosis of MDS-related disorders. Results: (1) When certain chromosomal abnormalities could not be detected by conventional MC methods, these abnormalities could be detected more efficiently by the SNP-A method. With SNP-A, the detection rates of submicroscopic or cryptic aberrations in the MDS, MPN, and AML patients with normal MC findings were 32.8, 30.8, and 30%, respectively. (2) The chromosomal abnormalities detected by SNP-A had a very important value for the prognosis of patients with MDSs, especially in the low-risk group. The survival of patients with abnormal chromosomes detected by SNP-A was significantly lower than that of patients with no detected chromosomal abnormalities; this difference was observed in overall survival (OS) (P = 0.001) and progression-free survival (PFS) [24 months vs. not reach (NR); P = 0.008]. The patients with multiple chromosomal abnormalities detected by SNP-A had an inferior prognosis, and SNP-A abnormalities (≥3 per patient) were found to be an independent predictor of poor prognosis in patients with MDSs [hazard ratio (HR) = 2.40, P = 0.002]. (3) Patients with ICUS may progress to myeloid malignancies, but most patients often maintain a stable ICUS status for many years without progression. An ICUS patient found to have an MDS-related karyotype would be rediagnosed with MDS. SNP-A can efficiently detect chromosomal abnormalities, which would be important for assessing the evolution of ICUS. In our study, 17 ICUS patients with SNP-A-detected abnormalities developed typical MDSs. Conclusions: SNP-A can help evaluate the prognosis of patients with MDSs and better assess the risk of disease progression for patients with ICUS.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Xiaoyuan He
- Department of Clinical Medicine, Nankai University School of Medicine, Tianjin, China
| | - Qing Li
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Wei Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Haibo Zhu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Weihong Yang
- Wuhan Kindstar Diagnostics Co./Kindstar Global Gene (Beijing) Technology, Inc., Wuhan, China
| | - Yuming Li
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Li Geng
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China.,Department of Clinical Medicine, Nankai University School of Medicine, Tianjin, China
| | - Zhong Chen
- Wuhan Kindstar Diagnostics Co./Kindstar Global Gene (Beijing) Technology, Inc., Wuhan, China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
24
|
Carraway HE, LaFramboise T. Myeloid neoplasms with germline predisposition: Practical considerations and complications in the search for new susceptibility loci. Best Pract Res Clin Haematol 2020; 33:101191. [PMID: 33038980 DOI: 10.1016/j.beha.2020.101191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
Abstract
Genomic research in hematological malignancies has focused far more prominently on somatic mutations than on germline variants. Although increasing numbers of germline variants are being identified, a substantial proportion of familial myeloid malignancies have no causal allele pinpointed. Here we review the biological, technological, and clinical challenges that stand in the way of the goal of establishing, implementing, and interpreting a comprehensive panel of germline variants for testing. Achieving this goal would inform care for large numbers of myeloid malignancy patients. Furthermore, knowledge of germline susceptibility variants and their corresponding genes will shed light on disease processes, potentially suggesting therapeutic strategies tailored to specific variants.
Collapse
Affiliation(s)
- Hetty E Carraway
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Thomas LaFramboise
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900, Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
25
|
Cumbo C, Tota G, Anelli L, Zagaria A, Specchia G, Albano F. TP53 in Myelodysplastic Syndromes: Recent Biological and Clinical Findings. Int J Mol Sci 2020; 21:E3432. [PMID: 32414002 PMCID: PMC7279310 DOI: 10.3390/ijms21103432] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
TP53 dysregulation plays a pivotal role in the molecular pathogenesis of myelodysplastic syndromes (MDS), identifying a subgroup of patients with peculiar features. In this review we report the recent biological and clinical findings of TP53-mutated MDS, focusing on the molecular pathways activation and on its impact on the cellular physiology. In MDS, TP53 mutational status is deeply associated with del(5q) syndrome and its dysregulation impacts on cell cycle, DNA repair and apoptosis inducing chromosomal instability and the clonal evolution of disease. TP53 defects influence adversely the MDS clinical outcome and the treatment response rate, thus new therapeutic approaches are being developed for these patients. TP53 allelic state characterization and the mutational burden evaluation can therefore predict prognosis and identify the subgroup of patients eligible for targeted therapy. For these reasons, in the era of precision medicine, the MDS diagnostic workup cannot do without the complete assessment of TP53 mutational profile.
Collapse
Affiliation(s)
| | | | | | | | | | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy; (C.C.); (G.T.); (L.A.); (A.Z.); (G.S.)
| |
Collapse
|
26
|
ETV6: A Candidate Gene for Predisposition to "Blend Pedigrees"? A Case Report from the NEXT-Famly Clinical Trial. Case Rep Hematol 2020; 2020:2795656. [PMID: 32148977 PMCID: PMC7057007 DOI: 10.1155/2020/2795656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background The identification of germline mutations in familial leukemia predisposition genes by next generation sequencing is of pivotal importance. Lately, some “blend pedigrees” characterized by both solid and hematologic malignancies have been described. Some genes were recognized as related to this double predisposition, while the involvement of others is still a matter of debate. ETV6 was associated with hematologic malignancies, in particular myeloid malignancies, and recently described as mutated also in oncologic patients. No clear evidences in its involvement in blend pedigrees are known. Case Presentation. We present our recent experience in the identification of an ETV6 was associated with hematologic malignancies, in particular myeloid malignancies, and recently described as mutated also in oncologic patients. No clear evidences in its involvement in blend pedigrees are known. ETV6 was associated with hematologic malignancies, in particular myeloid malignancies, and recently described as mutated also in oncologic patients. No clear evidences in its involvement in blend pedigrees are known. ETV6 was associated with hematologic malignancies, in particular myeloid malignancies, and recently described as mutated also in oncologic patients. No clear evidences in its involvement in blend pedigrees are known. Conclusion This evidence supports the involvement of ETV6 in the predisposition to both solid and hematologic neoplasia and the importance of the investigation of the noncoding regions of the genes as recently suggested by different expert groups.ETV6 was associated with hematologic malignancies, in particular myeloid malignancies, and recently described as mutated also in oncologic patients. No clear evidences in its involvement in blend pedigrees are known.
Collapse
|
27
|
Germline DDX41 mutations define a significant entity within adult MDS/AML patients. Blood 2020; 134:1441-1444. [PMID: 31484648 DOI: 10.1182/blood.2019000909] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022] Open
Abstract
Germline DDX41 mutations are involved in familial myelodysplastic syndromes (MDSs) and acute myeloid leukemias (AMLs). We analyzed the prevalence and characteristics of DDX41-related myeloid malignancies in an unselected cohort of 1385 patients with MDS or AML. Using targeted next-generation sequencing, we identified 28 different germline DDX41 variants in 43 unrelated patients, which we classified as causal (n = 21) or unknown significance (n = 7) variants. We focused on the 33 patients having causal variants, representing 2.4% of our cohort. The median age was 69 years; most patients were men (79%). Only 9 patients (27%) had a family history of hematological malignancy, and 15 (46%) had a personal history of cytopenia years before MDS/AML diagnosis. Most patients had a normal karyotype (85%), and the most frequent somatic alteration was a second DDX41 mutation (79%). High-risk DDX41 MDS/AML patients treated with intensive chemotherapy (n = 9) or azacitidine (n = 11) had an overall response rate of 100% or 73%, respectively, with a median overall survival of 5.2 years. Our study highlights that germline DDX41 mutations are relatively common in adult MDS/AML, often without known family history, arguing for systematic screening. Salient features of DDX41-related myeloid malignancies include male preponderance, frequent preexisting cytopenia, additional somatic DDX41 mutation, and relatively good outcome.
Collapse
|
28
|
Abstract
Modern management of acute myeloid leukaemia (AML) relies on the integration of phenotypic and genetic data to assign classification, establish prognosis, enhance monitoring and guide treatment. The prism through which we can now disperse a patient's leukaemia, interpret and apply our understanding has fundamentally changed since the completion of the first whole-genome sequencing (WGS) of an AML patient in 2008 and where possible, many clinicians would now prefer to delay treatment decisions until the karyotype and genetic status of a new patient is known. The success of global sequencing initiatives such as The Cancer Genome Atlas (TCGA) have brought us significantly closer to cataloguing the full spectrum of coding mutations involved in human malignancy. Indeed, genetic capability has raced ahead of our capacity to apply much of this knowledge into clinical practice and we are in the peculiar position of having routine access to genetic information on an individual patient's leukaemia that cannot be reliably interpreted or utilised. This is a measure of how rapid the progress has been, and this rate of change is likely to continue into the foreseeable future as research intensifies on the non-coding genome and the epigenome, as we scrutinise disease at a single cell level, and as initiatives like Beat AML and the Harmony Alliance progress. In this review, we will examine how interrogation of the coding genome is revolutionising our understanding of AML and improving our ability to underscore differences between paediatric and adult onset, sporadic and inherited forms of disease. We will look at how this knowledge is informing improvements in outcome prediction and the development of novel treatments, bringing us a step closer to personalised therapy for myeloid malignancy.
Collapse
Affiliation(s)
- Sarah Charrot
- Centre for Haemato-oncology, Barts Cancer Institute, QMUL, London, UK
| | - Hannah Armes
- Centre for Haemato-oncology, Barts Cancer Institute, QMUL, London, UK
| | - Ana Rio-Machin
- Centre for Haemato-oncology, Barts Cancer Institute, QMUL, London, UK
| | - Jude Fitzgibbon
- Centre for Haemato-oncology, Barts Cancer Institute, QMUL, London, UK
| |
Collapse
|
29
|
Li B, Zou D, Yang S, Ouyang G, Mu Q. Prognostic significance of U2AF1 mutations in myelodysplastic syndromes: a meta-analysis. J Int Med Res 2019; 48:300060519891013. [PMID: 31826693 PMCID: PMC7783272 DOI: 10.1177/0300060519891013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Although the effects of U2 small nuclear RNA auxiliary factor 1 gene (U2AF1) mutations on the outcomes of patients with myelodysplastic syndromes (MDS) have previously been investigated, their prognostic significance remains controversial. We performed a meta-analysis to investigate the impact of U2AF1 mutations on MDS progression. METHODS Two reviewers independently extracted information such as hazard ratios (HRs) and 95% confidential intervals (CIs) for overall survival (OS) and leukemia-free survival (LFS) as well as the number of surviving patients each year after diagnosis from the included studies. RESULTS Thirteen studies with a total of 3038 patients were included. The summary odds ratio (OR) for U2AF1 mutations with an OS of 5 years was 0.37, the summary HR for U2AF1 mutations in OS was 1.60, and the summary OR for an OS of 5 years in patients with U2AF1S34 and U2AF1Q157 was 3.68. There were no significant differences in leukemia-free survival or hypomethylating therapy response between patients with and without U2AF1 mutations. CONCLUSION U2AF1 mutations were associated with poor survival in MDS patients, and patients with U2AF1Q157 had a worse OS than those with U2AF1S34. Our findings suggest that MDS patients with U2AF1 mutations could benefit more from hypomethylation therapy.
Collapse
Affiliation(s)
- Bixia Li
- Ningbo University, Ningbo, P. R. China
| | - Duobing Zou
- Department of Hematology, Ningbo First Hospital, Ningbo, P. R. China
| | - Shujun Yang
- Department of Hematology, Ningbo First Hospital, Ningbo, P. R. China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, P. R. China
| | - Qitian Mu
- Department of Hematology, Ningbo First Hospital, Ningbo, P. R. China
| |
Collapse
|
30
|
Nordic Guidelines for Germline Predisposition to Myeloid Neoplasms in Adults: Recommendations for Genetic Diagnosis, Clinical Management and Follow-up. Hemasphere 2019; 3:e321. [PMID: 31976490 PMCID: PMC6924562 DOI: 10.1097/hs9.0000000000000321] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/10/2019] [Accepted: 10/24/2019] [Indexed: 12/18/2022] Open
Abstract
Myeloid neoplasms (MNs) with germline predisposition have recently been recognized as novel entities in the latest World Health Organization (WHO) classification for MNs. Individuals with MNs due to germline predisposition exhibit increased risk for the development of MNs, mainly acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Setting the diagnosis of MN with germline predisposition is of crucial clinical significance since it may tailor therapy, dictate the selection of donor for allogeneic hematopoietic stem cell transplantation (allo-HSCT), determine the conditioning regimen, enable relevant prophylactic measures and early intervention or contribute to avoid unnecessary or even harmful medication. Finally, it allows for genetic counseling and follow-up of at-risk family members. Identification of these patients in the clinical setting is challenging, as there is no consensus due to lack of evidence regarding the criteria defining the patients who should be tested for these conditions. In addition, even in cases with a strong suspicion of a MN with germline predisposition, no standard diagnostic algorithm is available. We present the first version of the Nordic recommendations for diagnostics, surveillance and management including considerations for allo-HSCT for patients and carriers of a germline mutation predisposing to the development of MNs.
Collapse
|
31
|
Palomo L, Ibáñez M, Abáigar M, Vázquez I, Álvarez S, Cabezón M, Tazón-Vega B, Rapado I, Fuster-Tormo F, Cervera J, Benito R, Larrayoz MJ, Cigudosa JC, Zamora L, Valcárcel D, Cedena MT, Acha P, Hernández-Sánchez JM, Fernández-Mercado M, Sanz G, Hernández-Rivas JM, Calasanz MJ, Solé F, Such E. Spanish Guidelines for the use of targeted deep sequencing in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol 2019; 188:605-622. [PMID: 31621063 PMCID: PMC7064979 DOI: 10.1111/bjh.16175] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 12/20/2022]
Abstract
The landscape of medical sequencing has rapidly changed with the evolution of next generation sequencing (NGS). These technologies have contributed to the molecular characterization of the myelodysplastic syndromes (MDS) and chronic myelomonocytic leukaemia (CMML), through the identification of recurrent gene mutations, which are present in >80% of patients. These mutations contribute to a better classification and risk stratification of the patients. Currently, clinical laboratories include NGS genomic analyses in their routine clinical practice, in an effort to personalize the diagnosis, prognosis and treatment of MDS and CMML. NGS technologies have reduced the cost of large-scale sequencing, but there are additional challenges involving the clinical validation of these technologies, as continuous advances are constantly being made. In this context, it is of major importance to standardize the generation, analysis, clinical interpretation and reporting of NGS data. To that end, the Spanish MDS Group (GESMD) has expanded the present set of guidelines, aiming to establish common quality standards for the adequate implementation of NGS and clinical interpretation of the results, hoping that this effort will ultimately contribute to the benefit of patients with myeloid malignancies.
Collapse
Affiliation(s)
- Laura Palomo
- Josep Carreras Leukaemia Research Institute, ICO Badalona-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Sadalona, Spain
| | - Mariam Ibáñez
- Department of Haematology, Hospital Universitari i Politècnic La Fe, València, Spain.,Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain.,Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, València, Spain
| | - María Abáigar
- Institute of Biomedical Research of Salamanca (IBSAL), Cancer Research Centre (IBMCC-CIC; Univ. of Salamanca-CSIC), Salamanca, Spain
| | - Iria Vázquez
- Haematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Sara Álvarez
- NIMGenetics, Genómica y Medicina, S.L., Madrid, Spain
| | - Marta Cabezón
- Haematology Service, ICO Badalona-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Bárbara Tazón-Vega
- Department of Haematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Inmaculada Rapado
- Haematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Haematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,Centro de investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Francisco Fuster-Tormo
- Josep Carreras Leukaemia Research Institute, ICO Badalona-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Sadalona, Spain
| | - José Cervera
- Department of Haematology, Hospital Universitari i Politècnic La Fe, València, Spain.,Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain.,Genetics Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Rocío Benito
- Institute of Biomedical Research of Salamanca (IBSAL), Cancer Research Centre (IBMCC-CIC; Univ. of Salamanca-CSIC), Salamanca, Spain
| | - María J Larrayoz
- Haematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | | | - Lurdes Zamora
- Haematology Service, ICO Badalona-Hospital Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - David Valcárcel
- Department of Haematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María T Cedena
- Haematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Haematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,Centro de investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Pamela Acha
- Josep Carreras Leukaemia Research Institute, ICO Badalona-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Sadalona, Spain
| | - Jesús M Hernández-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), Cancer Research Centre (IBMCC-CIC; Univ. of Salamanca-CSIC), Salamanca, Spain.,University of Salamanca (USAL), Salamanca, Spain
| | - Marta Fernández-Mercado
- Haematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain.,Advanced Genomics Laboratory, Centre for Applied Medical Research (CIMA), University of Navarra, Haemato-Oncology, Pamplona, Spain.,Biomedical Engineering Department, School of Engineering, University of Navarra, San Sebastian, Spain
| | - Guillermo Sanz
- Department of Haematology, Hospital Universitari i Politècnic La Fe, València, Spain.,Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Jesús M Hernández-Rivas
- Institute of Biomedical Research of Salamanca (IBSAL), Cancer Research Centre (IBMCC-CIC; Univ. of Salamanca-CSIC), Salamanca, Spain.,University of Salamanca (USAL), Salamanca, Spain.,Hospital Universitario de Salamanca, Salamanca, Spain
| | - María J Calasanz
- Haematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Francesc Solé
- Josep Carreras Leukaemia Research Institute, ICO Badalona-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Sadalona, Spain
| | - Esperanza Such
- Department of Haematology, Hospital Universitari i Politècnic La Fe, València, Spain.,Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain.,Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, València, Spain
| | | |
Collapse
|
32
|
Zhou J, Gou H, Zhang L, Wang X, Ye Y, Lu X, Ying B. ARID5B Genetic Polymorphisms Contribute to the Susceptibility and Prognosis of Male Acute Promyelocytic Leukemia. DNA Cell Biol 2019; 38:1374-1386. [PMID: 31599655 DOI: 10.1089/dna.2019.4926] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
This study was conducted using TagSNPs to systematically explore the relationship between ARID5B polymorphisms and the occurrence, clinical characterization, and prognosis of acute myeloid leukemia (AML). A total of 569 unrelated AML patients and 410 healthy individuals from West China were recruited, and ARID5B TagSNPs were genotyped using iMLDR® (improved multiplex ligation detection reaction). It was found that the association of ARID5B polymorphisms with AML was most significant in acute promyelocytic leukemia (APL), and exclusively in males, the mutant alleles of rs6415872, rs2393726, rs7073837, rs10821936, and rs7089424 were found to increase the risk of developing APL in men, the odds ratio (OR) were 1.36, 1.74, 1.45, 1.53, and 1.56 (all p < 0.05), respectively. Haplotype analysis revealed that haplotype [AACCG] increased the risk of male APL with an OR of 1.53 (95% confidence interval: 1.10-2.14, p = 0.012). Besides, there was a strong positive additive interaction between rs6415872 and rs10821936, rs7089424, respectively, and cases attributed to the interaction of rs6415872, rs10821936, and rs7089424 accounted for 100%. Furthermore, ARID5B single nucleotide polymorphisms were found associated with clinical features of AML, and rs6415872 was shown to be an independent prognosis factor in APL patients. Besides, dual luciferase report assay showed that rs6415872 may affect the binding activity of PPARG with ARID5B. ARID5B polymorphisms contribute to male APL risk, clinical feature, and prognosis, suggesting the importance of ARDI5B in AML pathogenesis and development, and the gender and subtype preference may prompt some specific mechanisms of ARID5B. Besides, ARID5B polymorphisms might be a potential prognosis biomarker.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Haimei Gou
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, P.R. China
| | - Li Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xinyi Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Yuanxin Ye
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaojun Lu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
33
|
When to obtain genomic data in acute myeloid leukemia (AML) and which mutations matter. Blood Adv 2019; 2:3070-3080. [PMID: 30425072 DOI: 10.1182/bloodadvances.2018020206] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022] Open
Abstract
Mutational profiling has fundamentally changed our approach to patients with acute myeloid leukemia (AML). Patients with AML are routinely profiled for the presence of mutations in FLT3, NPM1, CEBPA, and, more recently, TP53 In this chapter, we review the role of mutational profiling to help define disease biology in AML, particularly among patients with putatively intermediate-risk disease. We describe the body of evidence supporting the utility of mutational profiling when performed at the time of diagnosis (to identify prognostic and targetable mutations), at the time of complete remission (to assess minimal residual disease as a marker for relapse), and at the time of relapse (to identify therapeutic targets and eligibility for clinical trials). We further identify particular mutations that have been shown to affect prognosis across the established European LeukemiaNet risk categories and discuss which mutational events might be used to alter the approach to patient care at various time points during the disease course. We also review the evidence in support of molecular profiling for assessment of minimal/measurable residual disease and describe the current landscape of studies designed to validate this approach.
Collapse
|
34
|
D'Altri T, Schuster MB, Wenzel A, Porse BT. Heterozygous loss of Srp72 in mice is not associated with major hematological phenotypes. Eur J Haematol 2019; 103:319-328. [PMID: 31254415 DOI: 10.1111/ejh.13286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/19/2019] [Accepted: 06/24/2019] [Indexed: 02/01/2023]
Abstract
OBJECTIVES Familial cases of hematological malignancies are associated with germline mutations. In particular, heterozygous mutations of SRP72 correlate with the development of myelodysplasia and bone marrow aplasia in two families. The signal recognition particle 72 kDa protein (SRP72) is part of the SRP complex, responsible for targeting of proteins to the endoplasmic reticulum. The main objective of this study is to investigate the role of SRP72 in the hematopoietic system, thus explaining why a reduced dose could increase susceptibility to hematological malignancies. METHODS We developed an Srp72 null mouse model and characterized its hematopoietic system using flow cytometry, bone marrow transplantations, and gene expression analysis. RESULTS Heterozygous loss of Srp72 in mice is not associated with major changes in hematopoiesis, although causes mild reductions in blood and BM cellularity and minor changes within the stem/progenitor compartment. We did not observe any hematological disorder. Interestingly, gene expression analysis demonstrated that genes encoding secreted factors, including cytokines and receptors, were transcriptionally down-regulated in Srp72+/- animals. CONCLUSIONS The Srp72+/- mouse model only partially recapitulates the phenotype observed in families with inherited SRP72 lesions. Nonetheless, these results can provide mechanistic insights into why SRP72 mutations are associated with aplasia and myelodysplasia in humans.
Collapse
Affiliation(s)
- Teresa D'Altri
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel B Schuster
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Wenzel
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bo T Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Centre (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Turner SA, Shaver AC, Kovach AE, Oluwole OO, Mason EF. Myelodysplastic/myeloproliferative neoplasm with eosinophilia as a manifestation of Li Fraumeni Syndrome. Leuk Lymphoma 2019; 60:3312-3315. [DOI: 10.1080/10428194.2019.1630619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Scott A. Turner
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Aaron C. Shaver
- Division of Hematopathology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandra E. Kovach
- Division of Hematopathology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Olalekan O. Oluwole
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emily F. Mason
- Division of Hematopathology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
36
|
Fazio G, Massa V, Grioni A, Bystry V, Rigamonti S, Saitta C, Galbiati M, Rizzari C, Consarino C, Biondi A, Selicorni A, Cazzaniga G. First evidence of a paediatric patient with Cornelia de Lange syndrome with acute lymphoblastic leukaemia. J Clin Pathol 2019; 72:558-561. [PMID: 30948435 DOI: 10.1136/jclinpath-2019-205707] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/15/2019] [Accepted: 03/16/2019] [Indexed: 11/04/2022]
Abstract
Cornelia de Lange syndrome (CdLS) is a rare autosomal-dominant genetic disorder characterised by prenatal and postnatal growth and mental retardation, facial dysmorphism and upper limb abnormalities. Germline mutations of cohesin complex genes SMC1A, SMC3, RAD21 or their regulators NIPBL and HDAC8 have been identified in CdLS as well as somatic mutations in myeloid disorders. We describe the first case of a paediatric patient with CdLS with B-cell precursor Acute Lymphoblastic Leukaemia (ALL). The patient did not show any unusual cytogenetic abnormality, and he was enrolled into the high risk arm of AIEOP-BFM ALL2009 protocol because of slow early response, but 3 years after discontinuation, he experienced an ALL relapse. We identified a heterozygous mutation in exon 46 of NIPBL, causing frameshift and a premature stop codon (RNA-Targeted Next generation Sequencing Analysis). The analysis of the family indicated a de novo origin of this previously not reported deleterious variant. As for somatic cohesin mutations in acute myeloid leukaemia, also this ALL case was not affected by aneuploidy, thus suggesting a major impact of the non-canonical role of NIPBL in gene regulation. A potential biological role of NIPBL in leukaemia has still to be dissected.
Collapse
Affiliation(s)
- Grazia Fazio
- Centro di Ricerca Tettamanti, Clinica Pediatrica, Università di Milano, Bicocca, Monza, Italy
| | - Valentina Massa
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milano, Italy
| | - Andrea Grioni
- Centro di Ricerca Tettamanti, Clinica Pediatrica, Università di Milano, Bicocca, Monza, Italy.,Central European Institute of Technology, Masarykova Univerzita, Brno, Czech Republic
| | - Vojtech Bystry
- Central European Institute of Technology, Masarykova Univerzita, Brno, Czech Republic
| | - Silvia Rigamonti
- Centro di Ricerca Tettamanti, Clinica Pediatrica, Università di Milano, Bicocca, Monza, Italy.,Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milano, Italy
| | - Claudia Saitta
- Centro di Ricerca Tettamanti, Clinica Pediatrica, Università di Milano, Bicocca, Monza, Italy
| | - Marta Galbiati
- Centro di Ricerca Tettamanti, Clinica Pediatrica, Università di Milano, Bicocca, Monza, Italy
| | - Carmelo Rizzari
- Pediatric Department, Monza Brianza per il Bambino e la sua Mamma (MBBM) Foundation, Monza, Italy
| | - Caterina Consarino
- Ematologia ed Oncologia Pediatrica, Presidio Ospedaliero Ciaccio-De Lellis, Catanzaro, Italy
| | - Andrea Biondi
- Centro di Ricerca Tettamanti, Clinica Pediatrica, Università di Milano, Bicocca, Monza, Italy.,Pediatric Department, Monza Brianza per il Bambino e la sua Mamma (MBBM) Foundation, Monza, Italy
| | - Angelo Selicorni
- Department of Pediatrics, Presidio S. Fermo, ASST Lariana, Como, Italy
| | - Giovanni Cazzaniga
- Centro di Ricerca Tettamanti, Clinica Pediatrica, Università di Milano, Bicocca, Monza, Italy .,Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, Monza, Italy
| |
Collapse
|
37
|
Zhao X, Yin H, Li N, Zhu Y, Shen W, Qian S, He G, Li J, Wang X. An Integrated Regulatory Network Based on Comprehensive Analysis of mRNA Expression, Gene Methylation and Expression of Long Non-coding RNAs (lncRNAs) in Myelodysplastic Syndromes. Front Oncol 2019; 9:200. [PMID: 30984623 PMCID: PMC6450213 DOI: 10.3389/fonc.2019.00200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/08/2019] [Indexed: 12/12/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of disorders characterized by ineffective hematopoiesis, defective differentiation of hematopoietic precursors, and expansion of the abnormal clones. The prevalence of MDS has raised great concerns worldwide, but its pathogenetic mechanisms remain elusive. To provide insights on novel biomarkers for the diagnosis and therapy of MDS, we performed high-throughput genome-wide mRNA expression profiling, DNA methylation analysis, and long non-coding RNAs (lncRNA) analysis on bone marrows from four MDS patients and four age-matched healthy controls. We identified 1,937 differentially expressed genes (DEGs), 515 methylated genes, and 214 lncRNA that showed statistically significant differences. As the most significant module-related DEGs, TCL1A, PTGS2, and MME were revealed to be enriched in regulation of cell differentiation and cell death pathways. In addition, the GeneGo pathway maps identified by top DEGs were shown to converge on cancer, immunoregulation, apoptosis and regulation of actin cytoskeleton, most of which are known contributors in MDS etiology and pathogenesis. Notably, as potential biomarkers for diagnosis of MDS, four specific genes (ABAT, FADD, DAPP1, and SMPD3) were further subjected to detailed pathway analysis. Our integrative analysis on mRNA expression, gene methylation and lncRNAs profiling facilitates further understanding of the pathogenesis of MDS, and may promote the diagnosis and novel therapeutics for this disease.
Collapse
Affiliation(s)
- Xiaoli Zhao
- Key Laboratory of Hematology, Department of Hematology, Collaborative Innovation Center for Cancer Personalized Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China.,Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hua Yin
- Key Laboratory of Hematology, Department of Hematology, Collaborative Innovation Center for Cancer Personalized Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Nianyi Li
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Zhu
- Key Laboratory of Hematology, Department of Hematology, Collaborative Innovation Center for Cancer Personalized Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Wenyi Shen
- Key Laboratory of Hematology, Department of Hematology, Collaborative Innovation Center for Cancer Personalized Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Sixuan Qian
- Key Laboratory of Hematology, Department of Hematology, Collaborative Innovation Center for Cancer Personalized Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Guangsheng He
- Key Laboratory of Hematology, Department of Hematology, Collaborative Innovation Center for Cancer Personalized Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Jianyong Li
- Key Laboratory of Hematology, Department of Hematology, Collaborative Innovation Center for Cancer Personalized Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoqin Wang
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Churpek JE, Bresnick EH. Transcription factor mutations as a cause of familial myeloid neoplasms. J Clin Invest 2019; 129:476-488. [PMID: 30707109 DOI: 10.1172/jci120854] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The initiation and evolution of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are driven by genomic events that disrupt multiple genes controlling hematopoiesis. Human genetic studies have discovered germline mutations in single genes that instigate familial MDS/AML. The best understood of these genes encode transcription factors, such as GATA-2, RUNX1, ETV6, and C/EBPα, which establish and maintain genetic networks governing the genesis and function of blood stem and progenitor cells. Many questions remain unanswered regarding how genes and circuits within these networks function in physiology and disease and whether network integrity is exquisitely sensitive to or efficiently buffered from perturbations. In familial MDS/AML, mutations change the coding sequence of a gene to generate a mutant protein with altered activity or introduce frameshifts or stop codons or disrupt regulatory elements to alter protein expression. Each mutation has the potential to exert quantitatively and qualitatively distinct influences on networks. Consistent with this mechanistic diversity, disease onset is unpredictable and phenotypic variability can be considerable. Efforts to elucidate mechanisms and forge prognostic and therapeutic strategies must therefore contend with a spectrum of patient-specific leukemogenic scenarios. Here we illustrate mechanistic advances in our understanding of familial MDS/AML syndromes caused by germline mutations of hematopoietic transcription factors.
Collapse
Affiliation(s)
- Jane E Churpek
- Section of Hematology/Oncology and Center for Clinical Cancer Genetics, The University of Chicago, Chicago, Illinois, USA
| | - Emery H Bresnick
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
39
|
Roloff GW, Griffiths EA. When to obtain genomic data in acute myeloid leukemia (AML) and which mutations matter. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:35-44. [PMID: 30504289 PMCID: PMC6246019 DOI: 10.1182/asheducation-2018.1.35] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Mutational profiling has fundamentally changed our approach to patients with acute myeloid leukemia (AML). Patients with AML are routinely profiled for the presence of mutations in FLT3, NPM1, CEBPA, and, more recently, TP53 In this chapter, we review the role of mutational profiling to help define disease biology in AML, particularly among patients with putatively intermediate-risk disease. We describe the body of evidence supporting the utility of mutational profiling when performed at the time of diagnosis (to identify prognostic and targetable mutations), at the time of complete remission (to assess minimal residual disease as a marker for relapse), and at the time of relapse (to identify therapeutic targets and eligibility for clinical trials). We further identify particular mutations that have been shown to affect prognosis across the established European LeukemiaNet risk categories and discuss which mutational events might be used to alter the approach to patient care at various time points during the disease course. We also review the evidence in support of molecular profiling for assessment of minimal/measurable residual disease and describe the current landscape of studies designed to validate this approach.
Collapse
Affiliation(s)
| | - Elizabeth A. Griffiths
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and
- Roswell Park Cancer Institute, Buffalo, NY
| |
Collapse
|
40
|
Hereditary Myelodysplastic Syndrome and Acute Myeloid Leukemia: Diagnosis, Questions, and Controversies. Curr Hematol Malig Rep 2018; 13:426-434. [DOI: 10.1007/s11899-018-0473-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Germline duplication of ATG2B and GSKIP genes is not required for the familial myeloid malignancy syndrome associated with the duplication of chromosome 14q32. Leukemia 2018; 32:2720-2723. [PMID: 30087419 DOI: 10.1038/s41375-018-0231-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/11/2018] [Accepted: 07/18/2018] [Indexed: 11/08/2022]
|
42
|
Santini V. Society of Hematologic Oncology (SOHO) State of the Art Updates and Next Questions: Myelodysplastic Syndromes. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:495-500. [PMID: 29907542 DOI: 10.1016/j.clml.2018.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/17/2018] [Indexed: 10/16/2022]
Abstract
In the past few months, 2 main streams of research have dominated the panorama of myelodysplastic syndrome (MDS) investigations: deepening the insight into the pathogenic role, hierarchy, and prognostic effect of somatic mutations and, as a consequence, into the effect of inherited congenital predisposing conditions and the second, quite interlinked with the first, analyzing inflammation and innate immunity in patients with MDS. The research devoted to clarifying the mechanisms of action and mechanisms of resistance to hypomethylating agents has also advanced, mostly resulting from different approaches to the study of DNA methylation. Recent observations have reinforced support for targeted therapies for selected subgroups of MDS patients.
Collapse
Affiliation(s)
- Valeria Santini
- MDS Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy.
| |
Collapse
|