1
|
Joshi U, Bhetuwal U, Yadav SK, Budhathoki P, Gaire S, Sharma S, Niu C, Low SK, Neupane N, Agrawal V, Poudyal BS, Dhakal P. Survival Outcomes and Prognostic Factors in Therapy-Related Acute Myeloid Leukemia: A SEER Database Study, 2000-2020. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:e827-e834.e1. [PMID: 39095252 DOI: 10.1016/j.clml.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION With advances in therapeutics and longer survival across different cancer spectrums, the incidence of therapy-related acute myeloid leukemia (tAML) has continued to rise. This study aims to evaluate the trend of survival outcomes and their association with sociodemographic factors in tAML over the last 20 years. METHODS We identified tAML patients between 2000 and 2020 from the Surveillance, Epidemiology, and End Results database. Patients were divided into 4 age groups: 18-39, 40-59, 60-69, and >= 70 years, and 4 diagnostic periods: 2000-2005, 2006-2010, 2011-2015, and 2016-2020. Overall survival (OS) was compared using Kaplan Meier and log-rank methods. RESULTS The 1-year (and 5-year) OS in patients with tAML was 59.3% (33.7%), 48.2% (24.8%), 37.2% (11.1%), and 32.9% (5.5%) in age groups 18-39, 40-59, 60-69, and >=70 years, respectively. The 1-year (and 5-year) OS based on the year of diagnosis was 20.9% (13.2%), 36.8% (15.2%), 41.9% (13.88%), and 40.4% (not reached) for 2000-2005, 2006-2010, 2011-2015, and 2016-2020 respectively. Among the youngest cohort aged 18-39 years, 1-year OS was 35.7%, 57.7%, 66.7%, and 59.6%, respectively, in 4 diagnostic periods, whereas 1-year OS was 10.5%, 23.9%, 32.2%, and 36.9%, respectively, in the oldest cohort aged >=70 years. Age, year of diagnosis, and geographic location were independent prognostic markers of OS. CONCLUSION Our study demonstrates a significant improvement in the 1-year OS of tAML patients over the last decade, but the long-term prognosis remains dismal. Older patients continue to show improved survival in recent years with the addition of newer intensive and nonintensive options.
Collapse
Affiliation(s)
- Utsav Joshi
- Department of Hematology, Moffitt Cancer Center, Tampa, FL.
| | - Uttam Bhetuwal
- Department of Biostatistics, Biostatistics Epidemiology Research Design Informatics (BERDI) Core, Rhode Island Hospital, Providence, RI
| | - Sumeet K Yadav
- Department of Hospital Internal Medicine, Mayo Clinic Health System, Mankato, MN
| | | | - Suman Gaire
- Department of Internal Medicine, Mount Sinai Hospital, Chicago, IL
| | - Shiwani Sharma
- Department of Dermatology, National Academy of Medical Sciences, Kathmandu, Nepal
| | - Chengu Niu
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY
| | - Soon Khai Low
- Department of Hematology, Froedtert Hospital and Medical College of Wisconsin, Milwaukee, WI
| | - Niraj Neupane
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY
| | - Vishakha Agrawal
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Bishesh Sharma Poudyal
- Department of Hematology, Clinical Hematology and Bone Marrow Transplant Unit, Civil Service Hospital, Kathmandu, Nepal
| | - Prajwal Dhakal
- Division of Hematology, Oncology, Blood, and Marrow Transplantation, University of Iowa Health Care, Iowa City, IA
| |
Collapse
|
2
|
Menon A, Sukhanova M, Nocito KL, Gao J, Jennings LJ, Vormittag-Nocito ER. Detection and Interpretation of Clonal Hematopoiesis Variants during Routine Solid Tumor Next-Generation Sequencing: A Single-Institution Experience. J Mol Diagn 2024:S1525-1578(24)00213-7. [PMID: 39362468 DOI: 10.1016/j.jmoldx.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 10/05/2024] Open
Abstract
Clonal hematopoiesis (CH) and clonal cytopenia of undetermined significance (CCUS) are recently recognized diagnostic entities that serve as an independent risk factors for cardiovascular disease and myeloid malignancy. CH is an incidental finding, and evaluation of the incidence of CH/CCUS-associated mutations in solid tumor next-generation sequencing samples was undertaken to better understand the prevalence of mutations in this population. A retrospective review of clinical sequencing data for solid tumor malignancies diagnosed between February 2022 and April 2023 on next-generation sequencing data was performed. Cases were reviewed for variants in genes associated with CH/CCUS. Variant allele frequencies and other factors of the sequencing data were assessed for determining risk of CH/CCUS. A total of 2479 cases were evaluated during the study period. Of these, 29 cases demonstrated potential CH/CCUS-associated mutations, with a total of 33 variants identified. These were identified in a variety of tumor types, with gliomas being the most common. Significant cardiac histories were found in over half of cases identified, and few cases had abnormal blood counts. Detailed criteria for flagging variants as suspicious for CH and recommendations for these criteria as future guidelines for reporting are described. These variants are incidental findings that require more extensive follow-up or change in therapy management using a single institutional cohort.
Collapse
Affiliation(s)
- Adil Menon
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Madina Sukhanova
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kevin L Nocito
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Juehua Gao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Lawrence J Jennings
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Erica R Vormittag-Nocito
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
3
|
Kovach AE, Komova D, Itov A, Gaskova M, Kalinina I, Voronin K, Rumiantseva Y, Karachunskii A, Maschan M, Maschan A, Novichkova G, Olshanskaya Y, Bhojwani D, Raca G, Zerkalenkova E. Pediatric therapy-related hematologic neoplasms show enrichment for KMT2A rearrangement and lymphoblastic phenotype. Leuk Lymphoma 2024:1-13. [PMID: 39116419 DOI: 10.1080/10428194.2024.2376166] [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: 11/10/2023] [Revised: 06/25/2024] [Accepted: 06/29/2024] [Indexed: 08/10/2024]
Abstract
In children, therapy-related hematologic neoplasms (t-HN) are uncommon. Many are driven by genetic events independent of clonal hematopoiesis. We sought to understand the clinical and genetic factors of pediatric t-HN in a large independent cohort. Fifty-six t-HN were retrospectively identified. Chromosome microarray, next-generation and/or RNA sequencing were performed. Patients had primary hematologic, solid, or central nervous system tumors. t-HN included myeloid (t-MN) and lymphoblastic (t-ALL) phenotypes. Approximately half of the cases harbored KMTA2A rearrangement (KMT2Ar). Among t-HN without KMT2Ar, genetic drivers were heterogeneous, including diverse fusions or aneuploidy. Approximately 18% harbored 17p deletions and/or TP53 mutations. EFS/OS was not associated with t-HN lineage or KMT2Ar, but HSCT was associated with improved EFS and OS. We detail one of the largest cohorts to date of pediatric t-HN, confirming frequent KMT2Ar and t-ALL.
Collapse
Affiliation(s)
- Alexandra E Kovach
- Hematopathology, Division of Laboratory Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daria Komova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Albert Itov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Maria Gaskova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Irina Kalinina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Kirill Voronin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Yulia Rumiantseva
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexander Karachunskii
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Michael Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexey Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Galina Novichkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Yulia Olshanskaya
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Deepa Bhojwani
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Children's Hospital Los Angeles, CA, USA
| | - Gordana Raca
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Cytogenomics, Division of Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, CA, USA
| | - Elena Zerkalenkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| |
Collapse
|
4
|
Gołos A, Mikulski D, Grobelska-Kowalik M, Mądry K, Lis K, Sobas M, Ożańska A, Czemerska M, Hawrylecka D, Stelmach-Gołdyś A, Chromik K, Puła B, Sobczyk-Kruszelnicka M, Góra-Tybor J. Characteristics and outcomes of patients with lymphoma who developed therapy-related acute myeloid leukemia or myelodysplastic syndrome - a retrospective analysis of the Polish Adult Leukemia Group. Contemp Oncol (Pozn) 2024; 28:149-157. [PMID: 39421707 PMCID: PMC11480911 DOI: 10.5114/wo.2024.141727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 05/21/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Enhancing lymphoma outcomes increases the risk of therapy-related neoplasms such as acute myeloid leukemia (t-AML) and myelodysplastic syndrome (t-MDS). Material and methods Our study, conducted at seven Polish hematology centers between 2011 and 2018, explores clinical features, outcomes, and prognostic factors of t-AML and t-MDS arising after initial lymphoid neoplasms. Results The analysis included 57 patients of median age 65 with t-MDS (n = 38) and t-AML (n = 19). The median time to the onset of t-MDS/AML was 58.7 months. The median overall survival (OS) was 16.1 months. The presence of unfavorable cytogenetics and molecular risk factors (HR 2.88, 95% CI: 1.29-6.42, p = 0.009), hemoglobin level (HR 0.79, 95% CI: 0.65-0.95, p = 0.01) and platelets (HR 0.99, 95% CI: 0.99-0.9996, p = 0.03) were independent prognostic factors influencing OS. Therapy- related myelodysplastic syndrome/acute myeloid leukemia after lymphoma treatment is associated with a dismal prognosis mainly due to poor cytogenetic risk. Conclusions Anemia and thrombocytopenia may indicate more severe impairment of bone marrow function, resulting in further inferior treatment outcomes.
Collapse
Affiliation(s)
| | - Damian Mikulski
- Department Of Biostatistics And Translational Medicine, Medical University of Łódź, Łódź, Poland
| | - Monika Grobelska-Kowalik
- Hematooncology Department, Provincial Multi-Specialized Oncology and Trauma Center, Łódź, Poland
| | - Krzysztof Mądry
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Karol Lis
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marta Sobas
- Department and Clinic of Haematology, Blood Neoplasms, and Bone Marrow Transplantation, Wrocław Medical University, Wrocław, Poland
| | - Agnieszka Ożańska
- Department and Clinic of Haematology, Blood Neoplasms, and Bone Marrow Transplantation, Wrocław Medical University, Wrocław, Poland
| | | | | | | | - Karolina Chromik
- Department of Hematology and Bone Marrow Transplantation, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| | - Bartosz Puła
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Małgorzata Sobczyk-Kruszelnicka
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | |
Collapse
|
5
|
Travaglini S, Marinoni M, Visconte V, Guarnera L. Therapy-Related Myeloid Neoplasm: Biology and Mechanistic Aspects of Malignant Progression. Biomedicines 2024; 12:1054. [PMID: 38791019 PMCID: PMC11118122 DOI: 10.3390/biomedicines12051054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Therapy-related myeloid neoplasms (t-MN) arise after a documented history of chemo/radiotherapy as treatment for an unrelated condition and account for 10-20% of myelodysplastic syndromes and acute myeloid leukemia. T-MN are characterized by a specific genetic signature, aggressive features and dismal prognosis. The nomenclature and the subsets of these conditions have changed frequently over time, and despite the fact that, in the last classification, they lost their autonomous entity status and became disease qualifiers, the recognition of this feature remains of major importance. Furthermore, in recent years, extensive studies focusing on clonal hematopoiesis and germline variants shed light on the mechanisms of positive pressure underpinning the rise of driver gene mutations in t-MN. In this manuscript, we aim to review the evolution of defining criteria and characteristics of t-MN from a clinical and biological perspective, the advances in mechanistic aspects of malignant progression and the challenges in prevention and management.
Collapse
Affiliation(s)
- Serena Travaglini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Massimiliano Marinoni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Luca Guarnera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
6
|
Niscola P, Gianfelici V, Giovannini M, Piccioni D, Mazzone C, de Fabritiis P. Latest Insights and Therapeutic Advances in Myelodysplastic Neoplasms. Cancers (Basel) 2024; 16:1563. [PMID: 38672645 PMCID: PMC11048617 DOI: 10.3390/cancers16081563] [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: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic syndromes/neoplasms (MDSs) encompass a range of hematopoietic malignancies, commonly affecting elderly individuals. Molecular alterations in the hematopoietic stem cell compartment drive disease pathogenesis. Recent advancements in genomic profiling have provided valuable insights into the biological underpinnings of MDSs and have expanded therapeutic options, particularly for specific molecularly defined subgroups. This review highlights the diagnostic principles, classification updates, prognostic stratification systems, and novel treatments, which could inform future clinical trials and enhance the management of adult MDS patients, particularly for specific molecularly defined subgroups.
Collapse
Affiliation(s)
- Pasquale Niscola
- Division of Haematology, Sant’ Eugenio Hospital, 00144 Rome, Italy; (V.G.); (M.G.); (D.P.); (C.M.); (P.d.F.)
| | | | | | | | | | | |
Collapse
|
7
|
Mayeur S, Lhermitte B, Gantzer J, Molitor A, Stemmelen T, Meyer S, Kolmer A, Kurtz JE, Bahram S, Carapito R. Genomic profiling of a metastatic anaplastic melanocytic neuroectodermal tumor arising from a mature thymic teratoma as part of a mediastinal germ cell tumor. Cold Spring Harb Mol Case Stud 2023; 9:mcs.a006257. [PMID: 37160315 DOI: 10.1101/mcs.a006257] [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: 11/17/2022] [Accepted: 04/11/2023] [Indexed: 05/11/2023] Open
Abstract
Following chemotherapy, a mediastinal germ cell tumor can lead to a mature teratoma that is composed of tissues derived from all three germ layers. Although teratoma is usually curable, in rare cases it can give rise to various somatic tumors and exceptionally it undergoes melanocytic neuroectodermal tumor (MNT) transformation, a process that is not well-described. We report a patient with a postchemotherapy thymic teratoma associated with an MNT component who, 10 years later, additionally presented a vertebral metastasis corresponding to an anaplastic MNT. Using exome sequencing of the mature teratoma, the MNT and its metastatic vertebral anaplastic MNT components, we identified 19 somatic mutations shared by at least two components. Six mutations were common to all three components, and three of them were located in the known cancer-related genes KRAS (p.E63K), TP53 (p.P222X), and POLQ (p.S447P). Gene set enrichment analysis revealed that the melanoma tumorigenesis pathway was enriched in mutated genes including the four major driver genes KRAS, TP53, ERBB4, and KDR, indicating that these genes may be involved in the development of the anaplastic MNT transformation of the teratoma. To our knowledge, this is the first molecular study realized on MNT. Understanding the clinicopathological and molecular characteristics of these tumors is essential to better understand their development and to improve therapeutics.
Collapse
Affiliation(s)
- Sylvain Mayeur
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
- Department of Pathology, Strasbourg University Hospitals, Strasbourg 67200, France
| | - Benoit Lhermitte
- Department of Pathology, Strasbourg University Hospitals, Strasbourg 67200, France
| | - Justine Gantzer
- Pôle d'oncologie médico-chirurgicale et d'hématologie, ICANS-Europe, Strasbourg 67200, France
| | - Anne Molitor
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Tristan Stemmelen
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Sébastien Meyer
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Aline Kolmer
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France
| | - Jean-Emmanuel Kurtz
- Pôle d'oncologie médico-chirurgicale et d'hématologie, ICANS-Europe, Strasbourg 67200, France
| | - Seiamak Bahram
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France;
- Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg 67091, France
| | - Raphael Carapito
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg 67091, France;
- Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg 67091, France
| |
Collapse
|
8
|
Calvete O, Mestre J, Jerez A, Solé F. The Secondary Myelodysplastic Neoplasms (MDS) Jigsaw. Cancers (Basel) 2023; 15:1483. [PMID: 36900275 PMCID: PMC10000488 DOI: 10.3390/cancers15051483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/08/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
There is a great deal of controversy in the hematologic community regarding the classification of secondary myelodysplastic neoplasms (MDSs). Current classifications are based on the presence of genetic predisposition and MDS post-cytotoxic therapy (MDS-pCT) etiologies. However, since these risk factors are not exclusive for secondary MDSs and there are multiple overlapping scenarios, a comprehensive and definitive classification is yet to come. In addition, a sporadic MDS might arise after a primary tumor fulfills the diagnostic criteria of MDS-pCT without a causative cytotoxicity. In this review, we describe the triggering pieces of a secondary MDS jigsaw: previous cytotoxic therapy, germline predisposition and clonal hematopoiesis. Epidemiological and translational efforts are needed to put these pieces together and ascertain the real weight of each of these pieces in each MDS patient. Future classifications must contribute to understanding the role of secondary MDS jigsaw pieces in different concomitant or independent clinical scenarios associated with the primary tumor.
Collapse
Affiliation(s)
- Oriol Calvete
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Barcelona, Spain
| | - Julia Mestre
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Barcelona, Spain
| | - Andrés Jerez
- Experimental Hematology Unit, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, 08035 Barcelona, Spain
| | - Francesc Solé
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Barcelona, Spain
| |
Collapse
|
9
|
Volpe VO, Al Ali N, Chan O, Padron E, Sallman DA, Kuykendall A, Sweet K, Lancet JE, Komrokji RS. Splicing factor 3B subunit 1 (SF3B1) mutation in the context of therapy-related myelodysplastic syndromes. Br J Haematol 2022; 198:713-720. [PMID: 35751140 DOI: 10.1111/bjh.18319] [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: 03/28/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/27/2022]
Abstract
Splicing factor 3B subunit 1 (SF3B1) somatic mutation in the context of therapy-related myelodysplastic syndromes (t-MDS) has not been well defined. In a large cohort of patients with MDS, those with known SF3B1 somatic mutation were compared as de novo MDS (n = 289) and t-MDS with mutant SF3B1 (SF3B1mut ; n = 31). Baseline characteristics, concomitant mutations, and acute myeloid leukaemia (AML) transformation were similar between the two groups. The median overall survival (OS) of de novo MDS SF3B1mut was significantly longer compared to t-MDS SF3B1mut but not significantly different when adjusted for comorbidities. Comparing t-MDS wild-type SF3B1 (SF3B1WT ; n = 241) to t-MDS SF3B1mut (n = 31), complex cytogenetics were seen in 37.4% versus 10.3% (p = 0.009), tumour protein p53 (TP53) mutation was 36.1% versus 10% (p = 0.004), and AML transformation was 34.4% compared to 12.9% (p = 0.016) respectively. OS was significantly shorter in SF3B1WT versus SF3B1mut . When applying the International Working Group for Prognosis of MDS (IWG-PM) proposed SF3B1 criteria, OS was significantly shorter in SF3B1mut t-MDS compared to de novo MDS SF3B1mut with no significance in AML transformation. Survival was compared between t-MDS SF3B1mut who met the new proposed IWG-PM criteria to t-MDS SF3B1mut who did not meet criteria to survival of SF3B1WT t-MDS. OS was 53 versus 22 and 18 months respectively (p = 0.006). AML transformation was 0%, 26.7% and 32.3% (p = 0.021). Leukaemia-free survival was not reached among the three.
Collapse
Affiliation(s)
- Virginia O Volpe
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Najla Al Ali
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Onyee Chan
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Andrew Kuykendall
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Kendra Sweet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Jeffrey E Lancet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Rami S Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
10
|
Therapy-related myeloid neoplasms with different latencies: a detailed clinicopathologic analysis. Mod Pathol 2022; 35:625-631. [PMID: 34873304 DOI: 10.1038/s41379-021-00958-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 11/08/2022]
Abstract
Therapy-related myeloid neoplasm (t-MN) arising in patients with prior cytotoxic treatments is considered a distinct entity due to its unfavorable prognosis. Latencies between the initial cytotoxic therapy and the occurrence of t-MNs vary but usually fall between 1 and 10 years. t-MNs with unusually short or long latencies are not well characterized. It is unclear if they are biologically similar to the ones with ordinary latencies and should be kept in the t-MN entity. We compiled a cohort of t-MN cases including short (<1 year), ordinary (1-10 years), and extended (>10 years) latencies from two tertiary medical centers. Both the t-MNs with ordinary and extended latencies showed high likelihood of high-risk genetic abnormalities and demonstrated no significant survival differences. But the t-MNs with extended latencies were more likely associated with history of multiple cancers (p = 0.007) and were younger at the time of cytotoxic treatments (p < 0.001) when compared to the t-MNs with ordinary latencies. The t-MN with short latencies appears to be a very rare and highly heterogeneous group. In summary, the genetic composition appears similar in the t-MNs with ordinary and extended latencies. However, the association between the t-MN with extended latencies and history of multiple cancers raises a possibility that cancer predisposition may contribute to the accumulation of genetic abnormalities in these patients. Investigation into potential germline mutations in the t-MN patients with extended latencies may provide important information for related family members.
Collapse
|
11
|
Itonaga H, Kida M, Hamamura A, Uchida N, Ozawa Y, Fukuda T, Ueda Y, Kataoka K, Katayama Y, Ota S, Matsuoka KI, Kondo T, Eto T, Kanda J, Ichinohe T, Atsuta Y, Miyazaki Y, Ishiyama K. Outcome of therapy-related myelodysplastic syndrome and oligoblastic acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation: A propensity score matched analysis. Hematol Oncol 2022; 40:752-762. [PMID: 35299289 DOI: 10.1002/hon.2991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/26/2022] [Accepted: 03/16/2022] [Indexed: 11/11/2022]
Abstract
Therapy-related myelodysplastic syndromes (t-MDS) are generally progressive and associated with poorer outcomes than de novo MDS (d-MDS). To evaluate the outcome of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for t-MDS, we conducted a propensity score matched-pair analysis of patients with t-MDS and d-MDS using a nationwide database. A total of 178 patients with t-MDS underwent allo-HSCT between 2001 and 2018, and 178 out of 3,123 patients with d-MDS were selected. The probability of 3-year overall survival rate was 40.0% and 50.0% in the t-MDS and d-MDS groups, respectively (P=0.032). The 3-year transplant-related mortality was 30.9% and 19.0% in the t-MDS and d-MDS groups, respectively (P=0.005). The 3-year cumulative incidence of relapse was 32.8% and 33.0% in the t-MDS and d-MDS groups, respectively (P=0.983). A multivariate analysis identified four adverse factors for overall survival in the t-MDS group: age ≥55 years (hazard ratio [HR], 2.09; 95% CI, 1.11-3.94; P=0.023), the poor cytogenetic risk group (HR, 2.19; 95% CI, 1.40-4.19; P=0.019), performance status at allo-HSCT 2-4 (HR, 2.14; 95% CI, 1.19-3.86; P=0.011), and a shorter interval from diagnosis to transplantation (<8 months) (HR, 1.61; 95% CI, 1.00-2.57; P=0.048). The most frequent cause of transplant-related death was the infectious complications (21.6%) in t-MDS group and organ failure (12.5%) in d-MDS group. In conclusion, allo-HSCT potentially provides long-term remission in patients with t-MDS; however, further efforts to reduce transplant-related death are needed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hidehiro Itonaga
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan
| | - Michiko Kida
- Department of Hematology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Atsushi Hamamura
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Naoyuki Uchida
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Toranomon Hospital, Tokyo, Japan
| | - Yukiyasu Ozawa
- Department of Hematology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Aichi, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Yasunori Ueda
- Department of Hematology/Oncology and Transfusion and Hemapheresis Center, Kurashiki Central Hospital, Okayama, Japan
| | - Keisuke Kataoka
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuta Katayama
- Department of Hematology, Hiroshima Red Cross Hospital & Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Hokkaido, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Tadakazu Kondo
- Department of Hematology, Kyoto University Hospital, Kyoto, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - Junya Kanda
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Aichi, Japan.,Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Aichi, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan.,Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Ken Ishiyama
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Japan
| |
Collapse
|
12
|
Boquoi A, Banahan SM, Mohring A, Savickaite I, Strapatsas J, Hildebrandt B, Kobbe G, Gattermann N, Haas R, Schroeder T, Germing U, Fenk R. Therapy-related myeloid neoplasms following treatment for multiple myeloma-a single center analysis. Ann Hematol 2022; 101:1031-1038. [PMID: 35262868 PMCID: PMC8993729 DOI: 10.1007/s00277-022-04775-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022]
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) can be late complications following mutagenic treatment. Limited data is available on the outcome of patients developing therapy-related MDS and AML after treatment for multiple myeloma (MM). We identified 250 patients with therapy-associated MDS or AML in the Duesseldorf MDS registry. Of those, 50 patients were previously diagnosed with multiple myeloma (mm-MDS/AML). We compared them to patients with de novo MDS (n = 4862) and to patients with MDS following other underlying diseases (tMDS) (n = 200). mm-MDS patients and tMDS patients showed similar karyotypes and degrees of cytopenia. However, mm-MDS patients had significantly higher blast counts and more often belonged to the high-risk group according to the International Prognostic Scoring System (IPSS) (both p < 0.05). Although the rate of progression to AML was similar in mm-MDS and tMDS, both transformed significantly more often than de novo MDS (p < 0.05). Median overall survival of patients with mm-MDS (13 months; range: 1–99) and tMDS (13 months; range 0–160) was also similar yet significantly shorter than patients with de novo MDS (32 months; range 0–345 months; p < 0.05). Furthermore, survival of mm-MDS patients was not affected by myeloma activity. Despite significantly more high-risk disease and higher blast cell counts, myeloma-associated MDS-patients show features akin to other tMDS. Survival is similar to other tMDS and irrespective of myeloma remission status or transformation to AML. Thus, patient outcome is not determined by competing clones but rather by MDS governing the stem cell niche.
Collapse
Affiliation(s)
- A Boquoi
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany.
| | - S M Banahan
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - A Mohring
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - I Savickaite
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - J Strapatsas
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - B Hildebrandt
- Institute of Human Genetics, Heinrich-Heine-University, Duesseldorf, Germany
| | - G Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - N Gattermann
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - R Haas
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - T Schroeder
- Department of Hematology and Stem Cell Transplantation, University Medicine Essen, Essen, Germany
| | - U Germing
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - R Fenk
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| |
Collapse
|
13
|
Li G, Holly T, Kelly DR, Reddy V, Mikhail FM, Carroll AJ, Kutny MA. Therapy-related Myeloid Neoplasms in Children: A Single-institute Study. J Pediatr Hematol Oncol 2022; 44:e109-e113. [PMID: 33625084 DOI: 10.1097/mph.0000000000002097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/10/2021] [Indexed: 11/26/2022]
Abstract
Therapy-related myeloid neoplasm (t-MN) in the pediatric population is not well characterized. We studied 12 pediatric patients diagnosed with t-MN in our institution since 2006. The median age at the t-MN diagnoses was 14.8 years (range, 9 to 20 y). The primary malignancies included 9 solid tumors and 3 hematopoietic malignancies. Rhabdomyosarcoma (n=4) was the most common primary malignancy. Five of the 9 patients with solid tumors and all 3 patients with hematopoietic malignancies had primary neoplasms involving bone marrow. The median latency period was 5.2 years (range, 1.8 to 13.8 y). Thrombocytopenia was present in all patients at the t-MN diagnoses. Complete or partial monosomy of chromosome 5 or 7 were the 2 most common cytogenetic abnormalities. A quarter of patients demonstrated a genetic predisposition to t-MN: 1 with Li-Fraumeni syndrome with a germline TP53 R248Q mutation, 1 with Noonan syndrome with a somatic mutation (PTPN11 S502T), and 1 with a constitutive chromosomal translocation [t(X;9)(p22;q34)] and a germline TP53 L130V mutation. Outcomes remain poor. Two patients survived 3 and 5.1 years after hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Geling Li
- Department of Pathology and Laboratory Medicine, Children's of Alabama
- Departments of Pathology
| | - Taylor Holly
- Department of Pediatrics, Division of Hematology and Oncology
| | - David R Kelly
- Department of Pathology and Laboratory Medicine, Children's of Alabama
- Departments of Pathology
| | | | - Fady M Mikhail
- Genetics, University of Alabama at Birmingham, Birmingham, AL
| | | | - Matthew A Kutny
- Department of Pediatrics, Division of Hematology and Oncology
| |
Collapse
|
14
|
Pehalova L, Krejci D, Halamkova J, Smardova L, Snajdrova L, Dusek L. Significant current epidemiological trend: Haematological malignancies as subsequent primary tumours in cancer patients. Cancer Epidemiol 2021; 72:101929. [PMID: 33819838 DOI: 10.1016/j.canep.2021.101929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/09/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Numbers of patients who develop subsequent primary tumours have markedly increased recently. This study aimed to carry out a comprehensive analysis documenting the risk of incidence of subsequent haematological malignancies. METHODS The Czech National Cancer Registry was the main data source, containing records of 126,822 haematological malignancies diagnosed in the period 1977-2016. Subsequent haematological malignancies were identified according to IACR rules. Joinpoint regression was employed to assess the time trends. The risk of development of subsequent haematological malignancy was evaluated by the standardised incidence ratio. The Kaplan-Meier curves were used to assess the differences in survival. RESULTS Age-standardised incidence of subsequent haematological malignancies increased from 0.5 in 1977 to 9.1 in 2016. In 1992, there was a significant change in the trend: a sharp increase by 7.7 % annually was revealed thereafter. The risk of development of a haematological malignancy was approximately 1.5 times higher in persons with history of any cancer than in the general Czech population. Patients with haematological malignancies - mainly myelodysplastic syndromes, polycythaemia vera and non-Hodgkin lymphoma - were shown to be at the highest risk of developing a subsequent haematological malignancy. While the median survival following a first haematological malignancy was 2.3 years, it was only 1.1 years for subsequent haematological malignancies (p < 0.001). CONCLUSIONS Our study identified the highest-risk diagnoses in terms of development of subsequent haematological malignancy. The results might be useful to set up correctly follow-up procedures from which cancer patients could benefit.
Collapse
Affiliation(s)
- Lucie Pehalova
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic.
| | - Denisa Krejci
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| | - Jana Halamkova
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic; Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Medical Ethics, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Smardova
- Department of Internal Medicine, Haematology and Oncology, University Hospital Brno and Masaryk University, Brno, Czech Republic
| | - Lenka Snajdrova
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ladislav Dusek
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
15
|
Yarosh R, Roesler MA, Murray T, Cioc A, Hirsch B, Nguyen P, Warlick E, Poynter JN. Risk factors for de novo and therapy-related myelodysplastic syndromes (MDS). Cancer Causes Control 2021; 32:241-250. [PMID: 33392905 PMCID: PMC7878335 DOI: 10.1007/s10552-020-01378-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE Myelodysplastic syndromes (MDS) are classified as de novo and therapy-related (tMDS). We evaluated associations between MDS risk factors separately for de novo and tMDS. METHODS The study population included 346 de novo MDS cases, 37 tMDS cases and 682 population controls frequency matched by age and sex. Polytomous logistic regression was performed to calculate odds ratios (OR) and 95% confidence intervals (CI). RESULTS After adjustment, former smoking status (OR = 1.45, 95% CI: 1.10-1.93), personal history of autoimmune disease (OR = 1.34, 95% CI: 0.99-1.82) and exposure to benzene (OR = 1.48, 95% CI: 1.00-2.19) were associated with de novo MDS. Risk estimates for the associations between smoking, autoimmune disease, and benzene exposure were similar in magnitude but non-significant in tMDS cases. Among individuals with a previous diagnosis of cancer, de novo MDS cases and controls were more likely to have had a previous solid tumor, while tMDS cases more commonly had a previous hematologic malignancy. CONCLUSIONS We observed similar associations between smoking, history of autoimmune disease and benzene exposure in de novo and tMDS although estimates for tMDS were imprecise due to small sample sizes. Future analyses with larger sample sizes will be required to confirm whether environmental factors influence risk of tMDS.
Collapse
Affiliation(s)
- Rina Yarosh
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | | | - Thomas Murray
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | | | - Betsy Hirsch
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Phuong Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Erica Warlick
- Department of Medicine, Division of Hematology/Oncology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jenny N Poynter
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
16
|
Yuan X, Yu U, Chen S, Xu H, Yi M, Jiang X, Song J, Chen X, Chen S, Lin Z, Li C, Wen F, Liu S. Case Report: Myeloid Sarcoma Development During Treatment for B Cell Lymphoblastic Lymphoma in a Boy with KRAS/NRAS Gene Mutations. Onco Targets Ther 2021; 14:347-353. [PMID: 33469311 PMCID: PMC7812042 DOI: 10.2147/ott.s276912] [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: 09/27/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022] Open
Abstract
Here, we report a rare case of a 12-year-old boy who was initially diagnosed with B cell lymphoblastic lymphoma (BLBL) and developed myeloid sarcoma (MS) eight months after chemotherapy. Next-generation sequencing (NGS) showed mutations of KRAS and NRAS genes in both the bone marrow and lymph node. He presented an abnormal karyotype of 46, XY, -9, der (16) t (9; 16) (q13; q12), +mar. He received chemotherapy according to the South China Children's Leukemia Group 2016 protocol. Complete remission was achieved by the 15th day post-treatment. Eight months later and immediately prior to the start of maintenance therapy, the patient developed fever, skin nodules in both upper arms, and enlargement of bilateral testes. Pathological analysis of skin and testicular biopsies suggested the diagnosis of myeloid sarcoma (MS). Again, NGS examination showed mutations of KRAS and NRAS genes. The patient underwent haploidentical hematopoietic stem cell transplantation but unfortunately did not survive. The interval of eight-month interval between the initial disease onset and MS brings into question whether MS developed as part of the initial onset of disease or as a secondary tumor in association with chemotherapy. Thus, understanding the pathogenesis of MS involving abnormalities of lymphoid progenitors may assist in the prediction of prognosis and development of novel target therapies.
Collapse
Affiliation(s)
- Xiuli Yuan
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Uet Yu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Senmin Chen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Huanli Xu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Meng Yi
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Xianping Jiang
- Department of Pathology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Jianming Song
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Xiaowen Chen
- Institute for Medical Research, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Shiyang Chen
- Institute for Medical Research, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Zhenhu Lin
- Institute for Medical Research, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Changgang Li
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Feiqiu Wen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Sixi Liu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| |
Collapse
|
17
|
Chantadisai M, Kulkarni HR, Baum RP. Therapy-related myeloid neoplasm after peptide receptor radionuclide therapy (PRRT) in 1631 patients from our 20 years of experiences: prognostic parameters and overall survival. Eur J Nucl Med Mol Imaging 2020; 48:1390-1398. [PMID: 33247328 DOI: 10.1007/s00259-020-05127-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE To determine prognostic factors and overall survival (OS) in therapy-related myeloid neoplasm (t-MN) of patients after receiving peptide receptor radionuclide therapy (PRRT). METHODS All patients treated from February 1999 until September 2019 at our center who had bone marrow biopsy-proven t-MN after PRRT were included. Patient characteristics, laboratory results, and all tumor-directed therapies before t-MN diagnosis were collected. Cox regression analysis was performed to identify parameters associated with OS. Receiver operating characteristic (ROC) curve analysis was used to define cutoff values as well as sensitivity and specificity of the parameters. RESULTS Out of 1631 patients treated with PRRT, 30 patients developed t-MN comprising myelodysplastic syndrome (MDS) in 23 patients (77%) and acute myeloid leukemia (AML) in 7 patients (23%). The median OS of t-MN patients was 13 months (range 9.1-16.9 months): 6 months for AML and 15 months for the MDS subgroup, respectively. Higher platelet level was a significant prognostic parameter for longer OS (hazard ratio (HR): 0.99, P < 0.05). Using ROC analysis, the best cutoff value for thrombocyte count was 183.5 Gpt/L, resulting in a sensitivity of 92.3% and a specificity of 50%. Other factors, such as hemoglobin level, did not show a significant correlation with OS. CONCLUSION Even rarely occurred, the OS is gravely compromised in t-MN patients after PRRT, and even less in the AML subgroup (6 months). Higher platelet value was a significant prognostic parameter for longer OS in t-MN patients.
Collapse
Affiliation(s)
- M Chantadisai
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, 99437, Bad Berka, Germany. .,Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand.
| | - H R Kulkarni
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, 99437, Bad Berka, Germany
| | - R P Baum
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, 99437, Bad Berka, Germany.,Advanced Theranostics Center for Molecular Radiotherapy and Precision Oncology, ICPO Center of Excellence, CURANOSTICUM Wiesbaden-Frankfurt at DKD Helios Klinik, Wiesbaden, Germany
| |
Collapse
|
18
|
Brierley CK, Zabor EC, Komrokji RS, DeZern AE, Roboz GJ, Brunner AM, Stone RM, Sekeres MA, Steensma DP. Low participation rates and disparities in participation in interventional clinical trials for myelodysplastic syndromes. Cancer 2020; 126:4735-4743. [PMID: 32767690 DOI: 10.1002/cncr.33105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND The development of novel therapies for the myelodysplastic syndromes (MDS) is hampered by inadequate trial recruitment. Factors contributing to low trial accrual are incompletely understood. METHODS This study analyzed a pooled patient database from institutions of the US MDS Clinical Research Consortium to compare the characteristics of participants in interventional trials with those of patients who did not enroll in a trial. RESULTS Data were identified for 1919 patients with MDS, and 449 of these patients (23%) participated in an interventional clinical trial. The median age of all patients was 68 years, and 64% were male. Patients who participated in trials were significantly younger than nonparticipants (P = .014), and men were more likely to participate in a trial (71% of trial participants were male, whereas 61% of nonparticipants were; P < .001). Race and ethnicity were not associated with trial enrollment. Patients in more affluent ZIP codes had a higher participation rate (P < .001). Patients with intermediate- and high-risk disease according to the revised International Prognostic Scoring System were overrepresented (P = .004), and trial participants less frequently had treatment-related disease (P < .001). In multivariable analyses, participation in a clinical trial was associated with a reduced hazard of death (P = .004). Even at large referral centers, only a minority of patients with MDS enrolled in interventional trials. CONCLUSIONS Restrictive trial eligibility criteria that exclude patients with MDS on account of age, comorbidities, or a history of another cancer are limit enrollment of MDS patients to clinical trials. Gaining insight into the barriers to trial accrual may help investigators and study sponsors to design trials that will accrue more rapidly and augment treatment options for patients with MDS.
Collapse
Affiliation(s)
| | - Emily C Zabor
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Rami S Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Gail J Roboz
- Weill Cornell Medical College, New York, New York
| | - Andrew M Brunner
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard M Stone
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mikkael A Sekeres
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - David P Steensma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Albertini RJ, Kaden DA. Mutagenicity monitoring in humans: Global versus specific origin of mutations. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 786:108341. [PMID: 33339577 DOI: 10.1016/j.mrrev.2020.108341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 01/19/2023]
Abstract
An underappreciated aspect of human mutagenicity biomonitoring is tissue specificity reflected in different assays, especially those that measure events that can only occur in developing bone marrow (BM) cells. Reviewed here are 9 currently-employed human mutagenicity biomonitoring assays. Several assays measure chromosome-level events in circulating T-lymphocytes (T-cells), i.e., traditional analyses of aberrations, translocation studies involving chromosome painting and fluorescence in situ hybridization (FISH) and determinations of micronuclei (MN). Other T-cell assays measure gene mutations. i.e., hypoxanthine-guanine phosphoriboslytransferase (HPRT) and phosphoribosylinositol glycan class A (PIGA). In addition to the T-cell assays, also reviewed are those assays that measure events in peripheral blood cells that necessarily arose in BM cells, i.e., MN in reticulocytes; glycophorin A (GPA) gene mutations in red blood cells (RBCs), and PIGA gene mutations in RBC or granulocytes. This review considers only cell culture- or cytometry-based assays to describe endpoints measured, methods, optimal sampling times, and sample summaries of typical quantitative and qualitative results. However, to achieve its intended focus on the target cells where events occur, kinetics of the cells of peripheral blood that derive at some point from precursor cells are reviewed to identify body sites and tissues where the genotoxic events originate. Kinetics indicate that in normal adults, measured events in T-cells afford global assessments of in vivo mutagenicity but are not specific for BM effects. Therefore, an agent's capacity for inducing mutations in BM cells cannot be reliably inferred from T-cell assays as the magnitude of effect in BM, if any, is unknown. By contrast, chromosome or gene level mutations measured in RBCs/reticulocytes or granulocytes must originate in BM cells, i.e. in RBC or granulocyte precursors, thereby making them specific indicators for effects in BM. Assays of mutations arising directly in BM cells may quantitatively reflect the mutagenicity of potential leukemogenic agents.
Collapse
Affiliation(s)
- Richard J Albertini
- University of Vermont, 111 Colchester Avenue, Burlington, VT 05401, United States
| | - Debra A Kaden
- Ramboll US Consulting, Inc., 101 Federal Street, Suite 1900, Boston, MA 02110, United States.
| |
Collapse
|
20
|
Hwang SM. Classification of acute myeloid leukemia. Blood Res 2020; 55:S1-S4. [PMID: 32719169 PMCID: PMC7386892 DOI: 10.5045/br.2020.s001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/23/2020] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
The World Health Organization (WHO) Classification of Tumors of Hematopoietic and Lymphoid Tissues was revised in 2017 on the basis of recent high-throughput sequencing and gene expression data on hematologic malignancies. This review explores the current WHO classification of acute myeloid leukemia (AML) and related precursor neoplasms, highlighting the changes made in the current edition and focusing on the diagnosis of AML.
Collapse
Affiliation(s)
- Sang Mee Hwang
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
21
|
Tiruneh T, Enawgaw B, Shiferaw E. Genetic Pathway in the Pathogenesis of Therapy-Related Myeloid Neoplasms: A Literature Review. Oncol Ther 2020; 8:45-57. [PMID: 32700075 PMCID: PMC7360004 DOI: 10.1007/s40487-020-00111-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Therapy-related myeloid neoplasms are a life-threatening and often fatal complication, associated with poor prognosis outcomes and with high-risk unfavorable cytogenetic abnormalities including complex karyotype. They occur after the treatment of primary malignancies using chemotherapy and/or radiation therapy. Such therapy is not specific to cancer cells, and also damages the deoxyribonucleic acid (DNA) of normal cells, resulting in unbalanced and balanced translocations. There are eight genetic pathways, whose details are summarized in this review, depending on the cytogenetic abnormalities induced. This abnormality is the major contributor to the development of therapy-related myeloid neoplasms. The etiology of these neoplasms depends on the complex interaction between the nature and dose of the cytotoxic agent, the environment, and the presence of subsequent inherited mutations. This review aims to elaborate upon recent knowledge regarding the etiology, pathogenesis, and genetic pathways of therapy-related myeloid neoplasms. A deeper understanding of their etiology would aid physicians in more careful monitoring of patients during or after cytotoxic therapy for hematological malignancy. Ultimately, this knowledge could influence initial treatment strategies, with the aim of reducing both the incidence and serious complications of neoplasms. Therefore, early detection of DNA lesions is vital. The authors recommend that primary malignancy be treated with targeted therapy.
Collapse
Affiliation(s)
- Tegenaw Tiruneh
- Department Hematology and Immunohematology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia. .,School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Bamlaku Enawgaw
- School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Elias Shiferaw
- School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
22
|
Baek DW, Lee SJ, Sohn SK, Moon JH, Chae YS. Clinical Effects of Hypomethylating Agents in Patients with Newly Diagnosed Myelodysplastic Syndrome Who Received DNA-Damaging Chemotherapy for Metastatic Breast Cancer. J Breast Cancer 2020; 22:647-652. [PMID: 31897338 PMCID: PMC6933035 DOI: 10.4048/jbc.2019.22.e50] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/05/2019] [Indexed: 11/30/2022] Open
Abstract
The cumulative risk of therapy-related myelodysplastic syndrome (t-MDS) in breast cancer patients exposed to chemotherapy and/or radiotherapy is significantly high compared to that in other cancer patients. This report reviews the use of hypomethylating agents (HMAs) to treat a 57-year-old woman newly diagnosed with MDS during palliative chemotherapy for metastatic breast cancer. Over a period of 6 years, the patient received several DNA-damaging chemotherapeutics including doxorubicin, cyclophosphamide, and paclitaxel. Repeated thrombocytopenia was the main reason for suspecting secondary hematologic malignancy. She was diagnosed with t-MDS based on bone marrow examination and her treatment history for breast cancer. While azacitidine was originally administered to stabilize MDS, it also stabilized the patient's lung and lymph node metastases without any major toxicity. Therefore, the current case highlights the promising effects of HMAs for treating t-MDS following heavily pretreated breast cancer.
Collapse
Affiliation(s)
- Dong Won Baek
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Kyungpook National University Cancer Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | - Soo Jung Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Kyungpook National University Cancer Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | - Sang Kyun Sohn
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Kyungpook National University Cancer Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | - Joon Ho Moon
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Kyungpook National University Cancer Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | - Yee Soo Chae
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Kyungpook National University Cancer Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
23
|
Hartmann L, Nadarajah N, Meggendorfer M, Höllein A, Vetro C, Kern W, Haferlach T, Haferlach C, Stengel A. Molecular characterization of a second myeloid neoplasm developing after treatment for acute myeloid leukemia. Leukemia 2019; 34:811-820. [PMID: 31719678 DOI: 10.1038/s41375-019-0633-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/28/2019] [Indexed: 11/09/2022]
Abstract
Therapy-related myeloid neoplasms (tMN) following successful treatment of acute myeloid leukemia (AML) are rare and poorly characterized. To evaluate the presence of a common ancestral clone, we performed whole-exome sequencing of 25 patients at AML diagnosis, tMN diagnosis (tMDS: 13; tAML: 12), and matched remission samples, identifying 607 mutations affecting 504 different genes (46 recurrently mutated). Number of mutations was higher in tAML vs. tMDS cases (median 19 vs 13 mutations, p = 0.05). Focusing on 24 genes commonly mutated in hematological malignancies, 19/25 (76%) patients were found to share mutations between AML and tMN, mostly affecting epigenetic modifiers (21/32; 66%), splicing factors (6/32; 19%), and chromatin modifiers (3/32; 9%). Analysis of remission samples identified 13 persisting mutations in 10/22 patients, affecting DNMT3A (n = 6), TET2 (n = 5), IDH1 and SRSF2 (n = 1, each). Comparison of cytogenetics revealed that 9/12 patients with a normal karyotype (NK) in AML harbored aberrations in tMN, four aberrant AML cases presented with NK in tMN, four other patients showed unrelated cytogenetic aberrations. Our study provides novel insights into the pathogenesis of tMN, hypothesizing the presence of a common ancestral clone in AML and tMN. Mutations mostly affected epigenetic modifiers, which have previously been linked to clonal hematopoiesis.
Collapse
Affiliation(s)
- Luise Hartmann
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Niroshan Nadarajah
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Manja Meggendorfer
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Alexander Höllein
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Calogero Vetro
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Wolfgang Kern
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Torsten Haferlach
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Claudia Haferlach
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Anna Stengel
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany.
| |
Collapse
|
24
|
Chen KTJ, Gilabert-Oriol R, Bally MB, Leung AWY. Recent Treatment Advances and the Role of Nanotechnology, Combination Products, and Immunotherapy in Changing the Therapeutic Landscape of Acute Myeloid Leukemia. Pharm Res 2019; 36:125. [PMID: 31236772 PMCID: PMC6591181 DOI: 10.1007/s11095-019-2654-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 06/01/2019] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia that is becoming more prevalent particularly in the older (65 years of age or older) population. For decades, "7 + 3" remission induction therapy with cytarabine and an anthracycline, followed by consolidation therapy, has been the standard of care treatment for AML. This stagnancy in AML treatment has resulted in less than ideal treatment outcomes for AML patients, especially for elderly patients and those with unfavourable profiles. Over the past two years, six new therapeutic agents have received regulatory approval, suggesting that a number of obstacles to treating AML have been addressed and the treatment landscape for AML is finally changing. This review outlines the challenges and obstacles in treating AML and highlights the advances in AML treatment made in recent years, including Vyxeos®, midostaurin, gemtuzumab ozogamicin, and venetoclax, with particular emphasis on combination treatment strategies. We also discuss the potential utility of new combination products such as one that we call "EnFlaM", which comprises an encapsulated nanoformulation of flavopiridol and mitoxantrone. Finally, we provide a review on the immunotherapeutic landscape of AML, discussing yet another angle through which novel treatments can be designed to further improve treatment outcomes for AML patients.
Collapse
Affiliation(s)
- Kent T J Chen
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Interdisciplinary Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Roger Gilabert-Oriol
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Marcel B Bally
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- Cuprous Pharmaceuticals Inc., Vancouver, British Columbia, Canada.
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Ada W Y Leung
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Cuprous Pharmaceuticals Inc., Vancouver, British Columbia, Canada
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
Cooper JP, Khajaviyan S, Smith SD, Maloney DG, Shustov AR, Warren EH, Soma LA, Lynch RC, Ujjani C, Till B, Halpern AB, Gopal AK, Deeg HJ, Scott BL, Shadman M. Outcomes of Patients With Therapy-Related MDS After Chemoimmunotherapy for Chronic Lymphocytic Leukemia Compared With Patients With De Novo MDS: A Single-Institution Experience. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:390-395. [DOI: 10.1016/j.clml.2019.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 01/02/2023]
|
26
|
Gascón P, Krendyukov A, Mathieson N, Aapro M. Epoetin alfa for the treatment of myelodysplastic syndrome-related anemia: A review of clinical data, clinical guidelines, and treatment protocols. Leuk Res 2019; 81:35-42. [PMID: 31005849 DOI: 10.1016/j.leukres.2019.03.006] [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: 11/07/2018] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 01/12/2023]
Abstract
Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis, leading to hematopoietic precursor cell apoptosis and peripheral blood cytopenias. Anemia is the most frequently experienced cytopenia and is the main cause of MDS symptoms, with fatigue and dyspnea contributing to reduced quality of life and increased morbidity. As MDS disease course and prognosis is influenced by disease factors, prognostic scoring systems have been developed for MDS to aid clinical and therapeutic decisions following diagnosis. Erythropoiesis- stimulating agents (ESAs) have been used for many years to treat anemia in patients with lower-risk MDS without chromosomal abnormalities. The use of ESAs is recommended by international clinical practice guidelines, due to the large body of evidence demonstrating their effectiveness in lower-risk MDS. In March 2017, the European Medicines Agency approved epoetin alfa for the treatment of anemia in lower-risk MDS patients, based on the results from a Phase 3 clinical trial and three European registry studies. The effectiveness of biosimilar epoetin alfa (Binocrit®, Sandoz) to correct anemia in lower-risk MDS patients has also been demonstrated in a retrospective, single-center, observational study. The recent approval of epoetin alfa by the EMA in this setting will provide clinicians with a welcome, approved treatment option for lower-risk MDS.
Collapse
Affiliation(s)
- Pere Gascón
- Department of Hematology- Oncology, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain.
| | - Andriy Krendyukov
- Hematology/Nephrology, Sandoz Biopharmaceuticals, HEXAL AG, Holzkirchen, Germany.
| | - Nicola Mathieson
- Hematology/Nephrology, Sandoz Biopharmaceuticals, HEXAL AG, Holzkirchen, Germany.
| | - Matti Aapro
- Institut Multidisciplinaire d'Oncologie, Clinique de Genolier, Genolier, Switzerland.
| |
Collapse
|
27
|
Madanat YF, Gerds AT. Can allogeneic hematopoietic cell transplant cure therapy-related acute leukemia? Best Pract Res Clin Haematol 2019; 32:104-113. [DOI: 10.1016/j.beha.2019.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 01/16/2023]
|
28
|
Pombo-de-Oliveira MS, Andrade FG, Brisson GD, Dos Santos Bueno FV, Cezar IS, Noronha EP. Acute myeloid leukaemia at an early age: Reviewing the interaction between pesticide exposure and KMT2A-rearrangement. Ecancermedicalscience 2017; 11:782. [PMID: 29225689 PMCID: PMC5718248 DOI: 10.3332/ecancer.2017.782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Indexed: 12/29/2022] Open
Abstract
Acute myeloid leukaemia (AML) in early childhood is characterised by a high frequency of recurrent genomic aberrations associated with distinct myeloid subtypes, clinical outcomes and pathogenesis. Genomic instability is the first step of pathogenic mechanism in early childhood AML. A sum of adverse events is necessary to the development of infant AML (i-AML), which includes latency of biochemical-molecular and cellular effects. Inherited genetic susceptibility associated with exposures to biotransformation substances can modulate the risk of DNA damage and it is a very important piece in the pathogenic puzzle. In this review, we have aimed to explore the chain of events in the time-points of the natural history of i-AML, which includes maternal exposures during pregnancy, the speculations about the formation of somatic mutations during foetal life and the secondary genomic aberrations associated with i-AML. The modulation of risk conferred by xenobiotic metabolism´s genes variants is the bottom line of the pathogenic process. Since we have conducted observational and molecular investigations in early childhood leukaemia, the data focused here is based on Brazilian findings with summarised results of our experience with epidemiological and molecular studies in early-age leukaemia.
Collapse
Affiliation(s)
- Maria S Pombo-de-Oliveira
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil
| | - Francianne Gomes Andrade
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil
| | - Gisele Dallapicola Brisson
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil
| | - Filipe Vicente Dos Santos Bueno
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil
| | - Ingrid Sardou Cezar
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil
| | - Elda Pereira Noronha
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil
| |
Collapse
|
29
|
Aldoss I, Pham A, Li SM, Gendzekhadze K, Afkhami M, Telatar M, Hong H, Padeganeh A, Bedell V, Cao T, Khaled SK, Malki MMA, Salhotra A, Ali H, Aribi A, Palmer J, Aoun P, Spielberger R, Stein AS, Snyder D, O'Donnell MR, Murata-Collins J, Senitzer D, Weisenburger D, Forman SJ, Pullarkat V, Marcucci G, Pillai R, Nakamura R. Favorable impact of allogeneic stem cell transplantation in patients with therapy-related myelodysplasia regardless of TP53 mutational status. Haematologica 2017; 102:2030-2038. [PMID: 28971906 PMCID: PMC5709102 DOI: 10.3324/haematol.2017.172544] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/20/2017] [Indexed: 01/04/2023] Open
Abstract
Therapy-related myelodysplastic syndrome is a long-term complication of cancer treatment in patients receiving cytotoxic therapy, characterized by high-risk genetics and poor outcomes. Allogeneic hematopoietic cell transplantation is the only potential cure for this disease, but the prognostic impact of pre-transplant genetics and clinical features has not yet been fully characterized. We report here the genetic and clinical characteristics and outcomes of a relatively large cohort of patients with therapy-related myelodysplastic syndrome (n=67) who underwent allogeneic transplantation, comparing these patients to similarly treated patients with de novo disease (n=199). The 5-year overall survival was not different between patients with therapy-related and de novo disease (49.9% versus 53.9%; P=0.61) despite a higher proportion of individuals with an Intermediate-2/High International Prognostic Scoring System classification (59.7% versus 43.7%; P=0.003) and high-risk karyotypes (61.2% versus 30.7%; P<0.01) among the patients with therapy-related disease. In mutational analysis, TP53 alteration was the most common abnormality in patients with therapy-related disease (n=18: 30%). Interestingly, the presence of mutations in TP53 or in any other of the high-risk genes (EZH2, ETV6, RUNX1, ASXL1: n=29: 48%) did not significantly affect either overall survival or relapse-free survival. Allogeneic stem-cell transplantation is, therefore, a curative treatment for patients with therapy-related myelodysplastic syndrome, conferring a similar long-term survival to that of patients with de novo disease despite higher-risk features. While TP53 alteration was the most common mutation in therapy-related myelodysplastic syndrome, the finding was not detrimental in our case-series.
Collapse
Affiliation(s)
- Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Anh Pham
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Sierra Min Li
- Department of Information Sciences, Division of Biostatistics, City of Hope, Duarte, CA, USA
| | | | | | | | - Hao Hong
- Department of Pathology, City of Hope, Duarte, CA, USA
| | | | | | - Thai Cao
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Samer K Khaled
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Amandeep Salhotra
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Haris Ali
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Ahmed Aribi
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Joycelynne Palmer
- Department of Information Sciences, Division of Biostatistics, City of Hope, Duarte, CA, USA
| | - Patricia Aoun
- Department of Pathology, City of Hope, Duarte, CA, USA
| | - Ricardo Spielberger
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Anthony S Stein
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - David Snyder
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Margaret R O'Donnell
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | | | | | | | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Guido Marcucci
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| | - Raju Pillai
- Department of Pathology, City of Hope, Duarte, CA, USA
| | - Ryotaro Nakamura
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA, USA
| |
Collapse
|
30
|
Cîrstea M, Coliță A, Ionescu B, Ghiaur A, Vasilescu D, Dobrea C, Jardan C, Dragomir M, Gheorghe A, Várady Z, Lupu AR. Therapy-related myelodysplastic syndrome after successful treatment of acute promyelocytic leukemia: case report and literature review. REV ROMANA MED LAB 2017. [DOI: 10.1515/rrlm-2017-0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Abstract
In the 2016 revision of the World Health Organization classification the term therapy-related myeloid neoplasia (t-MN) defines a subgroup of acute myeloid leukemia (AML) comprising patients who develop myelodysplastic syndrome (MDS-t) or acute myeloid leukemia (AML-t) after treatment with cytotoxic and/or radiation therapy for various malignancies or autoimmune disorders. We report the case of a 36 year old patient with t-MN (t-MDS) after achieving complete remission (CR) of a PML-RARA positive acute promyelocytic leukemia (APL) at 32 months after diagnosis. Initially classified as low risk APL and treated according to the AIDA protocol - induction and 3 consolidation cycles - the patient achieved a complete molecular response in September 2013 and started maintenance therapy. On follow-up PML-RARA transcript remained negative. In January 2016 leukopenia and thrombocytopenia developed and a peripheral blood smear revealed hypogranular and agranular neutrophils. Immunophenotyping in the bone marrow aspirate identified undifferentiated blast cells that did not express cytoplasmic myeloperoxidase. The cytogenetic study showed normal karyotype. The molecular biology tests not identified PMLRARA transcript. A diagnosis of t-MDS (AREB-2 - WHO 2008) was established. Treatment of AML was started with 2 “3+7” regimens and 1 MEC cycle. Two months from diagnosis, while in CR, an allogeneic HSCT from an unrelated HLA compatible donor was performed after myeloablative regimen. An unfavorable clinical evolution was followed by death on day 9 after transplantation. The occurrence of t-MNs during CR of APL represents a particular problem in terms of follow-up and differential diagnosis of relapse and constitutes a dramatic complication for a disease with a favorable prognosis.
This work was supported by the grants PN 41-087 /PN2-099 from the Romanian Ministry of Research and Technology
Collapse
Affiliation(s)
- Mihaela Cîrstea
- Department of Onco-hematology, Fundeni Clinical Institute , Bucharest , Romania
| | - Adriana Coliță
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | - Bogdan Ionescu
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | | | | | - Camelia Dobrea
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | | | | | - Anca Gheorghe
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | - Zsofia Várady
- Fundeni Clinical Institute Romania , Bucharest , Romania
| | | |
Collapse
|
31
|
Maung SW, Burke C, Hayde J, Walshe J, McDermott R, Desmond R, McHugh J, Enright H. A review of therapy-related myelodysplastic syndromes and acute myeloid leukaemia (t-MDS/AML) in Irish patients: a single centre experience. ACTA ACUST UNITED AC 2017; 22:341-346. [PMID: 28196450 DOI: 10.1080/10245332.2017.1286539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVES To demonstrate the incidence, characteristics, treatment and outcomes of patients with therapy-related myelodysplastic syndromes and therapy-related acute myeloid leukaemia (t-MDS/AML) in a tertiary referral centre. METHODS Patients meeting the diagnostic criteria for t-MDS/AML from 2003 to 2014 were reviewed to analyse their diagnostic features, details of antecedent disorder and treatment, approach to management and survival. RESULTS 39 patients who developed t-MDS/AML were identified with incidence of 8.7%. Median age and gender distribution were similar to de novo MDS but t-MDS/AML patients had greater degree of cytopenia and adverse karyotypes. Time to development of t-MDS/AML was shortest for patients with antecedent haematological malignancy compared to solid tumours and autoimmune disorders (46, 85 and 109 months). Patients with prior acute leukaemia had the shortest latency and poor overall survival. Treatment options included best supportive care (56%), Azacitidine (31%) or intensive chemotherapy/allogeneic transplant (13%). Median OS of all patients was 14 months. Survival declined markedly after two years and 5-year OS was 13.8%. Longer survival was associated with blast count <5% at diagnosis, previous haematological disorder, lower risk IPSS-R and a normal karyotype. Four out of five patients who received intensive therapy/transplant remain alive with median OS of 14 months. Median OS of Azacitidine-treated group was 11 months. DISCUSSION t-MDS/AML patients showed unique characteristics which influenced their treatment and outcomes. IPSS-R may be useful in risk-adapted treatment approaches and can predict outcomes. Survival remains poor but improved outcomes were seen with allogeneic transplantation. Azacitidine may be effective in patients unfit for intensive therapies.
Collapse
Affiliation(s)
- Su W Maung
- a Department of Haematology , Tallaght Hospital , Dublin , Ireland
| | - Cathie Burke
- a Department of Haematology , Tallaght Hospital , Dublin , Ireland
| | - Jennifer Hayde
- b Department of Pharmacy , Tallaght Hospital , Dublin , Ireland
| | - Janice Walshe
- c Department of Medical Oncology , Tallaght Hospital , Dublin , Ireland
| | - Ray McDermott
- c Department of Medical Oncology , Tallaght Hospital , Dublin , Ireland
| | - Ronan Desmond
- a Department of Haematology , Tallaght Hospital , Dublin , Ireland
| | - Johnny McHugh
- a Department of Haematology , Tallaght Hospital , Dublin , Ireland
| | - Helen Enright
- a Department of Haematology , Tallaght Hospital , Dublin , Ireland
| |
Collapse
|
32
|
Cho HW, Choi YB, Yi ES, Lee JW, Sung KW, Koo HH, Yoo KH. Therapy-related myeloid neoplasms in children and adolescents. Blood Res 2016; 51:242-248. [PMID: 28090486 PMCID: PMC5234240 DOI: 10.5045/br.2016.51.4.242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/18/2016] [Accepted: 09/06/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND This retrospective study aimed to characterize and analyze the outcome of therapy-related myeloid neoplasms (t-MNs) in children and adolescents. METHODS The medical records of 16 patients under 21 years of age at the time of t-MN diagnosis were reviewed. RESULTS The median patient age was 11.5 years (range, 1.6-20.4 yr). Twelve patients had therapy-related acute myeloid leukemia, 3 patients had myelodysplastic syndrome, and 1 patient had chronic myelomonocytic leukemia. The median latency period was 29 months (range, 11-68 mo). Fourteen patients had cytogenetic aberrations, 8 of whom had an 11q23 abnormality. Of the 13 patients treated with curative intent, 12 patients received myeloid-type induction therapy that led to complete remission (CR) in 8 patients. Nine patients underwent allogeneic transplantation; 4 patients did not undergo transplantation due to chemotherapy-related toxic death (N=3) or parental refusal (N=1). The 5-year overall survival and event-free survival of the 13 patients treated with a curative intent were 46.2% and 30.8%, respectively. For the 9 patients who underwent allogeneic transplantation, the 5-year event-free survival was 66.7%. CONCLUSION A significant proportion of young patients with t-MNs can experience long-term survival, and allogeneic transplantation plays a key role for attaining cure in these patients.
Collapse
Affiliation(s)
- Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Bae Choi
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
| | - Eun Sang Yi
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.; Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
33
|
ESPíRITO Santo AE, Chacim S, Ferreira I, Leite L, Moreira C, Pereira D, Dantas Brito MD, Nunes M, Domingues N, Oliveira I, Moreira I, Martins A, Viterbo L, Mariz JM, Medeiros R. Effect of therapy-related acute myeloid leukemia on the outcome of patients with acute myeloid leukemia. Oncol Lett 2016; 12:262-268. [PMID: 27347135 DOI: 10.3892/ol.2016.4591] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 10/28/2015] [Indexed: 01/30/2023] Open
Abstract
Therapy-related acute myeloid leukemia (t-AML) is a rare and almost always fatal late side effect of antineoplastic treatment involving chemotherapy, radiotherapy or the two combined. The present retrospective study intended to characterize t-AML patients that were diagnosed and treated in a single referral to an oncological institution in North Portugal. Over the past 10 years, 231 cases of AML were diagnosed and treated at the Portuguese Institute of Oncology of Porto, of which 38 t-AML cases were identified. Data regarding the patient demographics, primary diagnosis and treatment, age at onset of therapy-related myeloid neoplasm, latency time of the neoplasm, cytogenetic characteristics, AML therapy and outcome were collected from medical records. A previous diagnosis with solid tumors was present in 28 patients, and 10 patients possessed a history of hematological conditions, all a lymphoproliferative disorder. Breast cancer was the most frequent solid tumor identified (39.5% of all solid tumors diagnosed). The mean latency time was 3 years. In the present study, t-AML patients were older (P<0.001) and more frequently carried cytogenetic abnormalities (P=0.009) compared with de novo AML patients. The overall survival time was observed to be significantly poorer among individuals with t-AML (P<0.001). However, in younger patients (age, <50 years) there was no difference between the overall survival time of patients with t-AML and those with de novo AML (P=0.983). Additionally, patients with promyelocytic leukemia possess a good prognosis, even when AML occurs as a secondary event (P=0.98). To the best of our knowledge, the present study is the first to evaluate t-AML in Portugal and the results are consistent with the data published previously in other populations. The present study concludes that although t-AML demonstrates a poor prognosis, this is not observed among younger patients or promyelocytic leukemia patients.
Collapse
Affiliation(s)
- Ana Espírito ESPíRITO Santo
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| | - Sérgio Chacim
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Isabel Ferreira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Luís Leite
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Claudia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Dulcineia Pereira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | | | - Marta Nunes
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Nelson Domingues
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Isabel Oliveira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Ilídia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Angelo Martins
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Luísa Viterbo
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - José Mário Mariz
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group Investigation Centre, Portuguese Institute of Oncology of Porto, Porto, Portugal; Abel Salazar Institute of Biomedical Sciences, University of Porot, Porto, Portugal; Department of Oncology, Portuguese Institute of Oncology, Porto, Portugal; Biomedical Research Centre, Faculty of Health Sciences of Fernando Pessoa University, Porto, Portugal; Department of Research, Portuguese League Against Cancer (Regional North Core), Porto, Portugal
| |
Collapse
|
34
|
Park EH, Lee H, Won YJ, Ju HY, Oh CM, Ingabire C, Kong HJ, Park BK, Yoon JY, Eom HS, Lee E, Park HJ. Nationwide statistical analysis of myeloid malignancies in Korea: incidence and survival rate from 1999 to 2012. Blood Res 2015; 50:204-17. [PMID: 26770948 PMCID: PMC4705046 DOI: 10.5045/br.2015.50.4.204] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/06/2015] [Accepted: 12/07/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Large-scale epidemiologic analysis for hematologic malignancies will be helpful to understand the trends in incidence and survival. METHODS The Korea Central Cancer Registry (KCCR) updated the nationwide analysis on the incidence and survival of myeloid malignancies, from the Korean National Cancer Incidence Database between 1999 and 2012. Myeloid malignancies were classified based on the International Classification of Diseases for Oncology 3(rd) edition (ICD-O-3). RESULTS Overall 3,771 cases of myeloid diseases, which was 1.7% of all cancers, were identified in 2012. The highest incidence of myeloid malignancies was observed in age 70s and male predominance was noted (1.3:1). Acute myeloid leukemia (AML) was the most frequent subtype, followed by myeloproliferative neoplasms (MPN), myelodysplastic syndrome (MDS) and MDS/MPN: age-standardized incidence rates (ASR) in 2012 for each disease were 2.02, 1.95, 1.13, and 0.12 per 100,000 persons, respectively. The ASR for all myeloid malignancies was increased from 3.31 in 1999 to 5.70 in 2012 with the annual percentage change (APC) of 5.4 %. Five-year relative survival rate (RS) for myeloid malignancies has gradually improved for decades. RS changed from 26.3% to 34.8% in AML, specifically from 51.6% to 69.6% in acute promyelocytic leukemia (APL) and from 23.8% to 29.9% in non-APL AML, between 1996-2000 and 2008-2012. RS also increased from 81.8% to 87.1% in MPN, with a significant improvement in CML (from 74.5% to 85.5%), and from 27.3% to 31.7% in MDS/MPN between 2001-2005 and 2008-2012. However, there was no survival improvement in MDS during the study period (45.6% in 2001-2005 to 44.4% in 2008-2012). CONCLUSION This report updated the nationwide statistical analysis on myeloid malignancies since 2008, showing increasing incidence and improving trends in survival.
Collapse
Affiliation(s)
- Eun-Hye Park
- The Korea Central Cancer Registry, Division of Cancer Registration and Surveillance, National Cancer Center, Goyang, Korea
| | - Hyewon Lee
- Hematologic Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Young-Joo Won
- The Korea Central Cancer Registry, Division of Cancer Registration and Surveillance, National Cancer Center, Goyang, Korea.; Department of Cancer Control and Policy, National Cancer Center, Goyang, Korea
| | - Hee Young Ju
- Center for Pediatric Oncology, National Cancer Center, Goyang, Korea
| | - Chang-Mo Oh
- The Korea Central Cancer Registry, Division of Cancer Registration and Surveillance, National Cancer Center, Goyang, Korea
| | - Cecile Ingabire
- The Korea Central Cancer Registry, Division of Cancer Registration and Surveillance, National Cancer Center, Goyang, Korea.; Department of Cancer Control and Policy, National Cancer Center, Goyang, Korea
| | - Hyun-Joo Kong
- The Korea Central Cancer Registry, Division of Cancer Registration and Surveillance, National Cancer Center, Goyang, Korea
| | - Byung-Kiu Park
- Center for Pediatric Oncology, National Cancer Center, Goyang, Korea
| | - Ju Young Yoon
- Center for Pediatric Oncology, National Cancer Center, Goyang, Korea
| | - Hyeon-Seok Eom
- Hematologic Oncology Clinic, National Cancer Center, Goyang, Korea.; Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Eunyoung Lee
- Hematologic Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Hyeon Jin Park
- Center for Pediatric Oncology, National Cancer Center, Goyang, Korea
| |
Collapse
|
35
|
Rare cytogenetic abnormalities in myelodysplastic syndromes. Mediterr J Hematol Infect Dis 2015; 7:e2015034. [PMID: 25960862 PMCID: PMC4418404 DOI: 10.4084/mjhid.2015.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/20/2015] [Indexed: 02/03/2023] Open
Abstract
The karyotype represents one of the main cornerstones for the International Prognostic Scoring System (IPSS) and the revised IPSS-R (IPSS-R) that are most widely used for prognostication in patients with myelodysplastic syndromes (MDS). The most frequent cytogenetic abnormalities in MDS, i.e. del(5q), -7/del(7q), +8, complex karyotypes, or -Y have been extensively explored for their prognostic impact. The IPSS-R also considers some less frequent abnormalities such as del(11q), isochromosome 17, +19, or 3q abnormalities. However, more than 600 different cytogenetic categories had been identified in a previous MDS study. This review aims to focus interest on selected rare cytogenetic abnormalities in patients with MDS. Examples are numerical gains of the chromosomes 11 (indicating rapid progression), of chromosome 14 or 14q (prognostically intermediate to favorable), -X (in females, with an intermediate prognosis), or numerical abnormalities of chromosome 21. Structural abnormalities are also considered, e.g. del(13q) that is associated with bone marrow failure syndromes and favorable response to immunosuppressive therapy. These and other rare cytogenetic abnormalities should be integrated into existing prognostication systems such as the IPSS-R. However, due to the very low number of cases, this is clearly dependent on international collaboration. Hopefully, this article will help to inaugurate this process.
Collapse
|
36
|
Singh ZN, Jethava Y, Post GR, Alapat D, Sawyer J, Waheed S, Nair B, Usmani SZ, Bailey C, Petty N, Van Rhee F, Barlogie B. Fulminant onset of acute leukemia from normal hematopoiesis within 3 months of follow up for multiple myeloma treated with total therapy protocols. Clin Case Rep 2015; 3:183-92. [PMID: 25838910 PMCID: PMC4377252 DOI: 10.1002/ccr3.180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 10/02/2014] [Accepted: 10/25/2014] [Indexed: 01/12/2023] Open
Abstract
Assiduous surveillance for genetic aberrations is necessary in patients on cytotoxic therapies to detect therapy-related myeloid neoplasms (t-MN). Current modalities include metaphase cytogenetics and FISH. Since t-MN may develop abruptly in cytogenetically normal patients, a discussion exploring additional methods such as SNP-array and targeted-deep-sequencing to detect subchromosomal abnormalities is needed.
Collapse
Affiliation(s)
- Zeba N Singh
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Yogesh Jethava
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Ginell R Post
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Daisy Alapat
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Jeffrey Sawyer
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Sarah Waheed
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Bijay Nair
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Saad Z Usmani
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Clyde Bailey
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Nathan Petty
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Frits Van Rhee
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Bart Barlogie
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| |
Collapse
|
37
|
Bobadilla-Morales L, Pimentel-Gutiérrez HJ, Gallegos-Castorena S, Paniagua-Padilla JA, Ortega-de-la-Torre C, Sánchez-Zubieta F, Silva-Cruz R, Corona-Rivera JR, Zepeda-Moreno A, González-Ramella O, Corona-Rivera A. Pediatric donor cell leukemia after allogeneic hematopoietic stem cell transplantation in AML patient from related donor. Mol Cytogenet 2015; 8:5. [PMID: 25674158 PMCID: PMC4324859 DOI: 10.1186/s13039-014-0105-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/26/2014] [Indexed: 12/26/2022] Open
Abstract
Here we present a male patient with acute myeloid leukemia (AML) initially diagnosed as M5 and with karyotype 46,XY. After induction therapy, he underwent a HLA-matched allogeneic hematopoietic stem cell transplantation, and six years later he relapsed as AML M1 with an abnormal karyotype //47,XX,+10[2]/47,XX,+11[3]/48,XX,+10,+11[2]/46,XX[13]. Based on this, we tested the possibility of donor cell origin by FISH and molecular STR analysis. We found no evidence of Y chromosome presence by FISH and STR analysis consistent with the success of the allogeneic hematopoietic stem cell transplantation from the female donor. FISH studies confirmed trisomies and no evidence of MLL translocation either p53 or ATM deletion. Additionally 28 fusion common leukemia transcripts were evaluated by multiplex reverse transcriptase-polymerase chain reaction assay and were not rearranged. STR analysis showed a complete donor chimerism. Thus, donor cell leukemia (DCL) was concluded, being essential the use of cytological and molecular approaches. Pediatric DCL is uncommon, our patient seems to be the sixth case and additionally it presented a late donor cell leukemia appearance. Different extrinsic and intrinsic mechanisms have been considered to explain this uncommon finding as well as the implications to the patient.
Collapse
Affiliation(s)
- Lucina Bobadilla-Morales
- Laboratorio de Citogenética, Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona Rivera"/Doctorado de Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Jalisco México.,Instituto de Investigación en Cáncer de la Infancia y la Adolescencia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco México.,Unidad de Citogenética, Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México.,Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México.,División de Pediatría, Centro de Registro e Investigación sobre Anomalías Congénitas (CRIAC), Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Helia J Pimentel-Gutiérrez
- Laboratorio de Citogenética, Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona Rivera"/Doctorado de Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Jalisco México.,Unidad de Citogenética, Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Sergio Gallegos-Castorena
- Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Jenny A Paniagua-Padilla
- Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Citlalli Ortega-de-la-Torre
- Laboratorio de Citogenética, Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona Rivera"/Doctorado de Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Jalisco México.,Unidad de Citogenética, Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México.,División de Pediatría, Centro de Registro e Investigación sobre Anomalías Congénitas (CRIAC), Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Fernando Sánchez-Zubieta
- Laboratorio de Citogenética, Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona Rivera"/Doctorado de Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Jalisco México.,Instituto de Investigación en Cáncer de la Infancia y la Adolescencia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco México.,Unidad de Citogenética, Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México.,Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Rocio Silva-Cruz
- Laboratorio de Citogenética, Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona Rivera"/Doctorado de Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Jalisco México.,División de Pediatría, Centro de Registro e Investigación sobre Anomalías Congénitas (CRIAC), Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Jorge R Corona-Rivera
- Laboratorio de Citogenética, Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona Rivera"/Doctorado de Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Jalisco México.,División de Pediatría, Centro de Registro e Investigación sobre Anomalías Congénitas (CRIAC), Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Abraham Zepeda-Moreno
- Instituto de Investigación en Cáncer de la Infancia y la Adolescencia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco México
| | - Oscar González-Ramella
- Instituto de Investigación en Cáncer de la Infancia y la Adolescencia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco México.,Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México
| | - Alfredo Corona-Rivera
- Laboratorio de Citogenética, Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona Rivera"/Doctorado de Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Jalisco México.,Instituto de Investigación en Cáncer de la Infancia y la Adolescencia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco México.,Unidad de Citogenética, Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México.,Servicio de Hematología y Oncología Pediátrica, División de Pediatría, Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México.,División de Pediatría, Centro de Registro e Investigación sobre Anomalías Congénitas (CRIAC), Nuevo Hospital Civil de Guadalajara, "Dr. Juan I. Menchaca", Guadalajara, Jalisco México.,Laboratorio de Citogenética Genotoxicidad y Biomonitoreo, Instituto de Genética Humana "Dr. Enrique Corona-Rivera", Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, #Sierra Mojada 950, S.L., Edificio P, Nivel 2, Col. Independencia, Guadalajara, Jalisco CP: 44340 México
| |
Collapse
|
38
|
Bang JI, Lee ES, Kim TS, Kim SK. Unexpected Second Primary Malignancies Detected by F-18 FDG PET/CT During Follow-up for Primary Malignancy: Two Case Reports. Nucl Med Mol Imaging 2014; 49:65-8. [PMID: 25767625 DOI: 10.1007/s13139-014-0302-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/13/2014] [Accepted: 10/15/2014] [Indexed: 10/24/2022] Open
Abstract
As the survival rate of cancer patients has increased over the last few decades, the risk of cancer survivors developing second primary malignancies has gained attention. We report two rare cases of second primary hematologic malignancy detected by (18)F-fluorodeoxyglucose (F-18 FDG) positron emission tomography/computed tomography (PET/CT) during follow-up for primary solid malignancies. Acute lymphoblastic leukemia developed in a breast cancer patient and non-Hodgkin lymphoma in an anal cancer patient. F-18 FDG PET/CT findings led to the diagnosis of unexpected second primary hematologic malignancy in cancer survivors in these two cases.
Collapse
Affiliation(s)
- Ji-In Bang
- Department of Nuclear Medicine, Research Institute and Hospital, National Cancer Center, Korea, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi 410-769 Republic of Korea
| | - Eun Seong Lee
- Department of Nuclear Medicine, Research Institute and Hospital, National Cancer Center, Korea, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi 410-769 Republic of Korea
| | - Tae-Sung Kim
- Department of Nuclear Medicine, Research Institute and Hospital, National Cancer Center, Korea, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi 410-769 Republic of Korea
| | - Seok-Ki Kim
- Department of Nuclear Medicine, Research Institute and Hospital, National Cancer Center, Korea, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi 410-769 Republic of Korea
| |
Collapse
|
39
|
Chen Y, Estrov Z, Pierce S, Qiao W, Borthakur G, Ravandi F, Kadia T, Brandt M, O'Brien S, Jabbour E, Garcia-Manero G, Cortes J, Beran M. Myeloid neoplasms after breast cancer: "therapy-related" not an independent poor prognostic factor. Leuk Lymphoma 2014; 56:1012-9. [PMID: 25048874 DOI: 10.3109/10428194.2014.946023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Two hundred and thirty-five consecutive patients presenting to a single center with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) after breast cancer treatment were compared with matched patients with de novo AML or MDS. There was no significant difference in median overall survival (OS) times between patients with therapy-related AML and those with de novo AML (8.7 months vs.10.2 months; p = 0.17). Patients with therapy-related MDS had slightly lower median baseline platelet counts and a higher frequency of poor cytogenetics than those with de novo MDS, but the two groups had similar OS times (13.6 months vs. 18.9 months; p = 0.06). Multivariate analysis revealed that cytogenetic risk, baseline white blood cell count, age and performance status were predictive for OS time in AML and that cytogenetic risk and performance status were predictive for OS time in MDS. Having therapy-related disease is not an independent risk factor in patients with myeloid neoplasms and with a history of breast cancer. Clinical trials should be designed to serve both populations.
Collapse
Affiliation(s)
- Yiming Chen
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center , Houston, TX
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Age related risk of myelodysplastic syndrome and acute myeloid leukemia among breast cancer survivors. Breast Cancer Res Treat 2013; 142:629-36. [PMID: 24265034 DOI: 10.1007/s10549-013-2773-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/10/2013] [Indexed: 10/26/2022]
Abstract
UNLABELLED Increased incidence of acute myeloid leukemia (AML) has been identified among breast cancer (BC) survivors but measurement has not included myelodysplastic syndrome (MDS). Our aim is to identify age and stage related MDS/AML incidence post BC diagnosis. We used the 2001-2009 Surveillance, Epidemiology, and end results (SEER) database to identify first primary stage I-III BC patients. Subsequent MDS or AML diagnosis was identified with observed rates compared to expected MDS/AML incidence in the general population. Age adjusted observed/expected rate ratios and 95 % confidence intervals (CI) were calculated. The unadjusted all age and stage MDS/AML incidence rate was .15 % (470/306,691) with a progressively higher rate by age (age 20-49 = .11, age 50-64 = .14, age 65+ =.21, and age 75+ =.18) and stage (stage I = .11, stage II = .18, and stage III = .22). Compared to the general population, BC patients had a 2.75-fold [95 % CI 2.51-3.00] increased relative risk of being diagnosed with MDS/AML. Young age survivors had highest relative risk [age 20-49: relative risk (RR) = 10.60 (95 % CI 8.57-12.93); age 50-64: 5.96 (95 % CI 5.13, 6.88); age 65-74 year-olds: 2.94 (95 % CI 2.45, 3.50); and age ≥75 year-olds: 1.28 (95 % CI 1.03, 1.56)]. Separately MDS relative risk was highest among young women [30.44 (95 % CI = 19.63, 44.62)]. MDS/AML relative risk increased from 1.87 to 5.66 for stage I-III. CONCLUSIONS Myelodysplastic syndrome and acute myeloid leukemia relative risk is substantially elevated among breast cancer survivors especially those aged 20-49. While the actual number is small, MDS/AML is a serious disease. More research is needed to identify the treatments that put women at risk and find less leukemogenic options, especially for young women.
Collapse
|
41
|
Tanizawa RSDS, Kumeda CA, de Azevedo Neto RS, Leal ADM, Ferreira PDB, Velloso EDRP. Karyotypic and fluorescent in situ hybridization study of the centromere of chromosome 7 in secondary myeloid neoplasms. Rev Bras Hematol Hemoter 2013; 33:425-31. [PMID: 23049358 PMCID: PMC3459367 DOI: 10.5581/1516-8484.20110117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Accepted: 09/05/2011] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Secondary myeloid neoplasms comprise a group of secondary diseases following exposure to myelotoxic agents or due to congenital diseases. The improvement of anticancer agents and immunosuppressive drugs seem to be associated with an increased incidence of secondary myeloid neoplasms. Karyotyping of bone marrow is essential for diagnosis and prognosis. Previous use of alkylating agents and radiation are associated with clonal abnormalities such as recurrent unbalanced -5/5q-, -7/7q- and complex karyotypes, whereas topoisomerase-II inhibitors lead to changes such as the balanced 11q23 rearrangement, t(8;21), t(15;17) and inv(16). OBJECTIVE To study the clinical and cytogenetic data of patients with secondary myeloid neoplasms who took antineoplastic and/or immunosuppressive drugs or progressed from aplastic anemia. METHODS The clinical and cytogenetic characteristics of 42 patients diagnosed with secondary myeloid neoplasms in one institution were retrospectively evaluated. Of these, 25, 11 and 6 patients had had oncological diseases, aplastic anemia and other diseases, respectively. Conventional cytogenetic and FISH analyses were performed for monosomy 7. RESULTS The cytogenetic study was conclusive in 32 cases with 84.4% of clonal abnormalities. Monosomy 7 and complex karyotypes were present in 44.4% and 37%, respectively. A high prevalence of unbalanced abnormalities (96.3%) was observed. Monosomy 7 was more prevalent in patients with myelodysplastic syndromes/myeloid neoplasms after aplastic anemia (66.6%). The median survival after diagnosis of myeloid neoplasms was only 5.7 months. Normal cytogenetics was associated to better survival (p-value = 0.03). There was a slightly worse trend of survival for patients with complex karyotypes (p-value = 0.057). Abnormal karyotype was an independent risk factor for poor survival (p-value = 0.012). CONCLUSION This study enhances the importance of cytogenetic analysis of patients at the time of diagnosis of secondary myeloid neoplasms.
Collapse
Affiliation(s)
- Roberta Sandra da Silva Tanizawa
- Cytogenetics Laboratory, Serviço de Hematologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo - USP, São Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
42
|
Mishra A, Corrales-Yepez M, Ali NA, Kharfan-Dabaja M, Padron E, Zhang L, Epling-Burnette PK, Pinilla-Ibarz J, Lancet JE, List AF, Komrokji RS. Validation of the revised International Prognostic Scoring System in treated patients with myelodysplastic syndromes. Am J Hematol 2013; 88:566-70. [PMID: 23605934 DOI: 10.1002/ajh.23454] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 04/02/2013] [Indexed: 11/07/2022]
Abstract
The International Prognostic Scoring System (IPSS) was recently revised (IPSS-R) under the auspices of the MDS Foundation as a collaborative international effort to refine its prognostic power. Our purpose was to externally validate this new risk model using a large single-institution cohort, determine its prognostic power in patients receiving active treatment, and explore its utility in guiding therapeutic decisions. Data were collected retrospectively from our myelodysplastic syndrome (MDS) database and verified by chart review. Of the data available for 1,088 patients, 152 (14%), 353 (32%), 237 (22%), 190 (18%), and 156 (14%) patients were classified as very low, low, intermediate, high, and very high risk, respectively, with median overall survival (OS) of 90 (95%CI 71-109), 54 (95%CI 50-59), 34 (95%CI 26-43), 21 (95%CI 17-25), and 13 months (95%CI 11-15), respectively (P < 0.005). We found that the IPSS-R further refined prognostic discrimination in all IPSS risk categories, particularly in the intermediate 1 and 2 groups. Among high and very high IPSS-R patients receiving azacitidine, OS was significantly improved versus patients not receiving azacitidine, with corresponding median OS of 25 versus 18 months (P = 0.028) and 15 versus 9 months (P = 0.005), respectively. Similarly, patients with IPSS-R high- and very high-risk disease who underwent allogeneic hematopoietic stem cell transplantation had significantly improved OS versus nontransplant approaches (P < 0.005). High and very high IPSS-R patients derived a survival advantage from disease-modifying therapies. Our data validate the prognostic value of the proposed IPSS-R and show that its refined IPSS prognostic discrimination can be applied to actively treated patients.
Collapse
Affiliation(s)
- Asmita Mishra
- H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | | | - Najla Al Ali
- H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | | | - Eric Padron
- H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Ling Zhang
- H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | | | | | | | - Alan F. List
- H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| | - Rami S. Komrokji
- H. Lee Moffitt Cancer Center and Research Institute; Tampa Florida
| |
Collapse
|
43
|
Huh HJ, Lee SH, Yoo KH, Sung KW, Koo HH, Kim K, Jang JH, Jung C, Kim SH, Kim HJ. Therapy-related myeloid neoplasms in 39 Korean patients: a single institution experience. Ann Lab Med 2013; 33:97-104. [PMID: 23483787 PMCID: PMC3589647 DOI: 10.3343/alm.2013.33.2.97] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 09/06/2012] [Accepted: 12/03/2012] [Indexed: 11/19/2022] Open
Abstract
Background Therapy-related myeloid neoplasms (t-MN) occur as late complications of cytotoxic therapy. This study reviewed clinical and cytogenetic characteristics of patients with t-MN at a single institution in Korea. Methods The study subjects included 39 consecutive patients diagnosed with t-MN. Each subject's clinical history of previous diseases, treatments, and laboratory data was reviewed, including cytogenetics. The primary diagnosis was hematologic malignancy in 14 patients and solid tumor in 25 patients. Results Therapy-related acute myeloid leukemia (t-AML, 66.7%) was found to be more common than therapy-related myelodysplastic syndrome (t-MDS). Primary hematologic malignancies that were commonly implicated included mature B-cell neoplasm and acute leukemia. Breast cancer was the most common primary solid tumor. The mean time interval from cytotoxic therapy initiation to t-MN detection was 49 months. Chromosomal aberrations were observed in 35 patients, and loss of chromosome 5, 7, or both accounted for 41% of all cases. Balanced rearrangements occurred in 13 patients; these patients showed shorter latency intervals (mean, 38 months) than patients with loss of chromosome 5 or 7 (mean, 61 months). Conclusions In this study, we determined the clinical and cytogenetic characteristics of Korean patients with t-MN. Although our results were generally consistent with those of previous reports, we found that t-MN resulting from de novo leukemia was common and that t-AML was more common than t-MDS at presentation. Multi-institutional studies involving a larger number of patients and additional parameters are required to investigate the epidemiology, genetic predisposition, and survival rate of t-MN in Korea.
Collapse
Affiliation(s)
- Hee Jae Huh
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Asou N, Iwanaga E, Nanri T, Mitsuya H. Successful treatment with low-dose imatinib mesylate of therapy-related myeloid neoplasm harboring TEL-PDGFRB in a patient with acute promyelocytic leukemia. Haematologica 2012; 95:e1. [PMID: 20807977 DOI: 10.3324/haematol.2010.27656] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
45
|
Larson RA. Cytogenetics, not just previous therapy, determines the course of therapy-related myeloid neoplasms. J Clin Oncol 2012; 30:2300-2. [PMID: 22585693 DOI: 10.1200/jco.2011.41.1215] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
46
|
Suvajdžić N, Cvetković Z, Dorđević V, Kraguljac-Kurtović N, Stanisavljević D, Bogdanović A, Djunić I, Colović N, Vidović A, Elezović I, Tomin D. Prognostic factors for therapy-related acute myeloid leukaemia (t-AML)--a single centre experience. Biomed Pharmacother 2012; 66:285-92. [PMID: 22401928 DOI: 10.1016/j.biopha.2011.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 12/23/2011] [Indexed: 12/15/2022] Open
Abstract
Prognostic parameters for treatment outcome in 42 consecutive patients with t-AML diagnosed and treated in a single centre between 2000-2010 (mean age: 56.07 years, range: 23-84; 30 females) were evaluated retrospectively/prospectively. Antecedent malignancy occurred in 37 patients (88.15%): 28 solid cancers (breast, n=14), nine haematological. History of previous chemotherapy (CT), radiotherapy (RT) alone and combined CT/RT was present in 42.9%, 6.19% and 30.1% patients, respectively. Primary disease was active in 11 patients (six relapsed or metastatic cancers; five autoimmune diseases). Myelodysplastic syndrome preceded t-AML in 29% of patients. Median latency period from prior CT/RT was 54.62 months (range: 6-243). Median WBC count was 27.23 × 10⁹/L, platelet count 62.29 × 10⁹/L, haemoglobin level 87.83 g/L, peripheral blood and bone marrow blast percentage 30.7% and 66.7% respectively, serum LDH 1216 U/L. Aberrant expression of B or T lymphoid markers was registered in seven out of 39 and six out of 39 patients, respectively. Aberrant karyotype was detected in 24 out of 33 (72.7%) of eligible patients: favourable: 15.2%, intermediate: 42.4% and unfavourable: 42.4%. Eastern Cooperative Oncology Group (ECOG) performance status greater or equal to 2 and Haematopoietic Cell Transplantation Specific Comorbidity Index (HCT-CI) greater or equal to 3 exhibited 83.3% and 76.2% patients, respectively. Intensive induction CT for t-AML was administered in 24 patients. The median follow-up and the median overall survival (OS) for the whole cohort were 2 months and 5.94 months (range: 0.5-34), respectively. In 10 patients (23.8%) achieving complete remission (CR), median disease free survival (DFS) was 11.8 months (range: 4-32). Only CD19 expression, pretreatment karyotype, ECOG PS, HCT-CI and activity of primary disease had impact on OS (P<0.05).
Collapse
Affiliation(s)
- Nada Suvajdžić
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Therapy-related myeloid malignancies in myeloma. Mediterr J Hematol Infect Dis 2011; 3:e2011047. [PMID: 22110897 PMCID: PMC3219649 DOI: 10.4084/mjhid.2011.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 10/14/2011] [Indexed: 12/22/2022] Open
Abstract
Therapy related myeloid malignancies are an increasingly recognized treatment complication in patients undergoing therapy for multiple myeloma. The main predisposing factors are the alkylating agents, topoisomerase II inhibitors and radiotherapy, but recently questions have been raised regarding the immunomodulatory agent lenalidomide. Little is known about the new antimyeloma agents in the context of therapy related myeloid malignancies. The duration of treatment and the time from diagnosis are the main contributing factors in alkylating induced myeloid malignancies which occur 5–10 years after treatment, chromosome 5 and 7 abnormalities being the characteristic finding. High dose therapy (HDT) does not seem to be a major contributing factor per se in multiple myeloma. In a number of large published series, all the factors related with therapy-induced myelodysplasia were defined prior to HDT. Topoisomerase II inhibitors induce mainly acute leukemias which invariably correlate with dysregulation of the MLL gene. Radiotherapy causes therapy related myelodysplasia if applied in bone marrow producing areas, especially if combined with chemotherapy. Therapy related myeloid malignancies generally herald a poor prognosis. Karyotypic abnormalities seem to be the main prognostic factor. In all cases the risk for therapy related myeloid malignancies drops sharply by 10 years after the treatment.
Collapse
|
48
|
Motlló C, Manuel Sancho J, García O, Granada I, Millá F, Ribera JM. Leucemias agudas secundarias a tratamiento con quimioterapia y/o radioterapia: estudio de 23 pacientes. Med Clin (Barc) 2011; 137:449-52. [DOI: 10.1016/j.medcli.2010.09.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 09/10/2010] [Accepted: 09/14/2010] [Indexed: 10/18/2022]
|
49
|
Komrokji RS, Corrales-Yepez M, Al Ali N, Kharfan-Dabaja M, Padron E, Fields T, Lancet JE, List AF. Validation of the MD Anderson Prognostic Risk Model for patients with myelodysplastic syndrome. Cancer 2011; 118:2659-64. [PMID: 21956402 DOI: 10.1002/cncr.26567] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 07/11/2011] [Accepted: 07/28/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND The proposed MD Anderson Risk Model Score (MDAS) for myelodysplastic syndromes (MDS) refines outcome discrimination compared with the International Prognostic Scoring System (IPSS). We applied the MDAS to Moffitt Cancer Center (MCC) MDS patients to validate its prognostic utility. METHODS This was a retrospective analysis of MDS cases, as defined by World Health Organization criteria, from the Moffitt database, with confirmatory chart review. RESULTS A total of 775 patients evaluated between January 2001 and December 2009 were included. Patients were reclassified by MDAS as low (20.6%), intermediate-1 (31%), intermediate-2 (21%), high risk (16.1%), and unknown (11.2%). Median overall survival (OS) from diagnosis was 92, 49, 27, and 14 months for low, intermediate-1, intermediate-2, and high-risk MDAS groups, respectively (P < .005). Median OS from referral was 61, 28, 15, and 8 months (P < .005), respectively. Among 484 patients classified by IPSS as low or intermediate-1 risk, 25% were up-staged as intermediate-2 or high risk by MDAS; 4 prognostically distinct subgroups were identified among lower risk IPSS categories with median OS of 93, 53, 31, and 18 months (P < .005). Among 201 intermediate-2 or high-risk IPSS patients, 15.4% were down-staged to intermediate-1 by MDAS, with 3 groups identified by MDAS having median OS of 33, 23, and 14 months. Acute myeloid leukemia transformation rate was highest among high-risk MDAS (50.4%). In Cox regression analysis, higher risk MDAS predicted inferior OS (hazard ratio 1.42; P < .005) independent of IPSS. CONCLUSIONS Our data validated MDAS' prognostic value. MDAS is complementary to IPSS and refines prognostic precision.
Collapse
Affiliation(s)
- Rami S Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Zinke-Cerwenka W, Valentin A, Posch U, Beham-Schmid C, Groselj-Strele A, Linkesch W, Wölfler A, Sill H. Reduced-intensity allografting in patients with therapy-related myeloid neoplasms and active primary malignancies. Bone Marrow Transplant 2011; 46:1540-4. [PMID: 21860429 DOI: 10.1038/bmt.2011.165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Therapy-related myeloid neoplasms (t-MNs) are severe long-term consequences of cytotoxic treatments for a primary, often, malignant disorder. So far, the majority of patients eligible for transplantation have undergone myeloablative allo haematopoietic SCT (HSCT) as a potentially curative treatment, but it has been associated with high transplantation-related mortality (TRM) rates. In this retrospective study, we analysed the outcome of patients with t-MNs undergoing HSCT with reduced-intensity conditioning (RIC). Of 55 patients, seen at a single centre over a 10-year period, 17 underwent RIC HSCT with related or unrelated donors. The estimated overall survival was 53% at 1 year and 47% at 3 years, and disease-free survival was 47% at 1 year. At 1 year, the cumulative incidence of relapse and TRM were 24% and 30%, respectively. Of five patients with active primary neoplasms who underwent transplantation, two are alive beyond 1 year and show CR of both t-MNs and the primary malignancy. These data indicate that RIC HSCT is an encouraging approach for patients with t-MNs. The issue of primary malignancies not being in remission at the time of transplantation should be explored in further studies.
Collapse
Affiliation(s)
- W Zinke-Cerwenka
- Division of Haematology, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | |
Collapse
|