351
|
Tsai FD, Lindsley RC. Clonal hematopoiesis in the inherited bone marrow failure syndromes. Blood 2020; 136:1615-1622. [PMID: 32736377 PMCID: PMC7530647 DOI: 10.1182/blood.2019000990] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/20/2020] [Indexed: 12/16/2022] Open
Abstract
Inherited bone marrow failure syndromes (IBMFSs) are characterized by ineffective hematopoiesis and increased risk for developing myeloid malignancy. The pathophysiologies of different IBMFSs are variable and can relate to defects in diverse biological processes, including DNA damage repair (Fanconi anemia), telomere maintenance (dyskeratosis congenita), and ribosome biogenesis (Diamond-Blackfan anemia, Shwachman-Diamond syndrome). Somatic mutations leading to clonal hematopoiesis have been described in IBMFSs, but the distinct mechanisms by which mutations drive clonal advantage in each disease and their associations with leukemia risk are not well understood. Clinical observations and laboratory models of IBMFSs suggest that the germline deficiencies establish a qualitatively impaired functional state at baseline. In this context, somatic alterations can promote clonal hematopoiesis by improving the competitive fitness of specific hematopoietic stem cell clones. Some somatic alterations relieve baseline fitness constraints by normalizing the underlying germline deficit through direct reversion or indirect compensation, whereas others do so by subverting senescence or tumor-suppressor pathways. Clones with normalizing somatic mutations may have limited transformation potential that is due to retention of functionally intact fitness-sensing and tumor-suppressor pathways, whereas those with mutations that impair cellular elimination may have increased risk for malignant transformation that is due to subversion of tumor-suppressor pathways. Because clonal hematopoiesis is not deterministic of malignant transformation, rational surveillance strategies will depend on the ability to prospectively identify specific clones with increased leukemic potential. We describe a framework by which an understanding of the processes that promote clonal hematopoiesis in IBMFSs may inform clinical surveillance strategies.
Collapse
Affiliation(s)
- Frederick D Tsai
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - R Coleman Lindsley
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
352
|
Bernard E, Nannya Y, Hasserjian RP, Devlin SM, Tuechler H, Medina-Martinez JS, Yoshizato T, Shiozawa Y, Saiki R, Malcovati L, Levine MF, Arango JE, Zhou Y, Solé F, Cargo CA, Haase D, Creignou M, Germing U, Zhang Y, Gundem G, Sarian A, van de Loosdrecht AA, Jädersten M, Tobiasson M, Kosmider O, Follo MY, Thol F, Pinheiro RF, Santini V, Kotsianidis I, Boultwood J, Santos FPS, Schanz J, Kasahara S, Ishikawa T, Tsurumi H, Takaori-Kondo A, Kiguchi T, Polprasert C, Bennett JM, Klimek VM, Savona MR, Belickova M, Ganster C, Palomo L, Sanz G, Ades L, Della Porta MG, Elias HK, Smith AG, Werner Y, Patel M, Viale A, Vanness K, Neuberg DS, Stevenson KE, Menghrajani K, Bolton KL, Fenaux P, Pellagatti A, Platzbecker U, Heuser M, Valent P, Chiba S, Miyazaki Y, Finelli C, Voso MT, Shih LY, Fontenay M, Jansen JH, Cervera J, Atsuta Y, Gattermann N, Ebert BL, Bejar R, Greenberg PL, Cazzola M, Hellström-Lindberg E, Ogawa S, Papaemmanuil E. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat Med 2020; 26:1549-1556. [PMID: 32747829 PMCID: PMC8381722 DOI: 10.1038/s41591-020-1008-z] [Citation(s) in RCA: 424] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 07/07/2020] [Indexed: 01/19/2023]
Abstract
Tumor protein p53 (TP53) is the most frequently mutated gene in cancer1,2. In patients with myelodysplastic syndromes (MDS), TP53 mutations are associated with high-risk disease3,4, rapid transformation to acute myeloid leukemia (AML)5, resistance to conventional therapies6-8 and dismal outcomes9. Consistent with the tumor-suppressive role of TP53, patients harbor both mono- and biallelic mutations10. However, the biological and clinical implications of TP53 allelic state have not been fully investigated in MDS or any other cancer type. We analyzed 3,324 patients with MDS for TP53 mutations and allelic imbalances and delineated two subsets of patients with distinct phenotypes and outcomes. One-third of TP53-mutated patients had monoallelic mutations whereas two-thirds had multiple hits (multi-hit) consistent with biallelic targeting. Established associations with complex karyotype, few co-occurring mutations, high-risk presentation and poor outcomes were specific to multi-hit patients only. TP53 multi-hit state predicted risk of death and leukemic transformation independently of the Revised International Prognostic Scoring System (IPSS-R)11. Surprisingly, monoallelic patients did not differ from TP53 wild-type patients in outcomes and response to therapy. This study shows that consideration of TP53 allelic state is critical for diagnostic and prognostic precision in MDS as well as in future correlative studies of treatment response.
Collapse
Affiliation(s)
- Elsa Bernard
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | | | - Sean M Devlin
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Juan S Medina-Martinez
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Yusuke Shiozawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Ryunosuke Saiki
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Hematology, IRCCS Fondazione Policlinico S. Matteo, Pavia, Italy
| | - Max F Levine
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan E Arango
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yangyu Zhou
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francesc Solé
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Barcelona, Spain
| | - Catherine A Cargo
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Detlef Haase
- Clinics of Hematology and Medical Oncology, University Medical Center, Göttingen, Germany
| | - Maria Creignou
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gunes Gundem
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Araxe Sarian
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Martin Jädersten
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Tobiasson
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Olivier Kosmider
- Department of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin and Université de Paris, Université Paris Descartes, Paris, France
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation Hannover Medical School, Hannover, Germany
| | - Ronald F Pinheiro
- Drug Research and Development Center, Federal University of Ceara, Ceara, Brazil
| | - Valeria Santini
- MDS Unit, Hematology, AOU Careggi, University of Florence, Florence, Italy
| | - Ioannis Kotsianidis
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| | - Jacqueline Boultwood
- Radcliffe Department of Medicine, University of Oxford and Oxford BRC Haematology Theme, Oxford, UK
| | - Fabio P S Santos
- Oncology-Hematology Center, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Julie Schanz
- Clinics of Hematology and Medical Oncology, University Medical Center, Göttingen, Germany
| | - Senji Kasahara
- Department of Hematology, Gifu Municipal Hospital, Gifu, Japan
| | - Takayuki Ishikawa
- Department of Hematology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hisashi Tsurumi
- Department of Hematology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toru Kiguchi
- Department of Hematology, Chugoku Central Hospital, Fukuyama, Japan
| | - Chantana Polprasert
- Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - John M Bennett
- Lab. Medicine and Pathology, Hematology/Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | - Virginia M Klimek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael R Savona
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Monika Belickova
- Department of Genomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Christina Ganster
- Clinics of Hematology and Medical Oncology, University Medical Center, Göttingen, Germany
| | - Laura Palomo
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Barcelona, Spain
| | - Guillermo Sanz
- Department of Hematology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Lionel Ades
- Department of Hematology, Hôpital St Louis and Paris University, Paris, France
| | - Matteo Giovanni Della Porta
- Humanitas Clinical and Research Center-IRCCS, Humanitas Cancer Center, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Harold K Elias
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Yesenia Werner
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Minal Patel
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Agnès Viale
- Integrated Genomics Operation, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katelynd Vanness
- Integrated Genomics Operation, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donna S Neuberg
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Kamal Menghrajani
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelly L Bolton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pierre Fenaux
- Department of Hematology, Hôpital St Louis and Paris University, Paris, France
| | - Andrea Pellagatti
- Radcliffe Department of Medicine, University of Oxford and Oxford BRC Haematology Theme, Oxford, UK
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation Hannover Medical School, Hannover, Germany
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Shigeru Chiba
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Carlo Finelli
- Institute of Hematology, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Maria Teresa Voso
- MDS Cooperative Group GROM-L, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Lee-Yung Shih
- Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan City, Taiwan
| | - Michaela Fontenay
- Department of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin and Université de Paris, Université Paris Descartes, Paris, France
| | - Joop H Jansen
- Laboratory Hematology, Department LABGK, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - José Cervera
- Department of Hematology and Genetics Unit, University Hospital La Fe, Valencia, Spain
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan
| | - Norbert Gattermann
- Department of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - Benjamin L Ebert
- Department of Medical Oncology and Howard Hughes Medical Institute, Dana-Farber Cancer Center, Boston, MA, USA
| | - Rafael Bejar
- UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | | | - Mario Cazzola
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Hematology, IRCCS Fondazione Policlinico S. Matteo, Pavia, Italy
| | - Eva Hellström-Lindberg
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Elli Papaemmanuil
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
353
|
Bejanyan N, Zhang M, Bo-Subait K, Brunstein C, Wang H, Warlick ED, Giralt S, Nishihori T, Martino R, Passweg J, Dias A, Copelan E, Hale G, Gale RP, Solh M, Kharfan-Dabaja MA, Diaz MA, Ganguly S, Gore S, Verdonck LF, Hossain NM, Kekre N, Savani B, Byrne M, Kanakry C, Cairo MS, Ciurea S, Schouten HC, Bredeson C, Munker R, Lazarus H, Cahn JY, van Der Poel M, Rizzieri D, Yared JA, Freytes C, Cerny J, Aljurf M, Palmisiano ND, Pawarode A, Bacher VU, Grunwald MR, Nathan S, Wirk B, Hildebrandt GC, Seo S, Olsson RF, George B, de Lima M, Hourigan CS, Sandmaier BM, Litzow M, Kebriaei P, Saber W, Weisdorf D. Myeloablative Conditioning for Allogeneic Transplantation Results in Superior Disease-Free Survival for Acute Myelogenous Leukemia and Myelodysplastic Syndromes with Low/Intermediate but not High Disease Risk Index: A Center for International Blood and Marrow Transplant Research Study. Transplant Cell Ther 2020; 27:68.e1-68.e9. [PMID: 33010430 DOI: 10.1016/j.bbmt.2020.09.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/05/2020] [Accepted: 09/19/2020] [Indexed: 11/26/2022]
Abstract
Compared with reduced-intensity conditioning (RIC), myeloablative conditioning (MAC) is generally associated with lower relapse risk after allogeneic hematopoietic cell transplantation (HCT) for acute myelogenous leukemia (AML) and myelodysplastic syndromes (MDS). However, disease-specific risk factors in AML/MDS can further inform when MAC and RIC may yield differential outcomes. We analyzed HCT outcomes stratified by the Disease Risk Index (DRI) in 4387 adults (age 40 to 65 years) to identify the impact of conditioning intensity. In the low/intermediate-risk DRI cohort, RIC was associated with lower nonrelapse mortality (NRM) (hazard ratio [HR], .74; 95% confidence interval [CI], .62 to .88; P < .001) but significantly greater relapse risk (HR, 1.54; 95% CI, 1.35 to 1.76; P < .001) and thus inferior disease-free survival (DFS) (HR, 1.19; 95% CI, 1.07 to 1.33; P = .001). In the high/very high-risk DRI cohort, RIC was associated with marginally lower NRM (HR, .83; 95% CI, .68 to 1.00; P = .051) and significantly higher relapse risk (HR, 1.23; 95% CI, 1.08 to 1.41; P = .002), leading to similar DFS using either RIC or MAC. These data support MAC over RIC as the preferred conditioning intensity for patients with AML/MDS with low/intermediate-risk DRI, but with a similar benefit as RIC in high/very high-risk DRI. Novel MAC regimens with less toxicity could benefit all patients, but more potent antineoplastic approaches are needed for the high/very-high risk DRI group.
Collapse
Affiliation(s)
- Nelli Bejanyan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.
| | - Meijie Zhang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Khalid Bo-Subait
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Claudio Brunstein
- Adult Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Hailin Wang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Erica D Warlick
- Adult Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Sergio Giralt
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Rodrigo Martino
- Division of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jakob Passweg
- Division of Hematology, Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Ajoy Dias
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Edward Copelan
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Gregory Hale
- Department of Hematology/Oncology, Johns Hopkins All Children's Hospital, St Petersburg, Florida
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Melhem Solh
- Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Miguel Angel Diaz
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Siddhartha Ganguly
- Division of Hematological Malignancy and Cellular Therapeutics, University of Kansas Health System, Kansas City, Kansas
| | - Steven Gore
- Section of Medical Oncology, Department of Internal Medicine, Yale New Haven Hospital, New Haven, Connecticut
| | - Leo F Verdonck
- Department of Hematology/Oncology, Isala Clinic, Zwolle, The Netherland
| | - Nasheed M Hossain
- Stem Cell Transplant Program, Division of Hematology/Oncology, Department of Medicine, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Natasha Kekre
- Blood & Marrow Transplant Program, Department of Medicine, Ottawa Hospital Ottawa, Ontario, Canada
| | - Bipin Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Byrne
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christopher Kanakry
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mitchell S Cairo
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, New York Medical College, Valhalla, New York
| | - Stefan Ciurea
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Harry C Schouten
- Department of Hematology, Academische Ziekenhuis, Maastricht, The Netherlands
| | - Christopher Bredeson
- Blood & Marrow Transplant Program, Department of Medicine, Ottawa Hospital Ottawa, Ontario, Canada
| | - Reinhold Munker
- Division of Medical Oncology, Markey Cancer Center, University of Kentucky School of Medicine, Lexington, Kentucky
| | - Hillard Lazarus
- Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Jean-Yves Cahn
- Department of Hematology, CHU Grenoble Alpes, Grenoble, France
| | - Marjolein van Der Poel
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - David Rizzieri
- Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, North Carolina
| | - Jean A Yared
- Blood & Marrow Transplantation Program, Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Cesar Freytes
- Hematopoietic Stem Cell Transplant Program, Texas Transplant Institute, San Antonio, Texas
| | - Jan Cerny
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical Center, Worcester, Massachusetts
| | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital Center & Research, Riyadh, Saudi Arabia
| | - Neil D Palmisiano
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Vera Ulrike Bacher
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Sunita Nathan
- Section of Bone Marrow Transplantation and Cellular Therapy, Division of Hematology, Oncology and Cell Therapy, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | - Baldeep Wirk
- Bone Marrow Transplant Program, Penn State Cancer Institute, Hershey, Pennsylvania
| | - Gerhard C Hildebrandt
- Division of Medical Oncology, Markey Cancer Center, University of Kentucky School of Medicine, Lexington, Kentucky
| | - Sachiko Seo
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | - Richard F Olsson
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Clinical Research Sormland, Uppsala University, Sweden
| | - Biju George
- Department of Haematology, Christian Medical College, Vellore, India
| | - Marcos de Lima
- Department of Medicine, Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Brenda M Sandmaier
- Division of Medical Oncology, University of Washington and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark Litzow
- Division of Hematology and Transplant Center, Mayo Clinic Rochester, Rochester, Minnesota
| | - Partow Kebriaei
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wael Saber
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daniel Weisdorf
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, 96
| |
Collapse
|
354
|
Neuendorff NR, Frenzel LP, Leuschner F, Fremd C. Integrating clonal haematopoiesis into geriatric oncology: The ARCH between aging, cardiovascular disease and malignancy. J Geriatr Oncol 2020; 12:479-482. [PMID: 32978103 DOI: 10.1016/j.jgo.2020.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/27/2020] [Accepted: 09/01/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Nina Rosa Neuendorff
- Medical Department V - Haematology, Oncology and Rheumatology, University Hospital Heidelberg, Im Neuenheimer Feld 410, Heidelberg, Germany.
| | - Lukas Peter Frenzel
- Department of Internal Medicine I, Center of Integrated Oncology Cologne-Bonn, University of Cologne, 50937 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Florian Leuschner
- Medical Department III - Cardiology, Angiology and Intensive Care, University Hospital Heidelberg, Im Neuenheimer Feld 410, Heidelberg, Germany
| | - Carlo Fremd
- National Center for Tumour Diseases, Department of Medical Oncology, University of Heidelberg, Im Neuenheimer Feld 460, Heidelberg, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 260, Heidelberg, Germany; Medical Department VI - Medical Oncology, University Hospital Heidelberg, Im Neuenheimer Feld 460, Heidelberg, Germany
| |
Collapse
|
355
|
Miller PG, Gibson CJ, Mehta A, Sperling AS, Frederick DT, Manos MP, Miao B, Hacohen N, Hodi FS, Boland GM, Ebert BL. Fitness Landscape of Clonal Hematopoiesis Under Selective Pressure of Immune Checkpoint Blockade. JCO Precis Oncol 2020; 4:2000186. [PMID: 33015529 DOI: 10.1200/po.20.00186] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2020] [Indexed: 02/01/2023] Open
Abstract
PURPOSE Conventional cytotoxic therapies increase the risk of clonal hematopoiesis and select for TP53-mutant clones, which carry a high risk for transformation to therapy-related myelodysplastic neoplasms. In contrast, the effect of immune checkpoint blockade (ICB) on clonal hematopoiesis is unknown. METHODS Paired peripheral-blood samples taken before and after treatment with ICB were obtained for 91 patients with either cutaneous melanoma or basal cell carcinoma. Error-corrected sequencing of a targeted panel of genes recurrently mutated in clonal hematopoiesis was performed on peripheral-blood genomic DNA. RESULTS The average interval between acquisition of the paired samples was 180 days. Forty-one percent of the patients had clonal hematopoiesis at a variant allele frequency (VAF) > 0.01 in the pretreatment sample. There was near-complete agreement in the distribution and burden of clonal hematopoiesis mutations in the paired blood samples, with 87 of 88 mutations identified across the cohort present in paired samples, regardless of the duration between sample collection. The VAF in the paired samples also showed a high correlation, with an R 2 = 0.95 (P < .0001). In contrast to cytotoxic therapy, exposure to ICB did not lead to selection of TP53- or PPM1D-mutant clones. However, consistent with the known effects of DNA-damaging therapy, we identified one patient who had eight unique TP53 mutations in the posttreatment blood sample after receiving two courses of radiation therapy. CONCLUSION There was no expansion of hematopoietic clones or selection for clones at high risk for malignant transformation in patients who received ICB, observations that warrant further validation in larger cohorts. These findings highlight an important difference between ICB and conventional cytotoxic therapies and their respective impacts on premalignant genetic lesions.
Collapse
Affiliation(s)
- Peter G Miller
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.,Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Christopher J Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.,Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Arnav Mehta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.,Massachusetts General Hospital Cancer Center, Boston, MA
| | - Adam S Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.,Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | | | - Michael P Manos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Benchun Miao
- Massachusetts General Hospital Cancer Center, Boston, MA
| | - Nir Hacohen
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA.,Massachusetts General Hospital Cancer Center, Boston, MA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Genevieve M Boland
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA.,Massachusetts General Hospital Cancer Center, Boston, MA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.,Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA.,Howard Hughes Medical Institute, Dana-Farber Cancer Institute
| |
Collapse
|
356
|
Konuma T, Ishiyama K, Igarashi A, Uchida N, Ozawa Y, Fukuda T, Ueda Y, Matsuoka KI, Mori T, Katayama Y, Onizuka M, Ichinohe T, Atsuta Y. Effects of Acute and Chronic Graft-versus-myelodysplastic Syndrome on Long-term Outcomes Following Allogeneic Hematopoietic Cell Transplantation. Clin Cancer Res 2020; 26:6483-6493. [PMID: 32895232 DOI: 10.1158/1078-0432.ccr-20-1104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/11/2020] [Accepted: 08/31/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Potent graft-versus-tumor (GVT) effects associated with graft-versus-host disease (GVHD) might be dependent on hematologic disease type and status. However, the data regarding the impact of GVHD on transplant outcomes for patients with myelodysplastic syndrome (MDS) are limited. EXPERIMENTAL DESIGN We retrospectively evaluated the impact of acute and chronic GVHD on transplant outcomes for a large cohort of adult patients with a low-risk (n = 1,193) and high-risk (n = 1,926) MDS treated by first allogeneic hematopoietic cell transplantation between 2001 and 2017. RESULTS The multivariate analysis, in which development of GVHD was treated as a time-dependent covariate, showed that acute and chronic GVHD at any grade or severity did not improve overall mortality, relapse, or nonrelapse mortality (NRM) in low-risk MDS. For patients with high-risk MDS, development of limited chronic GVHD was significantly associated with lower overall mortality [HR, 0.66; 95% confidence interval (CI), 0.50-0.86; P = 0.002]. This is probably due to that the reduced risk of relapse with grade III-IV acute GVHD (HR, 0.41; 95% CI, 0.25-0.65; P = 0.0002), or limited (HR, 0.57; 95% CI, 0.39-0.83; P = 0.003) or extensive (HR, 0.56; 95% CI, 0.41-0.77; P = 0.0004) chronic GVHD was offset by increased NRM with grade III-IV acute GVHD or extensive chronic GVHD in high-risk MDS. CONCLUSIONS These data demonstrated a survival benefit of the graft-versus-MDS effect is present only in high-risk MDS patients with limited chronic GVHD.See related commentary by Eckel and Deeg, p. 6404.
Collapse
Affiliation(s)
- Takaaki Konuma
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Ken Ishiyama
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Japan
| | - Aiko Igarashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Naoyuki Uchida
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Yukiyasu Ozawa
- Department of Hematology, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Yasunori Ueda
- Department of Hematology and Oncology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Takehiko Mori
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Katayama
- Department of Hematology, Hiroshima Red Cross Hospital & Atomic-Bomb Survivors Hospital, Hiroshima, Japan
| | - Makoto Onizuka
- Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | | |
Collapse
|
357
|
Bewersdorf JP, Shallis RM, Gowda L, Wei W, Hager K, Isufi I, Kim TK, Pillai MM, Seropian S, Podoltsev NA, Gore SD, Siddon AJ, Zeidan AM. Clinical outcomes and characteristics of patients with TP53-mutated acute myeloid leukemia or myelodysplastic syndromes: a single center experience. Leuk Lymphoma 2020; 61:2180-2190. [PMID: 32362171 PMCID: PMC7603787 DOI: 10.1080/10428194.2020.1759051] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/13/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
Mutations in the tumor suppressor gene TP53 are detected in 5-10% of patients with acute myeloid leukemia (AML) and myelodysplastic syndromes. TP53 mutations have been associated with complex karyotypes, therapy-related malignancies, lower response rates to cytotoxic chemotherapy, and an overall adverse prognosis. In this single-center retrospective study, we analyzed the clinicopathologic characteristics and outcomes of 83 patients with TP53-mutated myeloid malignancies treated at Yale Cancer Center between 9/2015 and 5/2019. Complex karyotypes (n = 75; 90%) and therapy-related malignancies (n = 32; 39%) were common. Median overall survival (OS) was 7.6 months. Intensive chemotherapy did not improve OS compared to lower-intensity treatment for AML patients. Patients who underwent allogeneic hematopoietic stem cell transplant (alloHSCT) had a significantly longer median OS, despite relatively limited follow-up. In conclusion, our data confirm the limited efficacy of intensive chemotherapy approaches for TP53-mutated patients with myeloid neoplasms and suggest that a minority of patients achieve long-term survival with alloHSCT.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Rory M. Shallis
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Lohith Gowda
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Wei Wei
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Karl Hager
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Iris Isufi
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Tae Kon Kim
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
- Division of Hematology/Oncology, Vanderbilt University Medical Center
| | - Manoj M. Pillai
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Stuart Seropian
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Nikolai A. Podoltsev
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Steven D. Gore
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| | - Alexa J. Siddon
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Section of Hematopathology, Yale School of Medicine, New Haven, CT, USA
| | - Amer M. Zeidan
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| |
Collapse
|
358
|
Hunter AM, Padron E. Molecular genetics of MDS/MPN overlap syndromes. Best Pract Res Clin Haematol 2020; 33:101195. [DOI: 10.1016/j.beha.2020.101195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/27/2020] [Indexed: 01/05/2023]
|
359
|
Do next-generation sequencing results drive diagnostic and therapeutic decisions in MDS? Blood Adv 2020; 3:3449-3453. [PMID: 31714960 DOI: 10.1182/bloodadvances.2018022434] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/12/2018] [Indexed: 01/01/2023] Open
Abstract
This article has a companion Counterpoint by Sanz et al.
Collapse
|
360
|
Byrne M, Danielson N, Sengsayadeth S, Rasche A, Culos K, Gatwood K, Wyatt H, Chinratanalab W, Dholaria B, Ferrell PB, Fogo K, Goodman S, Jagasia M, Jayani R, Kassim A, Mohan SR, Savani BN, Strickland SA, Engelhardt BG, Savona M. The use of venetoclax-based salvage therapy for post-hematopoietic cell transplantation relapse of acute myeloid leukemia. Am J Hematol 2020; 95:1006-1014. [PMID: 32390196 DOI: 10.1002/ajh.25859] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/26/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022]
Abstract
For patients with high risk myeloid disease, allogeneic hematopoietic cell transplantation (HCT) is the only potentially curative therapy. Unfortunately, many of these patients relapse after HCT and have a limited survival. The recent approval of venetoclax, an orally bioavailable BCL-2 inhibitor, resulted in significant responses in treatment naïve acute myeloid leukemia (AML), and off-label use in the relapsed/refractory setting is increasing. We report the outcomes of 21 patients who underwent allogeneic HCT for myeloid disease, relapsed with AML, and were treated with venetoclax. Several patients had poor risk features including antecedent hematologic malignancy (6/21), complex karyotype (6/21), and TP53 mutations (5/21). The median age was 64.5 years and time from HCT to relapse was 5.7 months (range: 0.9 to 44.9 months). Of the 19 patients who were assessed for response, there were meaningful treatment responses seen in eight patients: five CR, three CRi, zero PR, for an ORR of 42.1%. Treatment effect was seen in six additional patients, including four in the morphologic leukemia-free state. Nine patients maintained their response for ≥3 months and eight were receiving therapy at data cut. Post-HCT AML relapse has an exceedingly poor outcome, and venetoclax-based therapy is a potent therapy option that should be studied prospectively in this setting.
Collapse
Affiliation(s)
- Michael Byrne
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | | | - Salyka Sengsayadeth
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Tennessee Valley Healthcare System Nashville Tennessee USA
| | - Adrianne Rasche
- Department of Nursing Vanderbilt University Medical Center Nashville Tennessee USA
| | - Katie Culos
- Department of Pharmacy Vanderbilt University Medical Center Nashville Tennessee USA
| | - Katie Gatwood
- Department of Pharmacy Vanderbilt University Medical Center Nashville Tennessee USA
| | - Houston Wyatt
- Department of Pharmacy Vanderbilt University Medical Center Nashville Tennessee USA
| | - Wichai Chinratanalab
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Tennessee Valley Healthcare System Nashville Tennessee USA
| | - Bhagirathbhai Dholaria
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - P. Brent Ferrell
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Kristin Fogo
- Department of Nursing Vanderbilt University Medical Center Nashville Tennessee USA
| | - Stacey Goodman
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Tennessee Valley Healthcare System Nashville Tennessee USA
| | - Madan Jagasia
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Reena Jayani
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Adetola Kassim
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Sanjay R. Mohan
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Bipin N. Savani
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Stephen A. Strickland
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Brian G. Engelhardt
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
| | - Michael Savona
- Department of Medicine Vanderbilt University School of Medicine Nashville Tennessee USA
- Vanderbilt‐Ingram Cancer Center Nashville Tennessee USA
- Program in Cancer Biology Vanderbilt University Nashville Tennessee USA
| |
Collapse
|
361
|
Sallman DA. The Problem of TP53-Mutant MDS/AML. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20 Suppl 1:S65-S66. [PMID: 32862873 DOI: 10.1016/s2152-2650(20)30465-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- David A Sallman
- Malignant Hematology Department, H. Lee Moffitt Cancer Center, Tampa, Florida, United States.
| |
Collapse
|
362
|
Comorbid and inflammatory characteristics of genetic subtypes of clonal hematopoiesis. Blood Adv 2020; 3:2482-2486. [PMID: 31434682 DOI: 10.1182/bloodadvances.2018024729] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 07/04/2019] [Indexed: 01/13/2023] Open
Abstract
Key Points
CH may be associated with broader ill health (worse performance status, increased and potentially novel comorbidities). Serum interleukin-6 is elevated in people with CH and genetic subtypes, providing a view of the human systemic inflammatory landscape of CH.
Collapse
|
363
|
Nazha A, Hu ZH, Wang T, Lindsley RC, Abdel-Azim H, Aljurf M, Bacher U, Bashey A, Cahn JY, Cerny J, Copelan E, DeFilipp Z, Diaz MA, Farhadfar N, Gadalla SM, Gale RP, George B, Gergis U, Grunwald MR, Hamilton B, Hashmi S, Hildebrandt GC, Inamoto Y, Kalaycio M, Kamble RT, Kharfan-Dabaja MA, Lazarus HM, Liesveld JL, Litzow MR, Majhail NS, Murthy HS, Nathan S, Nishihori T, Pawarode A, Rizzieri D, Sabloff M, Savani BN, Schachter L, Schouten HC, Seo S, Shah NN, Solh M, Valcárcel D, Vij R, Warlick E, Wirk B, Wood WA, Yared JA, Alyea E, Popat U, Sobecks RM, Scott BL, Nakamura R, Saber W. A Personalized Prediction Model for Outcomes after Allogeneic Hematopoietic Cell Transplant in Patients with Myelodysplastic Syndromes. Biol Blood Marrow Transplant 2020; 26:2139-2146. [PMID: 32781289 DOI: 10.1016/j.bbmt.2020.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/21/2020] [Accepted: 08/01/2020] [Indexed: 01/01/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (HCT) remains the only potentially curative option for myelodysplastic syndromes (MDS). Mortality after HCT is high, with deaths related to relapse or transplant-related complications. Thus, identifying patients who may or may not benefit from HCT is clinically important. We identified 1514 patients with MDS enrolled in the Center for International Blood and Marrow Transplant Research Registry and had their peripheral blood samples sequenced for the presence of 129 commonly mutated genes in myeloid malignancies. A random survival forest algorithm was used to build the model, and the accuracy of the proposed model was assessed by concordance index. The median age of the entire cohort was 59 years. The most commonly mutated genes were ASXL1(20%), TP53 (19%), DNMT3A (15%), and TET2 (12%). The algorithm identified the following variables prior to HCT that impacted overall survival: age, TP53 mutations, absolute neutrophils count, cytogenetics per International Prognostic Scoring System-Revised, Karnofsky performance status, conditioning regimen, donor age, WBC count, hemoglobin, diagnosis of therapy-related MDS, peripheral blast percentage, mutations in RAS pathway, JAK2 mutation, number of mutations/sample, ZRSR2, and CUX1 mutations. Different variables impacted the risk of relapse post-transplant. The new model can provide survival probability at different time points that are specific (personalized) for a given patient based on the clinical and mutational variables that are listed above. The outcomes' probability at different time points may aid physicians and patients in their decision regarding HCT.
Collapse
Affiliation(s)
| | - Zhen-Huan Hu
- Department of Medicine, CIBMTR (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Tao Wang
- Department of Medicine, CIBMTR (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Hisham Abdel-Azim
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Ulrike Bacher
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Asad Bashey
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, Georgia
| | - Jean-Yves Cahn
- Department of Hematology, CHU Grenoble Alpes, Grenoble, France
| | - Jan Cerny
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Medical Center, Worcester, Masschusetts
| | - Edward Copelan
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Zachariah DeFilipp
- Blood and Marrow Transplantation Program, Massachusetts General Hospital, Boston, Massachusetts
| | - Miguel Angel Diaz
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Nosha Farhadfar
- Division of Hematology/Oncology, University Florida College of Medicine, Gainesville, Florida
| | - Shahinaz M Gadalla
- Division of Cancer Epidemiology & Genetics, NIH-NCI Clinical Genetics Branch, Rockville, Maryland
| | - Robert Peter Gale
- Hematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Usama Gergis
- Hematologic Malignancies & Bone Marrow Transplant, Department of Medical Oncology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Betty Hamilton
- Blood & Marrow Transplant Program, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Shahrukh Hashmi
- Department of Oncology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia; Department of Internal Medicine, Mayo Clinic, Minnesota, Rochester
| | | | - Yoshihiro Inamoto
- Division of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | | | - Rammurti T Kamble
- Division of Hematology and Oncology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Mohamed A Kharfan-Dabaja
- Divison of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | | | - Jane L Liesveld
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Mark R Litzow
- Division of Hematology and Transplant Center, Mayo Clinic Rochester, Rochester, Minnesota
| | - Navneet S Majhail
- Blood & Marrow Transplant Program, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Hemant S Murthy
- Divison of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | | | - Taiga Nishihori
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, Florida
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - David Rizzieri
- Divison of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, North Carolina
| | - Mitchell Sabloff
- Division of Hematology, Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Harry C Schouten
- Department of Hematology, Academische Ziekenhuis, Maastricht, Netherlands
| | - Sachiko Seo
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | - Nirav N Shah
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia
| | - David Valcárcel
- Department of Hematology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Ravi Vij
- Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Erica Warlick
- University of Minnesota Blood and Marrow Transplant Program, Minneapolis, Minnesota
| | - Baldeep Wirk
- Penn State Cancer Institute, Bone Marrow Transplant Program, Hershey, Pennsylvania
| | - William A Wood
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Jean A Yared
- Blood & Marrow Transplantation Program, Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Edwin Alyea
- Center of Hematologic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Bart L Scott
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ryotaro Nakamura
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Wael Saber
- Department of Medicine, CIBMTR (Center for International Blood and Marrow Transplant Research), Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
364
|
Najima Y, Sadato D, Harada Y, Oboki K, Hirama C, Toya T, Doki N, Haraguchi K, Yoshifuji K, Akiyama M, Inamoto K, Igarashi A, Kobayashi T, Kakihana K, Okuyama Y, Sakamaki H, Harada H, Ohashi K. Prognostic impact of TP53 mutation, monosomal karyotype, and prior myeloid disorder in nonremission acute myeloid leukemia at allo-HSCT. Bone Marrow Transplant 2020; 56:334-346. [PMID: 32760007 DOI: 10.1038/s41409-020-01016-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 07/09/2020] [Accepted: 07/24/2020] [Indexed: 11/09/2022]
Abstract
Outcomes after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in nonremission acute myeloid leukemia (AML) are dismal [2-year overall survival (OS): 20-30%]. Though several risk classifications have been used, some factors are unavailable until the start of conditioning or transplantation. We analyzed prognostic gene mutations by targeted next-generation sequencing to identify predisposing factors for predicting OS at 1 month before transplantation. We enrolled 120 patients with nonremission AML who underwent first allo-HSCT between 2005 and 2018. Mutations were found in 98 patients; frequently mutated genes were FLT3-ITD, TP53, RUNX1, and WT1. TP53 mutation was detected in 21 patients and was the only predictor of poor OS. Multivariate analysis using Cox regression hazard model revealed primary AML, monosomal karyotype (MK), and TP53 mutation as independent factors for predicting poor OS. Based on these, patients were stratified into three groups. The low-risk group included patients with prior myeloid disorder without MK (n = 26). Among the rest, patients with TP53 mutation were assigned to the high-risk group (n = 19) and the rest into the intermediate-risk group (n = 75). Two-year OS in low-, intermediate-, and high-risk groups differed significantly (50.0%, 24.9%, and 0%, respectively). This suggests that the indication of allo-HSCT should be carefully judged for high-risk patients.
Collapse
Affiliation(s)
- Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Daichi Sadato
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan.,Clinical Research Support Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan.,Center for Medical Research Cooperation, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yuka Harada
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan. .,Clinical Research Support Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan.
| | - Keisuke Oboki
- Center for Medical Research Cooperation, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chizuko Hirama
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan.,Clinical Research Support Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan.,Center for Medical Research Cooperation, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Toya
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kyoko Haraguchi
- Division of Transfusion and Cell Therapy, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kota Yoshifuji
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Megumi Akiyama
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kyoko Inamoto
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Aiko Igarashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Takeshi Kobayashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kazuhiko Kakihana
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Yoshiki Okuyama
- Division of Transfusion and Cell Therapy, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hisashi Sakamaki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hironori Harada
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan.,Laboratory of Oncology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kazuteru Ohashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| |
Collapse
|
365
|
Arslan S, Nakamura R. Decision Analysis of Transplantation for Patients with Myelodysplasia: "Who Should We Transplant Today?". Curr Hematol Malig Rep 2020; 15:305-315. [PMID: 32222884 PMCID: PMC8080957 DOI: 10.1007/s11899-020-00573-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Myelodysplastic syndrome (MDS) is a heterogeneous hematological disorder characterized by a spectrum of clinical presentation, cytogenetic, and somatic gene mutations and the risk of transformation to acute leukemia. Management options include observation, supportive care, blood transfusion, administration of growth factors and/or hypomethylating agents, and hematopoietic cell transplant (HCT) either upfront or after disease progression. RECENT FINDINGS Currently, HCT is the only curative therapy available for patients with MDS, with multiple factors such as donor availability, patient, and disease characteristics being involved in making the decision to proceed with transplant. In this article, we summarize (1) overall prognosis and natural history of MDS, (2) currently available non-HCT therapy with a focus on hypomethylating agents (HMA), (3) outcomes after HCT in patients with MDS, (4) factors to be considered to proceed to HCT for treatment of MDS, and (5) more recent/ongoing studies relevant to HCT decision-making processes.
Collapse
Affiliation(s)
- Shukaib Arslan
- Department of Hematology & Hematopoietic Cell Transplant, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Ryotaro Nakamura
- Department of Hematology & Hematopoietic Cell Transplant, City of Hope National Medical Center, Duarte, CA, 91010, USA.
| |
Collapse
|
366
|
Prognostic significance of serial molecular annotation in myelodysplastic syndromes (MDS) and secondary acute myeloid leukemia (sAML). Leukemia 2020; 35:1145-1155. [PMID: 32728186 DOI: 10.1038/s41375-020-0997-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 07/01/2020] [Accepted: 07/21/2020] [Indexed: 01/07/2023]
Abstract
The implementation of next-generation sequencing (NGS) has influenced diagnostic, prognostic, and therapeutic decisions in myeloid malignancies. However, the clinical relevance of serial molecular annotation in patients with myelodysplastic syndrome (MDS) undergoing active treatment is unknown. MDS or secondary acute myeloid leukemia (sAML) patients who had at least two NGS assessments were identified. Outcomes according to mutation clearance (NGS-) on serial assessment were investigated. Univariate and multivariate Cox regression models were used to evaluate the prognostic impact of NGS trajectory on overall survival (OS). A total of 157 patients (MDS [n = 95]; sAML [n = 52]; CMML [n = 10]) were identified, with 93% of patients receiving treatment between NGS assessments. Magnitude of VAF delta from baseline was significantly associated with quality of response to treatment. Patients achieving NGS- had significantly improved OS compared to patients with mutation persistence (median OS not reached vs. 18.5 months; P = 0.002), which was confirmed in multivariate analysis (HR,0.14; 95%CI = 0.03-0.56; P = 0.0064). Serial TP53 VAF evaluation predicts outcomes with TP53 clearance representing an independent covariate for superior OS (HR,0.22; 95%CI = 0.05-0.99; P = 0.048). Collectively, our study highlights the clinical value of serial NGS during treatment and warrants prospective validation of NGS negativity as a biomarker for treatment outcome.
Collapse
|
367
|
Deng J, Wu X, Ling Y, Liu X, Zheng X, Ye W, Gong Y. The prognostic impact of variant allele frequency (VAF) in TP53 mutant patients with MDS: A systematic review and meta-analysis. Eur J Haematol 2020; 105:524-539. [PMID: 32621334 DOI: 10.1111/ejh.13483] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Tumor protein p53 (TP53) is frequently expressed in patients with myelodysplastic syndromes (MDS). Studies have already reported the poor prognostic impact of TP53 gene mutations in MDS patients. However, parts of this subgroup of patients with low-risk MDS still have relatively better survival and longer remission times. Therefore, we performed a meta-analysis to evaluate the prognostic difference intra-gene of variant allele frequency (VAF). The primary endpoint was overall survival (OS), and event-free survival (EFS) was selected as the secondary endpoint. We extracted the hazard ratio (HR) and 95% confidence interval (CI) for OS and EFS from univariate and multivariate Cox proportional hazard models. A total of 4003 MDS patients and 1278 TP53-mutated patients from 13 cohorts of 11 studies up to February 24, 2020, were included in our meta-analysis. Pooled HRs suggested that a high mutant VAF had an adverse impact on OS (HR = 2.11, 95% CI: 1.48-3.01, P < .0001) but no impact on EFS (HR = 15.57, 95% CI: 0.75-324.44, P = .003) in MDS patients. Twenty percent is a proper threshold to set (HR = 2.02, 95% CI: 1.31-3.13, P = .001) and is a rough line between high clone burden and low clone burden, while 40% is an exact cutoff point (HR = 2.11, 95% CI: 1.26-3.55, P < .0001) to guide diagnosis and treatment. Beyond the traditional binary classification of gene mutation, we aimed to find a way to divide mutant molecular markers more specifically by VAF to provide clinical therapeutic values. Our meta-analysis indicates that a high VAF is an independent, adverse prognostic factor for OS in TP53 mutant MDS patients. Patients with mediate/low-frequency parts who could be treated like wide-type patients have relatively better survival and may choose allogeneic hematopoietic stem cell transplantation as conditions permitting. Further prospective studies are needed in the future, and a large subgroup analysis of the same cutoff point subgroups is needed to obtain a more reliable basis for the impact of other mutant gene VAFs on the prognosis of MDS.
Collapse
Affiliation(s)
- Jili Deng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Wu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yantao Ling
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyan Liu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Zheng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Wu Ye
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuping Gong
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
368
|
Patel NM, Jo H, Eberhard DA, Yin X, Hayward MC, Stein MK, Hayes DN, Grilley-Olson JE. Improved Tumor Purity Metrics in Next-generation Sequencing for Clinical Practice: The Integrated Interpretation of Neoplastic Cellularity and Sequencing Results (IINCaSe) Approach. Appl Immunohistochem Mol Morphol 2020; 27:764-772. [PMID: 30102605 PMCID: PMC6887630 DOI: 10.1097/pai.0000000000000684] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/08/2018] [Indexed: 12/18/2022]
Abstract
Neoplastic cellularity contributes to the analytic sensitivity of most present technologies for mutation detection, such that they underperform when stroma and inflammatory cells dilute a cancer specimen's variant fraction. Thus, tumor purity assessment by light microscopy is used to determine sample adequacy before sequencing and to interpret the significance of negative results and mutant allele fraction afterwards. However, pathologist estimates of tumor purity are imprecise and have limited reproducibility. With the advent of massively parallel sequencing, large amounts of molecular data can be analyzed by computational purity algorithms. We retrospectively compared tumor purity of 3 computational algorithms with neoplastic cellularity using hematoxylin and eosin light microscopy to determine which was best for clinical evaluation of molecular profiling. Data were analyzed from 881 cancer patients from a clinical trial cohort, LCCC1108 (UNCseq), whose tumors had targeted massively parallel sequencing. Concordance among algorithms was poor, and the specimens analyzed had high rates of algorithm failure partially due to variable tumor purity. Computational tumor purity estimates did not add value beyond the pathologist's estimate of neoplastic cellularity microscopy. To improve present methods, we propose a semiquantitative, clinically applicable strategy based on mutant allele fraction and copy number changes present within a given specimen, which when combined with the morphologic tumor purity estimate, guide the interpretation of next-generation sequencing results in cancer patients.
Collapse
Affiliation(s)
- Nirali M. Patel
- Department of Pathology and Laboratory Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Heejoon Jo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - David A. Eberhard
- Department of Pathology and Laboratory Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Xiaoying Yin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Michele C. Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Matthew K. Stein
- Department of Internal Medicine, Division of Hematology and Oncology
- West Cancer Center, University of Tennessee Health Science Center, Memphis, TN
| | - David Neil Hayes
- Department of Internal Medicine, Division of Hematology and Oncology
- West Cancer Center, University of Tennessee Health Science Center, Memphis, TN
| | - Juneko E. Grilley-Olson
- Department of Internal Medicine, Division of Hematology and Oncology, University of North Carolina School of Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
369
|
Robin M, Fenaux P. Which lower risk myelodysplastic syndromes should be treated with allogeneic hematopoietic stem cell transplantation? Leukemia 2020; 34:2552-2560. [PMID: 32661295 DOI: 10.1038/s41375-020-0967-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022]
Abstract
Indications of allogeneic hematopoietic stem cell transplantation (HSCT) remain controversial in patients with lower risk myelodysplastic syndrome. We review prognostic factors in lower risk MDS, delineating patients with relatively poor risk who may potentially benefit from HSCT during the disease course. Results of HSCT in those patients, and main efforts to decrease non-relapse mortality (NRM) are detailed. Prospective studies are needed to determine more precisely which lower risk MDS patients may benefit from transplantation.
Collapse
Affiliation(s)
- Marie Robin
- Service d'hématologie-greffe, hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP) and Université de Paris, Paris, France.
| | - Pierre Fenaux
- Service d'hématologie-sénior, hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP) and Université de Paris, Paris, France
| |
Collapse
|
370
|
Metal-dependent Ser/Thr protein phosphatase PPM family: Evolution, structures, diseases and inhibitors. Pharmacol Ther 2020; 215:107622. [PMID: 32650009 DOI: 10.1016/j.pharmthera.2020.107622] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Protein phosphatases and kinases control multiple cellular events including proliferation, differentiation, and stress responses through regulating reversible protein phosphorylation, the most important post-translational modification. Members of metal-dependent protein phosphatase (PPM) family, also known as PP2C phosphatases, are Ser/Thr phosphatases that bind manganese/magnesium ions (Mn2+/Mg2+) in their active center and function as single subunit enzymes. In mammals, there are 20 isoforms of PPM phosphatases: PPM1A, PPM1B, PPM1D, PPM1E, PPM1F, PPM1G, PPM1H, PPM1J, PPM1K, PPM1L, PPM1M, PPM1N, ILKAP, PDP1, PDP2, PHLPP1, PHLPP2, PP2D1, PPTC7, and TAB1, whereas there are only 8 in yeast. Phylogenetic analysis of the DNA sequences of vertebrate PPM isoforms revealed that they can be divided into 12 different classes: PPM1A/PPM1B/PPM1N, PPM1D, PPM1E/PPM1F, PPM1G, PPM1H/PPM1J/PPM1M, PPM1K, PPM1L, ILKAP, PDP1/PDP2, PP2D1/PHLPP1/PHLPP2, TAB1, and PPTC7. PPM-family members have a conserved catalytic core region, which contains the metal-chelating residues. The different isoforms also have isoform specific regions within their catalytic core domain and terminal domains, and these regions may be involved in substrate recognition and/or functional regulation of the phosphatases. The twenty mammalian PPM phosphatases are involved in regulating diverse cellular functions, such as cell cycle control, cell differentiation, immune responses, and cell metabolism. Mutation, overexpression, or deletion of the PPM phosphatase gene results in abnormal cellular responses, which lead to various human diseases. This review focuses on the structures and biological functions of the PPM-phosphatase family and their associated diseases. The development of specific inhibitors against the PPM phosphatase family as a therapeutic strategy will also be discussed.
Collapse
|
371
|
Recurrent genetic HLA loss in AML relapsed after matched unrelated allogeneic hematopoietic cell transplantation. Blood Adv 2020; 3:2199-2204. [PMID: 31324640 DOI: 10.1182/bloodadvances.2019000445] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Immune evasion is a hallmark of cancer and a central mechanism underlying acquired resistance to immune therapy. In allogeneic hematopoietic cell transplantation (alloHCT), late relapses can arise after prolonged alloreactive T-cell control, but the molecular mechanisms of immune escape remain unclear. To identify mechanisms of immune evasion, we performed a genetic analysis of serial samples from 25 patients with myeloid malignancies who relapsed ≥1 year after alloHCT. Using targeted sequencing and microarray analysis to determine HLA allele-specific copy number, we identified copy-neutral loss of heterozygosity events and focal deletions spanning class 1 HLA genes in 2 of 12 recipients of matched unrelated-donor HCT and in 1 of 4 recipients of mismatched unrelated-donor HCT. Relapsed clones, although highly related to their antecedent pretransplantation malignancies, frequently acquired additional mutations in transcription factors and mitogenic signaling genes. Previously, the study of relapse after haploidentical HCT established the paradigm of immune evasion via loss of mismatched HLA. Here, in the context of matched unrelated-donor HCT, HLA loss provides genetic evidence that allogeneic immune recognition may be mediated by minor histocompatibility antigens and suggests opportunities for novel immunologic approaches for relapse prevention.
Collapse
|
372
|
Ma S, Shi W, Li Z, Tang L, Wang H, Xia L, Hu Y. Reduced-intensity versus Myeloablative Conditioning Regimens for Younger Adults with Acute Myeloid Leukemia and Myelodysplastic Syndrome: A systematic review and meta-analysis. J Cancer 2020; 11:5223-5235. [PMID: 32742468 PMCID: PMC7378925 DOI: 10.7150/jca.46081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/15/2020] [Indexed: 01/06/2023] Open
Abstract
Background: Historically, reduced-intensity conditioning (RIC) was recommended to be performed for older patients who were considered ineligible for myeloablative conditioning (MAC) before allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the evidence regarding the optimal conditioning intensity in younger patients with AML or MDS is weak and contradictory. Methods: PubMed, Medline, Embase, and other online sources were searched from the initial period to February 25, 2020. Odds ratios and 95% confidence intervals were calculated to estimate pooling effects. Results: Four randomized controlled trials (RCTs) about conditioning intensity involving 633 patients were included. There were no significant differences of 1/2/4/5 years progression-free survival (PFS) and relapse incidence (RI) between two conditioning intensities. Overall survival (OS) was similar at 1/2/4 years, but patients receiving RIC had a higher OS at 5 years. Additionally, RIC were associated with lower non-relapse mortality, less grade II-IV and grade III-IV acute graft-versus-host disease (GVHD), and lower incidence of chronic GVHD compared with MAC regimens. Subgroup analysis showed similar OS and RI for AML patients, and there was a trend towards lower NRM and grade II-IV aGVHD in RIC group. Available data for MDS indicated that OS, PFS, and RI were comparable. For intermediate-risk patients, there was no evidence that RIC is inferior to MAC. However, for high-risk patients, MAC tends to perform better. Conclusions: Based on the above results, it might be concluded that RIC is a feasible treatment option for adults with AML or MDS younger than 66 years, particularly those with intermediate-risk disease. Future RCTs incorporating of risk stratifications are warranted to guide the optimal decision under certain conditions.
Collapse
Affiliation(s)
- Shengling Ma
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Ziying Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huafang Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
373
|
Higgins A, Shah MV. Genetic and Genomic Landscape of Secondary and Therapy-Related Acute Myeloid Leukemia. Genes (Basel) 2020; 11:E749. [PMID: 32640569 PMCID: PMC7397259 DOI: 10.3390/genes11070749] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/22/2022] Open
Abstract
A subset of acute myeloid leukemia (AML) arises either from an antecedent myeloid malignancy (secondary AML, sAML) or as a complication of DNA-damaging therapy for other cancers (therapy-related myeloid neoplasm, t-MN). These secondary leukemias have unique biological and clinical features that distinguish them from de novo AML. Over the last decade, molecular techniques have unraveled the complex subclonal architecture of sAML and t-MN. In this review, we compare and contrast biological and clinical features of de novo AML with sAML and t-MN. We discuss the role of genetic mutations, including those involved in RNA splicing, epigenetic modification, tumor suppression, transcription regulation, and cell signaling, in the pathogenesis of secondary leukemia. We also discuss clonal hematopoiesis in otherwise healthy individuals, as well as in the context of another malignancy, and how it challenges the conventional notion of sAML/t-MN. We conclude by summarizing the current and emerging treatment strategies, including allogenic transplant, in these complex scenarios.
Collapse
|
374
|
Hellström-Lindberg E, Tobiasson M, Greenberg P. Myelodysplastic syndromes: moving towards personalized management. Haematologica 2020; 105:1765-1779. [PMID: 32439724 PMCID: PMC7327628 DOI: 10.3324/haematol.2020.248955] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
The myelodysplastic syndromes (MDS) share their origin in the hematopoietic stem cell but have otherwise very heterogeneous biological and genetic characteristics. Clinical features are dominated by cytopenia and a substantial risk for progression to acute myeloid leukemia. According to the World Health Organization, MDS is defined by cytopenia, bone marrow dysplasia and certain karyotypic abnormalities. The understanding of disease pathogenesis has undergone major development with the implementation of next-generation sequencing and a closer integration of morphology, cytogenetics and molecular genetics is currently paving the way for improved classification and prognostication. True precision medicine is still in the future for MDS and the development of novel therapeutic compounds with a propensity to markedly change patients' outcome lags behind that for many other blood cancers. Treatment of higher-risk MDS is dominated by monotherapy with hypomethylating agents but novel combinations are currently being evaluated in clinical trials. Agents that stimulate erythropoiesis continue to be first-line treatment for the anemia of lower-risk MDS but luspatercept has shown promise as second-line therapy for sideroblastic MDS and lenalidomide is an established second-line treatment for del(5q) lower-risk MDS. The only potentially curative option for MDS is hematopoietic stem cell transplantation, until recently associated with a relatively high risk of transplant-related mortality and relapse. However, recent studies show increased cure rates due to better tools to target the malignant clone with less toxicity. This review provides a comprehensive overview of the current status of the clinical evaluation, biology and therapeutic interventions for this spectrum of disorders.
Collapse
Affiliation(s)
- Eva Hellström-Lindberg
- Karolinska Institutet, Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Tobiasson
- Karolinska Institutet, Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Greenberg
- Stanford Cancer Institute, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
375
|
Coombs CC, Dickherber T, Crompton BD. Chasing ctDNA in Patients With Sarcoma. Am Soc Clin Oncol Educ Book 2020; 40:e351-e360. [PMID: 32598183 DOI: 10.1200/edbk_280749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Liquid biopsies are new technologies that allow cancer profiling of tumor fragments found in body fluids, such as peripheral blood, collected noninvasively from patients with malignancies. These assays are increasingly valuable in clinical oncology practice as prognostic biomarkers, as guides for therapy selection, for treatment monitoring, and for early detection of disease progression and relapse. However, application of these assays to rare cancers, such as pediatric and adult sarcomas, have lagged. In this article, we review the technical challenges of applying liquid biopsy technologies to sarcomas, provide an update on progress in the field, describe common pitfalls in interpreting liquid biopsy data, and discuss the intersection of sarcoma clinical care and commercial assays emerging on the horizon.
Collapse
Affiliation(s)
| | | | - Brian D Crompton
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| |
Collapse
|
376
|
Nonsense Suppression Therapy: New Hypothesis for the Treatment of Inherited Bone Marrow Failure Syndromes. Int J Mol Sci 2020; 21:ijms21134672. [PMID: 32630050 PMCID: PMC7369780 DOI: 10.3390/ijms21134672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Inherited bone marrow failure syndromes (IBMFS) are a group of cancer-prone genetic diseases characterized by hypocellular bone marrow with impairment in one or more hematopoietic lineages. The pathogenesis of IBMFS involves mutations in several genes which encode for proteins involved in DNA repair, telomere biology and ribosome biogenesis. The classical IBMFS include Shwachman–Diamond syndrome (SDS), Diamond–Blackfan anemia (DBA), Fanconi anemia (FA), dyskeratosis congenita (DC), and severe congenital neutropenia (SCN). IBMFS are associated with high risk of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML), and solid tumors. Unfortunately, no specific pharmacological therapies have been highly effective for IBMFS. Hematopoietic stem cell transplantation provides a cure for aplastic or myeloid neoplastic complications. However, it does not affect the risk of solid tumors. Since approximately 28% of FA, 24% of SCN, 21% of DBA, 20% of SDS, and 17% of DC patients harbor nonsense mutations in the respective IBMFS-related genes, we discuss the use of the nonsense suppression therapy in these diseases. We recently described the beneficial effect of ataluren, a nonsense suppressor drug, in SDS bone marrow hematopoietic cells ex vivo. A similar approach could be therefore designed for treating other IBMFS. In this review we explain in detail the new generation of nonsense suppressor molecules and their mechanistic roles. Furthermore, we will discuss strengths and limitations of these molecules which are emerging from preclinical and clinical studies. Finally we discuss the state-of-the-art of preclinical and clinical therapeutic studies carried out for IBMFS.
Collapse
|
377
|
External validation and comparison of multiple prognostic scores in allogeneic hematopoietic stem cell transplantation. Blood Adv 2020; 3:1881-1890. [PMID: 31221661 DOI: 10.1182/bloodadvances.2019032268] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/16/2019] [Indexed: 12/23/2022] Open
Abstract
Clinical decisions in allogeneic hematopoietic stem cell transplantation (allo-HSCT) are supported by the use of prognostic scores for outcome prediction. Scores vary in their features and in the composition of development cohorts. We sought to externally validate and compare the performance of 8 commonly applied scoring systems on a cohort of allo-HSCT recipients. Among 528 patients studied, acute myeloid leukemia was the leading transplant indication (44%) and 46% of patients had a matched sibling donor. Most models successfully grouped patients into higher and lower risk strata, supporting their use for risk classification. However, discrimination varied (2-year overall survival area under the receiver operating characteristic curve [AUC]: revised Pretransplantation Assessment of Mortality [rPAM], 0.64; PAM, 0.63; revised Disease Risk Index [rDRI], 0.62; Endothelial Activation and Stress Index [EASIx], 0.60; combined European Society for Blood and Marrow Transplantation [EBMT]/Hematopoietic Cell Transplantation-specific Comorbidity Index [HCT-CI], 0.58; EBMT, 0.58; Comorbidity-Age, 0.58; HCT-CI, 0.55); AUC ranges from 0.5 (random) to 1.0 (perfect prediction). rPAM and PAM, which had the greatest predictive capacity across all outcomes, are comprehensive models including patient, disease, and transplantation information. Interestingly, EASIx, a biomarker-driven model, had comparable performance for nonrelapse mortality (NRM; 2-year AUC, 0.65) but no predictive value for relapse (2-year AUC, 0.53). Overall, allo-HSCT prognostic systems may be useful for risk stratification, but individual prediction remains a challenge, as reflected by the scores' limited discriminative capacity.
Collapse
|
378
|
Clonal hematopoiesis is associated with adverse outcomes in multiple myeloma patients undergoing transplant. Nat Commun 2020; 11:2996. [PMID: 32533060 PMCID: PMC7293239 DOI: 10.1038/s41467-020-16805-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/20/2020] [Indexed: 12/30/2022] Open
Abstract
Multiple myeloma (MM) is a plasma-cell neoplasm that is treated with high-dose chemotherapy, autologous stem cell transplant (ASCT) and long-term immunomodulatory drug (IMiD) maintenance. The presence of somatic mutations in the peripheral blood is termed clonal hematopoiesis of indeterminate potential (CHIP) and is associated with adverse outcomes. Targeted sequencing of the stem cell product from 629 MM patients treated by ASCT at the Dana-Farber Cancer Institute (2003–2011) detects CHIP in 136/629 patients (21.6%). The most commonly mutated genes are DNMT3A, TET2, TP53, ASXL1 and PPM1D. Twenty-one from fifty-six patients (3.3%) receiving first-line IMiD maintenance develop a therapy-related myeloid neoplasm (TMN). However, regardless of CHIP status, the use of IMiD maintenance associates with improved PFS and OS. In those not receiving IMiD maintenance, CHIP is associated with decreased overall survival (OS) (HR:1.34, p = 0.02) and progression free survival (PFS) (HR:1.45, p < 0.001) due to an increase in MM progression. Multiple myeloma (MM) is treated with induction chemotherapy, autologous stem cell transplant (ASCT) and long-term immunomodulatory drug (IMiD) maintenance. Here, the authors show that the presence of clonal haematopoiesis of indeterminate potential (CHIP) at time of ASCT is associated with adverse outcomes in MM patients.
Collapse
|
379
|
Carraway HE, LaFramboise T. Myeloid neoplasms with germline predisposition: Practical considerations and complications in the search for new susceptibility loci. Best Pract Res Clin Haematol 2020; 33:101191. [PMID: 33038980 DOI: 10.1016/j.beha.2020.101191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
Abstract
Genomic research in hematological malignancies has focused far more prominently on somatic mutations than on germline variants. Although increasing numbers of germline variants are being identified, a substantial proportion of familial myeloid malignancies have no causal allele pinpointed. Here we review the biological, technological, and clinical challenges that stand in the way of the goal of establishing, implementing, and interpreting a comprehensive panel of germline variants for testing. Achieving this goal would inform care for large numbers of myeloid malignancy patients. Furthermore, knowledge of germline susceptibility variants and their corresponding genes will shed light on disease processes, potentially suggesting therapeutic strategies tailored to specific variants.
Collapse
Affiliation(s)
- Hetty E Carraway
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Thomas LaFramboise
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900, Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
380
|
Goyal H, Chachoua I, Pecquet C, Vainchenker W, Constantinescu SN. A p53-JAK-STAT connection involved in myeloproliferative neoplasm pathogenesis and progression to secondary acute myeloid leukemia. Blood Rev 2020; 42:100712. [PMID: 32660739 DOI: 10.1016/j.blre.2020.100712] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/25/2020] [Accepted: 05/27/2020] [Indexed: 01/14/2023]
Abstract
Since the discovery of JAK2 V617F as a highly prevalent somatic acquired mutation in the majority of myeloproliferative neoplasms (MPNs), it has become clear that these diseases are driven by pathologic activation of JAK2 and eventually of STAT5 and other members of the STAT family. The concept was strengthened by the discovery of the other activating driver mutations in MPL (thrombopoietin receptor, TpoR) and in calreticulin gene, which all lead to persistent activation of wild type JAK2. Although with a rare frequency, MPNs can evolve to secondary acute myeloid leukemia (sAML), a condition that is resistant to treatment. Here we focus on the role of p53 in this transition. In sAML mutations in TP53 or amplification in genes coding for negative regulators of p53 are much more frequent than in de novo AML. We review studies that explore a signaling and biochemical interaction between activated STATs and p53 in MPNs and other cancers. With the development of advanced sequencing efforts, strong evidence has been presented for dominant negative effects of mutated p53 in leukemia. In other studies, gain of function effects have been described that might be cell type specific. A more profound understanding of the potential interaction between p53 and activated STATs is necessary in order to take full advantage of novel p53-targeted therapies.
Collapse
Affiliation(s)
- Harsh Goyal
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium; Université catholique de Louvain and de Duve Institute, Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium
| | - Ilyas Chachoua
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium; Université catholique de Louvain and de Duve Institute, Brussels, Belgium; Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden
| | - Christian Pecquet
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium; Université catholique de Louvain and de Duve Institute, Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium
| | - William Vainchenker
- INSERM, Unité Mixte de Recherche 1170, Institut Gustave Roussy, Villejuif, France; Paris-Saclay, Unité Mixte de Recherche 1170, Institut Gustave Roussy, Villejuif, France; Gustave Roussy, Unité Mixte de Recherche 1170, Villejuif, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium; Université catholique de Louvain and de Duve Institute, Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium.
| |
Collapse
|
381
|
Myeloablative versus Reduced-Intensity Hematopoietic Cell Transplantation in Myelodysplastic Syndromes: Systematic Review and Meta-analysis. Biol Blood Marrow Transplant 2020; 26:e138-e141. [DOI: 10.1016/j.bbmt.2020.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 11/21/2022]
|
382
|
Shreve J, Nazha A. The Evolving Landscape of Myelodysplastic Syndrome Prognostication. Clin Hematol Int 2020; 2:43-48. [PMID: 32879911 PMCID: PMC7462414 DOI: 10.2991/chi.d.200408.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/07/2020] [Indexed: 11/10/2022] Open
Abstract
Myelodysplastic syndromes (MDSs) are potentially devastating monoclonal deviations of hematopoiesis that lead to bone marrow dysplasia and variable cytopenias. Predicting severity of disease progression and likelihood to undergo acute myeloid leukemia transformation is the basis of treatment strategy. Some patients belong to a low-risk cohort best managed with conservative supportive care, whereas others are included in a high-risk cohort that requires decisive therapy with hematopoietic cell transplantation or hypomethylating agent administration. Risk scoring systems for MDS prognostication were traditionally based on karyotype characteristics and clinical factors readily available from chart review, and validation was typically conducted on de novo MDS patients. However, retrospective analysis found a large subset of patients incorrectly risk-stratified. In this review, the most commonly used scoring systems are evaluated, and pitfalls therein are identified. Emerging technologies such as personal genomics and machine learning are then explored for efficacy in MDS risk modeling. Barriers to clinical adoption of artificial intelligence-derived models are discussed, with focus on approaches meant to increase model interpretability and clinical relevance. Finally, a guiding set of recommendations is proposed for best designing an accurate and universally applicable prognostic model for MDS, which is supported by more than 20 years of observation of traditional scoring system performance, as well as modern efforts in creating hybrid genomic-clinical scoring systems.
Collapse
Affiliation(s)
- Jacob Shreve
- Department of Hematology and Medical Oncology, Taussig Cancer Center, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH, USA
| | - Aziz Nazha
- Department of Hematology and Medical Oncology, Taussig Cancer Center, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH, USA
| |
Collapse
|
383
|
Wedge E, Sengeløv H, Hansen JW, Andersen NS, Schjødt I, Petersen SL, Kornblit B, Grønbæk K, Friis LS. Improved Outcomes after Allogenic Hematopoietic Stem Cell Transplantation with Fludarabine/Treosulfan for Patients with Myelodysplastic Syndromes. Biol Blood Marrow Transplant 2020; 26:1091-1098. [DOI: 10.1016/j.bbmt.2020.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/13/2022]
|
384
|
Feld J, Belasen A, Navada SC. Myelodysplastic syndromes: a review of therapeutic progress over the past 10 years. Expert Rev Anticancer Ther 2020; 20:465-482. [PMID: 32479130 DOI: 10.1080/14737140.2020.1770088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Myelodysplastic syndromes (MDS) represent a range of bone marrow disorders, with patients affected by cytopenias and risk of progression to AML. There are limited therapeutic options available for patients, including hypomethylating agents (azacitidine/decitabine), growth factor support, lenalidomide, and allogeneic stem cell transplant. AREAS COVERED This review provides an overview of the progress made over the past decade for emerging therapies for lower- and higher-risk MDS (MDS-HR). We also cover advances in prognostication, supportive care, and use of allogeneic SCT in MDS. EXPERT OPINION While there have been no FDA-approved therapies for MDS in the past decade, we anticipate the approval of luspatercept based on results from the MEDALIST trial for patients with lower-risk MDS (MDS-LR) and ringed sideroblasts who have failed or are ineligible for erythropoiesis stimulating agents (ESAs). With growing knowledge of the biologic and molecular mechanisms underlying MDS, it is anticipated that new therapies will be approved in the coming years.
Collapse
Affiliation(s)
- Jonathan Feld
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine , New York, USA
| | - Abigail Belasen
- Department of Medicine, Icahn School of Medicine , New York, USA
| | - Shyamala C Navada
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine , New York, USA
| |
Collapse
|
385
|
|
386
|
Myers K, Hebert K, Antin J, Boulad F, Burroughs L, Hofmann I, Kamble R, MacMillan ML, Eapen M. Hematopoietic Stem Cell Transplantation for Shwachman-Diamond Syndrome. Biol Blood Marrow Transplant 2020; 26:1446-1451. [PMID: 32428734 DOI: 10.1016/j.bbmt.2020.04.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/20/2020] [Accepted: 04/29/2020] [Indexed: 11/15/2022]
Abstract
We report the outcomes of hematopoietic stem cell transplantation (HSCT) for 52 patients with Shwachman-Diamond syndrome (SDS) who underwent transplantation between 2000 and 2017. The median age at transplantation was 11 years, and the median duration of follow-up was 60 months. The indication for HSCT was bone marrow failure (BMF; cytopenia or aplastic anemia) in 39 patients and myelodysplasia (MDS)/acute myelogenous leukemia (AML) in 13 patients. The donor type was an HLA-matched sibling for 18 patients, an HLA-matched or mismatched relative for 6 patients, and an HLA-matched or mismatched unrelated donor for 28 patients. Preparative regimens for BMF were myeloablative in 13 patients and reduced intensity in 26. At the time of this report, 29 of the 39 patients with BMF were alive, and the 5-year overall survival was 72% (95% confidence interval, 57% to 86%). Graft failure and graft-versus-host disease were the predominant causes of death. Preparative regimens for patients with MDS/AML were myeloablative in 8 and reduced intensity in 5. At the time of this report, only 2 of 13 patients were alive (15%), with relapse the predominant cause of death. Survival after transplantation for SDS-related BMF is better compared with historical reports, but strategies are needed to overcome graft failure and graft-versus-host disease. For SDS- related MDS or AML, transplantation does not extend survival. Rigorous surveillance and novel treatments for leukemia are urgently needed.
Collapse
Affiliation(s)
- Kasiani Myers
- Division of Blood and Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kyle Hebert
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Joseph Antin
- Division of Hematology and Oncology, Dana Farber Cancer Institute at Brigham and Women's Hospital, Boston, Massachusetts
| | - Farid Boulad
- Division of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lauri Burroughs
- Data Abstraction Department- Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Inga Hofmann
- Division of Blood and Bone Marrow Transplant, University of Wisconsin Hospital and Clinics, Madison, Wisconsin
| | - Rammurti Kamble
- Division of Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Margaret L MacMillan
- Division of Pediatrics-Blood and Marrow Transplant Program, University of Minnesota Blood and Marrow Transplant Program, Minneapolis, Minnesota
| | - Mary Eapen
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
387
|
Abstract
As people age, their tissues accumulate an increasing number of somatic mutations. Although most of these mutations are of little or no functional consequence, a mutation may arise that confers a fitness advantage on a cell. When this process happens in the hematopoietic system, a substantial proportion of circulating blood cells may derive from a single mutated stem cell. This outgrowth, called "clonal hematopoiesis," is highly prevalent in the elderly population. Here we discuss recent advances in our knowledge of clonal hematopoiesis, its relationship to malignancies, its link to nonmalignant diseases of aging, and its potential impact on immune function. Clonal hematopoiesis provides a glimpse into the process of mutation and selection that likely occurs in all somatic tissues.
Collapse
Affiliation(s)
- Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Benjamin L Ebert
- Department of Medical Oncology, Howard Hughes Medical Institute, Boston, MA. .,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
388
|
Cumbo C, Tota G, Anelli L, Zagaria A, Specchia G, Albano F. TP53 in Myelodysplastic Syndromes: Recent Biological and Clinical Findings. Int J Mol Sci 2020; 21:E3432. [PMID: 32414002 PMCID: PMC7279310 DOI: 10.3390/ijms21103432] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
TP53 dysregulation plays a pivotal role in the molecular pathogenesis of myelodysplastic syndromes (MDS), identifying a subgroup of patients with peculiar features. In this review we report the recent biological and clinical findings of TP53-mutated MDS, focusing on the molecular pathways activation and on its impact on the cellular physiology. In MDS, TP53 mutational status is deeply associated with del(5q) syndrome and its dysregulation impacts on cell cycle, DNA repair and apoptosis inducing chromosomal instability and the clonal evolution of disease. TP53 defects influence adversely the MDS clinical outcome and the treatment response rate, thus new therapeutic approaches are being developed for these patients. TP53 allelic state characterization and the mutational burden evaluation can therefore predict prognosis and identify the subgroup of patients eligible for targeted therapy. For these reasons, in the era of precision medicine, the MDS diagnostic workup cannot do without the complete assessment of TP53 mutational profile.
Collapse
Affiliation(s)
| | | | | | | | | | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy; (C.C.); (G.T.); (L.A.); (A.Z.); (G.S.)
| |
Collapse
|
389
|
Do next-generation sequencing results drive diagnostic and therapeutic decisions in MDS? Blood Adv 2020; 3:3454-3460. [PMID: 31714959 DOI: 10.1182/bloodadvances.2019000680] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
This article has a companion Point by Thol and Platzbecker.
Collapse
|
390
|
An WB, Liu C, Wan Y, Chang LX, Chen XY, Zhu XF. [Clinical features and gene mutations of children with Shwachman-Diamond syndrome and malignant myeloid transformation]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:460-465. [PMID: 32434641 PMCID: PMC7389397 DOI: 10.7499/j.issn.1008-8830.2001133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/01/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To study the clinical features and genetic mutations of children with Shwachman-Diamond syndrome (SDS) and malignant myeloid transformation. METHODS Next-generation sequencing was used to analyze the gene mutations in 11 SDS children with malignant myeloid transformation, and their clinical features and genetic mutations were analyzed. RESULTS Of the 11 children with SDS, 9 (82%) presented with refractory cytopenia of childhood (RCC), 1 (9%) had myelodysplastic syndrome with excess blasts (MDS-EB), and 1 (9%) had acute myeloid leukemia with myelodysplasia-related changes (AML-MRC). The median age of onset of malignant myeloid transformation was 48 months (ranged 7 months to 14 years). Of the 11 children, 45% had abnormalities in the hematological system alone. Mutations of the SBDS gene were detected in all 11 children, among whom 5 (45%) had c.258+2T>C homozygous mutation and 3 (27%) had c.184A>T+c.258+2T>C compound heterozygous mutation. The new mutations of the SBDS gene, c.634_635insAACATACCTGT+c.637_638delGA and c.8T>C, were rated as "pathogenic" and "possibly pathogenic" respectively. The 3-year predicted overall survival rates of children transformed to RCC and MDS-EB/AML-MRC were 100% and 0% respectively (P=0.001). CONCLUSIONS SDS children may have hematological system symptoms as the only manifestation, which needs to be taken seriously in clinical practice. The type of malignant transformation is associated with prognosis.
Collapse
Affiliation(s)
- Wen-Bin An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| | | | | | | | | | | |
Collapse
|
391
|
Plasma cell-free DNA variant analysis compared with methylated DNA analysis in renal cell carcinoma. Genet Med 2020; 22:1366-1373. [DOI: 10.1038/s41436-020-0801-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 11/09/2022] Open
|
392
|
Stoddart A, Wang J, Fernald AA, Davis EM, Johnson CR, Hu C, Cheng JX, McNerney ME, Le Beau MM. Cytotoxic Therapy-Induced Effects on Both Hematopoietic and Marrow Stromal Cells Promotes Therapy-Related Myeloid Neoplasms. Blood Cancer Discov 2020; 1:32-47. [PMID: 32924016 DOI: 10.1158/2643-3230.bcd-19-0028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Therapy-related myeloid neoplasms (t-MNs) following treatment with alkylating agents are characterized by a del(5q), complex karyotypes, alterations of TP53, and a dismal prognosis. To decipher the molecular pathway(s) leading to the pathogenesis of del(5q) t-MN and the effect(s) of cytotoxic therapy on the marrow microenvironment, we developed a mouse model with loss of two key del(5q) genes, EGR1 and APC, in hematopoietic cells. We used the well-characterized drug, N-ethyl-N-nitrosurea (ENU) to demonstrate that alkylating agent exposure of stromal cells in the microenvironment increases the incidence of myeloid disease. In addition, loss of Trp53 with Egr1 and Apc was required to drive the development of a transplantable leukemia, and accompanied by the acquisition of somatic mutations in DNA damage response genes. ENU treatment of mesenchymal stromal cells induced cellular senescence, and led to the acquisition of a senescence-associated secretory phenotype, which may be a critical microenvironmental alteration in the pathogenesis of myeloid neoplasms.
Collapse
Affiliation(s)
| | - Jianghong Wang
- Department of Medicine, University of Chicago, Chicago, IL
| | | | | | | | - Chunmei Hu
- Department of Medicine, University of Chicago, Chicago, IL
| | - Jason X Cheng
- Department of Pathology, University of Chicago, Chicago, IL.,University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL
| | - Megan E McNerney
- Department of Pathology, University of Chicago, Chicago, IL.,University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL.,Department of Pediatrics, University of Chicago, Chicago IL
| | - Michelle M Le Beau
- Department of Medicine, University of Chicago, Chicago, IL.,University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL
| |
Collapse
|
393
|
Hourigan CS, Dillon LW, Gui G, Logan BR, Fei M, Ghannam J, Li Y, Licon A, Alyea EP, Bashey A, Deeg HJ, Devine SM, Fernandez HF, Giralt S, Hamadani M, Howard A, Maziarz RT, Porter DL, Scott BL, Warlick ED, Pasquini MC, Horwitz ME. Impact of Conditioning Intensity of Allogeneic Transplantation for Acute Myeloid Leukemia With Genomic Evidence of Residual Disease. J Clin Oncol 2020; 38:1273-1283. [PMID: 31860405 PMCID: PMC7164487 DOI: 10.1200/jco.19.03011] [Citation(s) in RCA: 316] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Patients with acute myeloid leukemia (AML) in remission remain at risk for relapse even after allogeneic hematopoietic cell transplantation (alloHCT). AML measurable residual disease (MRD) status before alloHCT has been shown to be prognostic. Whether modulation of the intensity of the alloHCT conditioning regimen in patients with AML who test positive for MRD can prevent relapse and improve survival is unknown. METHODS Ultra-deep, error-corrected sequencing for 13 commonly mutated genes in AML was performed on preconditioning blood from patients treated in a phase III clinical trial that randomly assigned adult patients with myeloid malignancy in morphologic complete remission to myeloablative conditioning (MAC) or reduced-intensity conditioning (RIC). RESULTS No mutations were detected in 32% of MAC and 37% of RIC recipients; these groups had similar survival (3-year overall survival [OS], 56% v 63%; P = .96). In patients with a detectable mutation (next-generation sequencing [NGS] positive), relapse (3-year cumulative incidence, 19% v 67%; P < .001) and survival (3-year OS, 61% v 43%; P = .02) was significantly different between the MAC and RIC arms, respectively. In multivariable analysis for NGS-positive patients, adjusting for disease risk and donor group, RIC was significantly associated with increased relapse (hazard ratio [HR], 6.38; 95% CI, 3.37 to 12.10; P < .001), decreased relapse-free survival (HR, 2.94; 95% CI, 1.84 to 4.69; P < .001), and decreased OS (HR, 1.97; 95% CI, 1.17 to 3.30; P = .01) compared with MAC. Models of AML MRD also showed benefit for MAC over RIC for those who tested positive. CONCLUSION This study provides evidence that MAC rather than RIC in patients with AML with genomic evidence of MRD before alloHCT can result in improved survival.
Collapse
MESH Headings
- Adult
- Aged
- Circulating Tumor DNA/blood
- Circulating Tumor DNA/genetics
- Clinical Trials, Phase III as Topic
- Female
- Hematopoietic Stem Cell Transplantation/methods
- High-Throughput Nucleotide Sequencing
- Humans
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Mutation
- Neoplasm, Residual
- Prognosis
- Randomized Controlled Trials as Topic
- Transplantation Conditioning/methods
- Transplantation, Homologous
- Young Adult
Collapse
Affiliation(s)
| | - Laura W. Dillon
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Gege Gui
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | | | - Jack Ghannam
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Yuesheng Li
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | | | - Asad Bashey
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA
| | | | - Steven M. Devine
- National Marrow Donor Program and Center for International Blood and Marrow Transplant Research, Minneapolis, MN
| | | | - Sergio Giralt
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Alan Howard
- National Marrow Donor Program and Center for International Blood and Marrow Transplant Research, Minneapolis, MN
| | - Richard T. Maziarz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | | | | | | | | | | |
Collapse
|
394
|
Freeman S, Craddock C. Less Is Not Necessarily More: Toward a Rational Selection of the Conditioning Regimen in Acute Myeloid Leukemia. J Clin Oncol 2020; 38:1249-1251. [DOI: 10.1200/jco.19.03161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Sylvie Freeman
- Institute of Infection and Immunity University of Birmingham, Edgbaston, Birmingham, United Kingdom
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
- Cancer Research UK Cancer Trials Unit, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
395
|
Schratz KE, DeZern AE. Genetic Predisposition to Myelodysplastic Syndrome in Clinical Practice. Hematol Oncol Clin North Am 2020; 34:333-356. [PMID: 32089214 PMCID: PMC7875473 DOI: 10.1016/j.hoc.2019.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Myelodysplastic syndromes (MDSs) are a heterogeneous group of marrow failure disorders that primarily affect older persons but also occur at a lower frequency in children and young adults. There is increasing recognition of an inherited predisposition to MDS as well as other myeloid malignancies for patients of all ages. Germline predisposition to MDS can occur as part of a syndrome or sporadic disease. The timely diagnosis of an underlying genetic predisposition in the setting of MDS is important. This article delineates germline genetic causes of MDS and provides a scaffold for the diagnosis and management of patients in this context.
Collapse
Affiliation(s)
- Kristen E Schratz
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Bloomberg 11379, 1800 Orleans Street, Baltimore, MD 21287, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD 21287, USA
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD 21287, USA; Division of Hematologic Malignancies, Johns Hopkins University School of Medicine, CRBI Room 3M87, 1650 Orleans Street, Baltimore, MD 21287-0013, USA.
| |
Collapse
|
396
|
Shreve J, Nazha A. Novel Prognostic Models for Myelodysplastic Syndromes. Hematol Oncol Clin North Am 2020; 34:369-378. [DOI: 10.1016/j.hoc.2019.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
397
|
|
398
|
Cesaro S, Pegoraro A, Sainati L, Lucidi V, Montemitro E, Corti P, Ramenghi U, Nasi C, Menna G, Zecca M, Danesino C, Nicolis E, Pasquali F, Perobelli S, Tridello G, Farruggia P, Cipolli M. A Prospective Study of Hematologic Complications and Long-Term Survival of Italian Patients Affected by Shwachman-Diamond Syndrome. J Pediatr 2020; 219:196-201.e1. [PMID: 32037152 DOI: 10.1016/j.jpeds.2019.12.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/30/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To describe the hematologic outcome and long-term survival of patients enrolled in the Shwachman-Diamond syndrome Italian Registry. STUDY DESIGN A retrospective and prospective study of patients recorded in the Shwachman-Diamond syndrome Italian Registry. RESULTS The study population included 121 patients, 69 males and 52 females, diagnosed between 1999 and 2018. All patients had the clinical diagnosis confirmed by mutational analysis on the SBDS gene. During the study period, the incidence of SDS was 1 in 153 000 births. The median age of patients with SDS at diagnosis was 1.3 years (range, 0-35.6 years). At the first hematologic assessment, severe neutropenia was present in 25.8%, thrombocytopenia in 25.5%, and anemia in 4.6% of patients. A normal karyotype was found in 40 of 79 patients, assessed whereas the most frequent cytogenetic abnormalities were isochromosome 7 and interstitial deletion of the long arm of chromosome 20. The cumulative incidence of severe neutropenia, thrombocytopenia, and anemia at 30 years of age were 59.9%, 66.8%, and 20.2%, respectively. The 20-year cumulative incidence of myelodysplastic syndrome/leukemia and of bone marrow failure/severe cytopenia was 9.8% and 9.9%, respectively. Fifteen of 121 patients (12.4%) underwent allogeneic stem cell transplantation. Fifteen patients (12.4%) died; the probability of overall survival at 10 and 20 years was 95.7% and 87.4%, respectively. CONCLUSIONS Despite an improvement in survival, hematologic complications still cause death in patients with SDS. Further studies are needed to optimize type and modality of hematopoietic stem cell transplantation and to assess the long-term outcome in nontransplanted patients.
Collapse
Affiliation(s)
- Simone Cesaro
- Pediatric Hematology and Oncology Unit, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | - Anna Pegoraro
- Pediatric Hematology and Oncology Unit, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Laura Sainati
- Pediatric Hematology and Oncology Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Vincenzina Lucidi
- Cystic Fibrosis Unit, Department of Specialized Pediatrics, "Bambino Gesù" Children's Hospital, Istituto di Ricerca e Cura a Carattere Scientifico, Roma, Italy
| | - Enza Montemitro
- Cystic Fibrosis Unit, Department of Specialized Pediatrics, "Bambino Gesù" Children's Hospital, Istituto di Ricerca e Cura a Carattere Scientifico, Roma, Italy
| | - Paola Corti
- Pediatric Hematology Oncology, Department of Pediatrics, University Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Ugo Ramenghi
- Department of Pediatric and Public Health Sciences, University of Torino, Torino, Italy
| | - Cristina Nasi
- Division of Pediatrics, Azienda Sanitaria ASL 17, Savigliano, Italy
| | - Giuseppe Menna
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Hospital, Napoli, Italy
| | - Marco Zecca
- Pediatric Hematology and Oncology Unit, Deparment of Pediatrics, Istituto di Ricerca e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Cesare Danesino
- Department of Molecular Medicine, University of Pavia and Fondazione Istituto di Ricerca e Cura a Carattere Scientifico Policlinico S. Matteo, Pavia, Italy
| | - Elena Nicolis
- L Transfusion Medicine and Immunology Unit, Department of Transfusion Medicine, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| | - Francesco Pasquali
- Medical Genetics, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Sandra Perobelli
- Cystic Fibrosis Center, Department of Cardiovascular and Thoracic Surgery, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Gloria Tridello
- Pediatric Hematology and Oncology Unit, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Piero Farruggia
- Pediatric Hematology and Oncology Unit, Oncology Department, Azienda Ospedaliera di Rilieno Nazionale di Alta Specializzazione, Ospedale Civico, Palermo, Italy
| | - Marco Cipolli
- Cystic Fibrosis Center, Department of Cardiovascular and Thoracic Surgery, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
399
|
Shallis RM, Podoltsev NA, Gowda L, Zeidan AM, Gore SD. Cui bono? Finding the value of allogeneic stem cell transplantation for lower-risk myelodysplastic syndromes. Expert Rev Hematol 2020; 13:447-460. [PMID: 32182435 DOI: 10.1080/17474086.2020.1744433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction: The myelodysplastic syndromes (MDS) vary in their risk of disease progression; progression includes increasingly severe bone marrow failure, reclassification as acute myeloid leukemia (AML), and death. Prognostic tools guide recommendations for allogeneic stem cell transplantation (alloSCT), the only curative option. AlloSCT is typically reserved for patients with higher-risk MDS as defined by existing prognostic tools, although additional clinical and biological factors in lower-risk patients may influence this dogma.Areas covered: This review discusses the current understanding of MDS risk stratification as it pertains to the use of alloSCT in subpopulations of MDS patients with a particular focus on the use of alloSCT in patients with lower-risk disease.Expert commentary: Though high-quality data are lacking, some lower-risk MDS patients may benefit from alloSCT, which offers the only prospect of cure. Understanding the etiologic role and prognostic impact of recurring genetic events may improve existing risk stratification and become integral facets of prognostic schemata. The identification of additional factors influencing the prognoses of patients currently lumped together as 'lower-risk' will likewise improve the selection of MDS patients for early intervention or aggressive therapies such as alloSCT.
Collapse
Affiliation(s)
- Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Cancer Center, New Haven, CT, USA
| | - Nikolai A Podoltsev
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Cancer Center, New Haven, CT, USA
| | - Lohith Gowda
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Cancer Center, New Haven, CT, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Cancer Center, New Haven, CT, USA
| | - Steven D Gore
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Cancer Center, New Haven, CT, USA
| |
Collapse
|
400
|
Watson CJ, Papula AL, Poon GYP, Wong WH, Young AL, Druley TE, Fisher DS, Blundell JR. The evolutionary dynamics and fitness landscape of clonal hematopoiesis. Science 2020; 367:1449-1454. [PMID: 32217721 DOI: 10.1126/science.aay9333] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/24/2020] [Indexed: 12/15/2022]
Abstract
Somatic mutations acquired in healthy tissues as we age are major determinants of cancer risk. Whether variants confer a fitness advantage or rise to detectable frequencies by chance remains largely unknown. Blood sequencing data from ~50,000 individuals reveal how mutation, genetic drift, and fitness shape the genetic diversity of healthy blood (clonal hematopoiesis). We show that positive selection, not drift, is the major force shaping clonal hematopoiesis, provide bounds on the number of hematopoietic stem cells, and quantify the fitness advantages of key pathogenic variants, at single-nucleotide resolution, as well as the distribution of fitness effects (fitness landscape) within commonly mutated driver genes. These data are consistent with clonal hematopoiesis being driven by a continuing risk of mutations and clonal expansions that become increasingly detectable with age.
Collapse
Affiliation(s)
- Caroline J Watson
- Department of Oncology, University of Cambridge, Cambridge, UK.
- Early Detection Programme, CRUK Cambridge Cancer Centre, University of Cambridge, Cambridge, UK
| | - A L Papula
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Gladys Y P Poon
- Department of Oncology, University of Cambridge, Cambridge, UK
- Early Detection Programme, CRUK Cambridge Cancer Centre, University of Cambridge, Cambridge, UK
| | - Wing H Wong
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew L Young
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Todd E Druley
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Fisher
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Jamie R Blundell
- Department of Oncology, University of Cambridge, Cambridge, UK.
- Early Detection Programme, CRUK Cambridge Cancer Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|