1
|
Goldsmith SR, Shouse G, Wong FL, Bosworth A, Iukuridze A, Chen S, Rhee JW, Mei M, Htut M, Janakiram M, Forman SJ, Pillai R, Budde LE, Armenian SH. Clonal Hematopoiesis is Associated With Severe Cytokine Release Syndrome in Patients Treated With Chimeric Antigen Receptor T-Cell (CART) Therapy. Transplant Cell Ther 2024; 30:927.e1-927.e9. [PMID: 38871057 DOI: 10.1016/j.jtct.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
Among patients receiving CD19 or B-cell maturation antigen (BCMA) CAR T therapy, inflammation pre- and post-CAR T infusion is implicated in the development of toxicities including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and likely contributes to prolonged cytopenias. Clonal hematopoiesis (CH), the clonal expansion of hematopoietic stem cells harboring somatic mutations, has been associated with inflammasome upregulation. Herein, we examined the prevalence of pre-CAR T CH in a predominantly transplant-naïve cohort of recipients with non-Hodgkin lymphoma (NHL) or multiple myeloma (MM), and assessed the relationship between the presence of CH mutations and CAR T-related outcomes including CRS, ICANS, prolonged cytopenia, progression-free survival (PFS), and overall survival (OS). This study included 62 patients with NHL or MM who underwent CD19 or BCMA CAR T therapy from 2017 to 2022 at City of Hope and had available pre-CAR T cryopreserved peripheral blood mononuclear cells (PBMCs). DNA was isolated with QIAamp DNA Mini Kit (Qiagen) from PBMC samples (94% collected <30d of CART infusion), on which we performed targeted exome sequencing (108 pre-defined gene panel with 1000x sequencing depth) to determine the presence of CH (variant allele frequency [VAF] ≥2%). Multivariable logistic regression was used to examine the association between CH and absolute neutrophil count (ANC) recovery at day +30 and +60, maximum grade CRS and ICANS, grade <2 versus 2+, and OS and PFS at 1y. Covariates considered were age at CART, baseline ANC, sex, race, CAR-HEMATOTOX, LDH, bridging therapy (Y/N), and number of prior lines of therapy. Fifteen (24%) patients had at least one pathogenic CH mutation; 2 (13%) had ≥2 CH mutations concurrently. DMT3A mutations were the most common; 29% of mutations had VAFs >10%. Patients with CH were significantly more likely to develop grade ≥2 CRS (60% versus 28%, p = .03) compared to those without CH (odds ratio [OR] 3.9, 95% CI 1.2-13.2; p = .027). Accounting for baseline ANC (which was higher among the CH cohort and associated with delayed ANC recovery, p = .02) patients with CH did not have a significantly different rate of delayed ANC recovery compared to those without CH (adjusted OR 0.37, 95% CI 0.09-1.5; p = .17). There was no association between CH and ICANS, nor with 1y PFS or OS. CH was frequent (24%) in this cohort of CAR T recipients and was associated with a higher risk of development of grade ≥2 CRS after CAR T. Additional validation studies are currently underway, which may set the stage for consideration of pre-CAR T CH as a biomarker for risk stratification towards more proactive CRS prophylaxis. Translational studies could aim to prove a direct relationship between CH-mutated myeloid cells and CRS.
Collapse
Affiliation(s)
- Scott R Goldsmith
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California.
| | - Geoffrey Shouse
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| | - F Lennie Wong
- Department of Population Sciences Program, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Alysia Bosworth
- Department of Population Sciences Program, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Aleksi Iukuridze
- Department of Population Sciences Program, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Sitong Chen
- Department of Population Sciences Program, City of Hope Comprehensive Cancer Center, Duarte, California
| | - June-Wha Rhee
- Department of Population Sciences Program, City of Hope Comprehensive Cancer Center, Duarte, California; Department of Medicine, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Matthew Mei
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Myo Htut
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Murali Janakiram
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Stephen J Forman
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Raju Pillai
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - L Elizabeth Budde
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Saro H Armenian
- Department of Population Sciences Program, City of Hope Comprehensive Cancer Center, Duarte, California; Department of Pediatrics, City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
2
|
Cesana D, Cicalese MP, Calabria A, Merli P, Caruso R, Volpin M, Rudilosso L, Migliavacca M, Barzaghi F, Fossati C, Gazzo F, Pizzi S, Ciolfi A, Bruselles A, Tucci F, Spinozzi G, Pais G, Benedicenti F, Barcella M, Merelli I, Gallina P, Giannelli S, Dionisio F, Scala S, Casiraghi M, Strocchio L, Vinti L, Pacillo L, Draghi E, Cesana M, Riccardo S, Colantuono C, Six E, Cavazzana M, Carlucci F, Schmidt M, Cancrini C, Ciceri F, Vago L, Cacchiarelli D, Gentner B, Naldini L, Tartaglia M, Montini E, Locatelli F, Aiuti A. A case of T-cell acute lymphoblastic leukemia in retroviral gene therapy for ADA-SCID. Nat Commun 2024; 15:3662. [PMID: 38688902 PMCID: PMC11061298 DOI: 10.1038/s41467-024-47866-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Hematopoietic stem cell gene therapy (GT) using a γ-retroviral vector (γ-RV) is an effective treatment for Severe Combined Immunodeficiency due to Adenosine Deaminase deficiency. Here, we describe a case of GT-related T-cell acute lymphoblastic leukemia (T-ALL) that developed 4.7 years after treatment. The patient underwent chemotherapy and haploidentical transplantation and is currently in remission. Blast cells contain a single vector insertion activating the LIM-only protein 2 (LMO2) proto-oncogene, confirmed by physical interaction, and low Adenosine Deaminase (ADA) activity resulting from methylation of viral promoter. The insertion is detected years before T-ALL in multiple lineages, suggesting that further hits occurred in a thymic progenitor. Blast cells contain known and novel somatic mutations as well as germline mutations which may have contributed to transformation. Before T-ALL onset, the insertion profile is similar to those of other ADA-deficient patients. The limited incidence of vector-related adverse events in ADA-deficiency compared to other γ-RV GT trials could be explained by differences in transgenes, background disease and patient's specific factors.
Collapse
Affiliation(s)
- Daniela Cesana
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Cicalese
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Merli
- IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Monica Volpin
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Rudilosso
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maddalena Migliavacca
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Fossati
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Gazzo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Tucci
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Spinozzi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Pais
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Barcella
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Pierangela Gallina
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Giannelli
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Dionisio
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Miriam Casiraghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Lucia Pacillo
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of Pediatrics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Eleonora Draghi
- Immunogenetics, Leukemia Genomics and Immunobiology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marcella Cesana
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Sara Riccardo
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- NEGEDIA S.r.l., Pozzuoli, Italy
| | - Chiara Colantuono
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- NEGEDIA S.r.l., Pozzuoli, Italy
| | - Emmanuelle Six
- Laboratory of Human Lympho-hematopoiesis, INSERM, Paris, France
| | | | - Filippo Carlucci
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Caterina Cancrini
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of Pediatrics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine University of Rome Tor Vergata, Rome, Italy
| | - Fabio Ciceri
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
- Haematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- Università Vita-Salute San Raffaele, Milan, Italy
- Immunogenetics, Leukemia Genomics and Immunobiology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132, Milan, Italy
- Haematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples "Federico II", Naples, Italy
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Haematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
3
|
Calvete O, Mestre J, Risueño RM, Manzanares A, Acha P, Xicoy B, Solé F. Two-Time Multiplexed Targeted Next-Generation Sequencing Might Help the Implementation of Germline Screening Tools for Myelodysplastic Syndromes/Hematologic Neoplasms. Biomedicines 2023; 11:3222. [PMID: 38137443 PMCID: PMC10740751 DOI: 10.3390/biomedicines11123222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Next-generation sequencing (NGS) tools have importantly helped the classification of myelodysplastic syndromes (MDS), guiding the management of patients. However, new concerns are under debate regarding their implementation in routine clinical practice for the identification of germline predisposition. Cost-effective targeted NGS tools would improve the current standardized studies and genetic counseling. Here, we present our experience in a preliminary study detecting variants using a two-time multiplexed library strategy. Samples from different MDS patients were first mixed before library preparation and later multiplexed for a sequencing run. Two different mixes including a pool of three (3×) and four (4×) samples were evaluated. The filtered variants found in the individually sequenced samples were compared with the variants found in the two-time multiplexed studies to determine the detection efficiency scores. The same candidate variants were found in the two-time multiplexed studies in comparison with the individual tNGS. The variant allele frequency (VAF) values of the candidate variants were also compared. No significant differences were found between the expected and observed VAF percentages in both the 3× (p-value 0.74) and 4× (p-value 0.34) multiplexed studies. Our preliminary results suggest that the two-time multiplexing strategy might have the potential to help reduce the cost of evaluating germline predisposition.
Collapse
Affiliation(s)
- Oriol Calvete
- MDS Group, Josep Carreras Leukaemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Julia Mestre
- MDS Group, Josep Carreras Leukaemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
- Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Ruth M. Risueño
- Leukos Biotech, 08021 Barcelona, Spain
- Faculty of Education, University of Atlántico Medio, 35017 Las Palmas, Spain
| | - Ana Manzanares
- MDS Group, Josep Carreras Leukaemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Pamela Acha
- MDS Group, Josep Carreras Leukaemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Blanca Xicoy
- Hematology Service, ICO-Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Francesc Solé
- MDS Group, Josep Carreras Leukaemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| |
Collapse
|
4
|
Renneville A, Bernard E, Micol JB. Therapy-related myelodysplastic syndromes in the genomics era. Bull Cancer 2023; 110:1129-1140. [PMID: 37391357 DOI: 10.1016/j.bulcan.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 07/02/2023]
Abstract
Therapy-related myelodysplastic syndromes (t-MDS) represent a heterogeneous group of malignancies that arise as a late complication of prior exposure to chemotherapy and/or radiotherapy administered for a primary condition. T-MDS account for approximately 20% of all MDS and are characterized by resistance to current treatment strategies and poor prognosis. Our understanding of t-MDS pathogenesis has considerably improved over the last 5 years with the availability of deep sequencing technologies. T-MDS development is now considered as a multifactorial process resulting from complex interactions between an underlying germline genetic susceptibility, the stepwise acquisition of somatic mutations in hematopoietic stem cells, the clonal selection pressure exerted by cytotoxic therapies, and alterations of the bone marrow microenvironment. The survival of patients with t-MDS is generally poor. This can be explained by both patient-related factors including poor performance status and less tolerance to treatment and disease-related factors, such as the presence of chemoresistant clones, high-risk cytogenetic alterations and molecular features (e.g. high frequency of TP53 mutations). Around 50% of t-MDS patients are classified as high/very high risk based on IPSS-R or IPSS-M scores, versus 30% in de novo MDS. Long-term survival is only achieved in a minority of t-MDS patients who receive allogeneic stem cell transplantation, but the development of novel drugs may open new therapeutic opportunities, especially in unfit patients. Further investigations are needed to improve the identification of patients at higher risk of developing t-MDS and determine whether primary disease treatment can be modified to prevent the occurrence of t-MDS.
Collapse
Affiliation(s)
- Aline Renneville
- Gustave-Roussy, Department of Medical Biology and Pathology, Villejuif, France; Gustave-Roussy, université Paris-Saclay, Inserm U1287, Villejuif, France
| | - Elsa Bernard
- Gustave Roussy, université Paris-Saclay, Inserm U981, Villejuif, France; Memorial Sloan Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, USA
| | - Jean-Baptiste Micol
- Gustave-Roussy, université Paris-Saclay, Inserm U1287, Villejuif, France; Gustave-Roussy, université Paris-Saclay, Department of Hematology, Villejuif, France; Gustave-Roussy, université Paris-Saclay, Interception Program, Personalized Cancer Prevention Center, Villejuif, France.
| |
Collapse
|
5
|
Falconi G, Galossi E, Hajrullaj H, Fabiani E, Voso MT. Bone Marrow Microenvironment Involvement in t-MN: Focus on Mesenchymal Stem Cells. Mediterr J Hematol Infect Dis 2023; 15:e2023055. [PMID: 37705521 PMCID: PMC10497308 DOI: 10.4084/mjhid.2023.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 09/15/2023] Open
Abstract
Therapy-related myeloid neoplasms (t-MN) are a late complication of cytotoxic therapy (CT) used in the treatment of both malignant and non-malignant diseases. Historically, t-MN has been considered to be a direct consequence of DNA damage induced in normal hematopoietic stem or progenitor cells (HSPC) by CT. However, we now know that treatment-induced mutations in HSC are not the only players involved in t-MN development, but additional factors may contribute to the onset of t-MN. One of the known drivers involved in this field is the bone marrow microenvironment (BMM) and, in particular, bone marrow mesenchymal stem cells (BM-MSC), whose role in t-MN pathogenesis is the topic of this mini-review. BM-MSCs, physiologically, support HSC maintenance, self-renewal, and differentiation through hematopoietic-stromal interactions and the production of cytokines. In addition, BM-MSCs maintain the stability of the BM immune microenvironment and reduce the damage caused to HSC by stress stimuli. In the t-MN context, chemo/radiotherapy may induce damage to the BM-MSC and likewise alter BM-MSC functions by promoting pro-inflammatory response, clonal selection and/or the production of factors that may favor malignant hematopoiesis. Over the last decade, it has been shown that BM-MSC isolated from patients with de novo and therapy-related MN exhibit decreased proliferative and clonogenic capacity, altered morphology, increased senescence, defective osteogenic differentiation potential, impaired immune-regulatory properties, and reduced ability to support HSC growth and differentiation, as compared to normal BM-MSC. Although the understanding of the genetic and gene expression profile associated with ex vivo-expanded t-MN-MSCs remains limited and debatable, its potential role in prognostic and therapeutic terms is acting as a flywheel of attraction for many researchers.
Collapse
Affiliation(s)
- Giulia Falconi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - E Galossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - H Hajrullaj
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - E Fabiani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - M T Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
6
|
Calvete O, Mestre J, Jerez A, Solé F. The Secondary Myelodysplastic Neoplasms (MDS) Jigsaw. Cancers (Basel) 2023; 15:1483. [PMID: 36900275 PMCID: PMC10000488 DOI: 10.3390/cancers15051483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/08/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
There is a great deal of controversy in the hematologic community regarding the classification of secondary myelodysplastic neoplasms (MDSs). Current classifications are based on the presence of genetic predisposition and MDS post-cytotoxic therapy (MDS-pCT) etiologies. However, since these risk factors are not exclusive for secondary MDSs and there are multiple overlapping scenarios, a comprehensive and definitive classification is yet to come. In addition, a sporadic MDS might arise after a primary tumor fulfills the diagnostic criteria of MDS-pCT without a causative cytotoxicity. In this review, we describe the triggering pieces of a secondary MDS jigsaw: previous cytotoxic therapy, germline predisposition and clonal hematopoiesis. Epidemiological and translational efforts are needed to put these pieces together and ascertain the real weight of each of these pieces in each MDS patient. Future classifications must contribute to understanding the role of secondary MDS jigsaw pieces in different concomitant or independent clinical scenarios associated with the primary tumor.
Collapse
Affiliation(s)
- Oriol Calvete
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Barcelona, Spain
| | - Julia Mestre
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Barcelona, Spain
| | - Andrés Jerez
- Experimental Hematology Unit, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, 08035 Barcelona, Spain
| | - Francesc Solé
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Barcelona, Spain
| |
Collapse
|
7
|
Ha H, Kim HJ, Park JH, Shin A, Lee KN, Han K, Lee NR, Hong J. Epidemiologic outlook of therapy-related myeloid neoplasms and selection of high-risk patients: A Korean nationwide study. Cancer 2022; 128:3888-3896. [PMID: 36069361 DOI: 10.1002/cncr.34453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Although a considerable proportion of patients with cancer receive chemotherapy (CT) or radiotherapy (RT), only a very few patients eventually develop therapy-related myeloid neoplasms (t-MNs). METHODS To identify subsets of cancer patients who have substantially elevated risk of developing t-MNs. Incidences and risks of t-MNs after contemporary CT or RT in patients newly diagnosed major cancers during 2009-2013 were analyzed. By merging two Korean nationwide health care big data sets, patients were selected and observed on follow-up to until t-MN development or December 2019. RESULTS Among 250,155 patients, 555 (0.22%) were diagnosed with t-MNs with a standard incidence ratio (SIR) of 3.40 (95% CI, 3.13-3.70). Patients had bone/joint cancers (SIR, 94.25; 95% CI, 50.71-137.80) and a remarkably high SIR for t-MN development. Patients receiving both CT and RT had the highest SIR (4.64; 95% CI, 4.08-5.20), followed by those receiving CT only (SIR, 3.30; 95% CI, 2.89-3.70). Contrarily, RT alone did not increase t-MN risk (SIR, 1.16; 95% CI, 0.76-1.56). More exposure to leukemogenic agents resulted in the higher t-MNs development. CONCLUSIONS The increased risk of developing acute myeloid leukemia or myelodysplastic syndrome after CT and/or RT was confirmed and subsets with substantially elevated risk for developing t-MNs were found. Such patients would be suitable for a prospective cohort for investigating t-MN pathogenesis by time series analyses.
Collapse
Affiliation(s)
- Hyerim Ha
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Inha University College of Medicine, Incheon, Republic of Korea
| | - Hyo Jeong Kim
- National Evidence-based Healthcare Collaborating Agency (NECA), Seoul, South Korea
| | - Ju Hyun Park
- Korea Institute for Health and Social Affairs (KIHASA), Sejong-si, South Korea
| | - Aesun Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyu Na Lee
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Na Rae Lee
- National Evidence-based Healthcare Collaborating Agency (NECA), Seoul, South Korea
| | - Junshik Hong
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Biomedical Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
Voso M, Pandzic T, Falconi G, Denčić‐Fekete M, De Bellis E, Scarfo L, Ljungström V, Iskas M, Del Poeta G, Ranghetti P, Laidou S, Cristiano A, Plevova K, Imbergamo S, Engvall M, Zucchetto A, Salvetti C, Mauro FR, Stavroyianni N, Cavelier L, Ghia P, Stamatopoulos K, Fabiani E, Baliakas P. Clonal haematopoiesis as a risk factor for therapy‐related myeloid neoplasms in patients with chronic lymphocytic leukaemia treated with chemo‐(immuno)therapy. Br J Haematol 2022; 198:103-113. [DOI: 10.1111/bjh.18129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/23/2022] [Indexed: 02/06/2023]
Affiliation(s)
- Maria‐Teresa Voso
- Department of Biomedicine and Prevention University of Tor Vergata Rome Italy
| | - Tatjana Pandzic
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala University Uppsala Sweden
- Department of Clinical Genetics Uppsala University Hospital Uppsala Sweden
| | - Giulia Falconi
- Department of Biomedicine and Prevention University of Tor Vergata Rome Italy
| | | | - Eleonora De Bellis
- Department of Biomedicine and Prevention University of Tor Vergata Rome Italy
| | - Lydia Scarfo
- Strategic Research Programme in CLL, Division of Experimental Oncology IRCCS Ospedale San Raffaele and Università Vita‐Salute San Raffaele Milan Italy
| | - Viktor Ljungström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala University Uppsala Sweden
- Department of Clinical Genetics Uppsala University Hospital Uppsala Sweden
| | - Michail Iskas
- Hematology Department and HCT Unit, G. Papanicolaou Hospital Thessaloniki Greece
| | - Giovanni Del Poeta
- Department of Biomedicine and Prevention University of Tor Vergata Rome Italy
| | - Pamela Ranghetti
- Strategic Research Programme in CLL, Division of Experimental Oncology IRCCS Ospedale San Raffaele and Università Vita‐Salute San Raffaele Milan Italy
| | - Stamatia Laidou
- Institute of Applied Biosciences Center for Research and Technology Hellas Thessaloniki Greece
| | - Antonio Cristiano
- Department of Biomedicine and Prevention University of Tor Vergata Rome Italy
| | - Karla Plevova
- Institute of Medical Genetics and Genomics Faculty of Medicine Masaryk University Brno Czech Republic
- Department of Internal Medicine – Haematology and Oncology University Hospital Brno Brno Czech Republic
| | - Silvia Imbergamo
- Hematology and Clinical Immunology, Department of Hematology Hospital of Padua Padua Italy
| | - Marie Engvall
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala University Uppsala Sweden
- Department of Clinical Genetics Uppsala University Hospital Uppsala Sweden
| | - Antonella Zucchetto
- Clinical and Experimental Onco‐Hematology Unit Centro di Riferimento Oncologico di Aviano (CRO), IRCCS Aviano Italy
| | | | - Francesca R. Mauro
- Department of Translational and Precision Medicine, Hematology 'Sapienza' University Rome Italy
| | - Niki Stavroyianni
- Hematology Department and HCT Unit, G. Papanicolaou Hospital Thessaloniki Greece
| | - Lucia Cavelier
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala University Uppsala Sweden
- Department of Clinical Genetics Uppsala University Hospital Uppsala Sweden
| | - Paolo Ghia
- Strategic Research Programme in CLL, Division of Experimental Oncology IRCCS Ospedale San Raffaele and Università Vita‐Salute San Raffaele Milan Italy
| | - Kostas Stamatopoulos
- Institute of Applied Biosciences Center for Research and Technology Hellas Thessaloniki Greece
- Department of Molecular Medicine and Surgery Karolinska Institutet Stockholm Sweden
| | - Emiliano Fabiani
- Department of Biomedicine and Prevention University of Tor Vergata Rome Italy
- UniCamillus‐Saint Camillus International University of Health Sciences Rome Italy
| | - Panagiotis Baliakas
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala University Uppsala Sweden
- Department of Clinical Genetics Uppsala University Hospital Uppsala Sweden
| |
Collapse
|
9
|
Pillai RK, Afkhami M. Advances in Diagnosis and Risk Stratification of Acute Myeloid Leukemia and Myelodysplastic Syndromes. Cancer Treat Res 2021; 181:1-16. [PMID: 34626352 DOI: 10.1007/978-3-030-78311-2_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Advances in high-throughput DNA sequencing technology in the past decade have made a tremendous impact on basic science and clinical practice. Methods using the latest next generation sequencing technology can sequence an entire human genome within a few hours. Diagnosis and prognostication of hematologic neoplasms have moved from traditional histology and immunophenotyping to integration of cytogenetic and genomic alterations. Using illustrative cases, this chapter provides an overview of the utility of using genomic data for prognostication as well as treatment decision-making for patients with bone marrow neoplasms.
Collapse
Affiliation(s)
- Raju K Pillai
- City of Hope Medical Center, 1500 E Duarte Rd, Duarte, CA, 91010, USA.
| | - Michelle Afkhami
- City of Hope Medical Center, 1500 E Duarte Rd, Duarte, CA, 91010, USA
| |
Collapse
|
10
|
Lin SH, Wang Y, Hartley SW, Karyadi DM, Lee OW, Zhu B, Zhou W, Brown DW, Beilstein-Wedel E, Hazra R, Kacanek D, Chadwick EG, Marsit CJ, Poirier MC, Brummel SS, Chanock SJ, Engels EA, Machiela MJ. In-utero exposure to zidovudine-containing antiretroviral therapy and clonal hematopoiesis in HIV-exposed uninfected newborns. AIDS 2021; 35:1525-1535. [PMID: 33756513 PMCID: PMC8286286 DOI: 10.1097/qad.0000000000002894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Zidovudine (ZDV) has been extensively used in pregnant women to prevent vertical transmission of HIV but few studies have evaluated potential mutagenic effects of ZDV during fetal development. DESIGN Our study investigated clonal hematopoiesis in HIV-exposed uninfected (HEU) newborns, 94 of whom were ZDV-exposed and 91 antiretroviral therapy (ART)-unexposed and matched for potential confounding factors. METHODS Utilizing high depth sequencing and genotyping arrays, we comprehensively examined blood samples collected during the first week after birth for potential clonal hematopoiesis associated with fetal ZDV exposure, including clonal single nucleotide variants (SNVs), small insertions and deletions (indels), and large structural copy number or copy neutral alterations. RESULTS We observed no statistically significant difference in the number of SNVs and indels per person in ZDV-exposed children (adjusted ratio [95% confidence interval, CI] for expected number of mutations = 0.79 [0.50--1.22], P = 0.3), and no difference in the number of large structural alterations. Mutations in common clonal hematopoiesis driver genes were not found in the study population. Mutational signature analyses on SNVs detected no novel signatures unique to the ZDV-exposed children and the mutational profiles were similar between the two groups. CONCLUSION Our results suggest that clonal hematopoiesis at levels detectable in our study is not strongly influenced by in-utero ZDV exposure; however, additional follow-up studies are needed to further evaluate the safety and potential long-term impacts of in-utero ZDV exposure in HEU children as well as better investigate genomic aberrations occurring late in pregnancy.
Collapse
Affiliation(s)
- Shu-Hong Lin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Youjin Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Stephen W Hartley
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Danielle M Karyadi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Olivia W Lee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Weiyin Zhou
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Derek W Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Erin Beilstein-Wedel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rohan Hazra
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Deborah Kacanek
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ellen G Chadwick
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Carmen J Marsit
- Departments of Environmental Health and Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Miriam C Poirier
- Carcinogen-DNA Interactions Section, Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sean S Brummel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| |
Collapse
|
11
|
|
12
|
What's new in the pathogenesis and treatment of therapy-related myeloid neoplasms. Blood 2021; 138:749-757. [PMID: 33876223 DOI: 10.1182/blood.2021010764] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/02/2021] [Indexed: 12/14/2022] Open
Abstract
Therapy-related myeloid neoplasms (t-MN) include diseases onsetting in patients treated with chemo- and/or radiotherapy for a primary cancer, or an autoimmune disorder. Genomic variants, in particular in familial cancer genes, may play a predisposing role. Recent advances in deep sequencing techniques have shed light on the pathogenesis of t-MN, identifying clonal hematopoiesis of indeterminate potential (CHIP) as a frequent first step in the multi-hit model of t-MN. CHIP is often detectable prior to any cytotoxic treatment, probably setting the fertile genomic background for secondary leukemogenesis. The evolution pattern towards t-MN is then a complex process, shaped by the type of cancer therapy, the aging process, and the individual exposures, that favor additional hits, such as the acquisition of TP53 mutations and unfavorable karyotype abnormalities. The pathogenesis of t-MN differs from MN associated with environmental exposure. Indeed, the genetic aberration patterns of MN developing in atomic bomb survivors show few mutations in classical DNA methylation genes, and a high prevalence of 11q and ATM alterations, together with TP53 mutations. Survival in t-MN is poor. In addition to the biology of t-MN, the patient's previous disease history and the remission status at t-MN diagnosis are significant factors contributing to unfavorable outcome. New drugs active in secondary leukemias include CPX-351, or venetoclax in combination with hypomethylating agents, monoclonal antibodies as magrolimab, or targeted drugs against pathogenic mutations. Allogeneic stem cell transplantation remains the best currently available therapeutic option with curative intent for fit patients with unfavorable genetic profiles.
Collapse
|
13
|
From Clonal Hematopoiesis to Therapy-Related Myeloid Neoplasms: The Silent Way of Cancer Progression. BIOLOGY 2021; 10:biology10020128. [PMID: 33562056 PMCID: PMC7914896 DOI: 10.3390/biology10020128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/29/2022]
Abstract
Simple Summary In the last decades the improved management of cancer patients and the overall prolonged life expectancy contributed to the increased number of patients at risk of late clonal events such as therapy-related myeloid neoplasms (t-MN). The discovery of clonal hematopoiesis of indeterminate potential (CHIP) in normal individuals has shed light on the pathophysiologic mechanism behind the process of myeloid evolution, defining CHIP carriers at higher risk of progression. Moreover, different patterns of clonal evolution have been identified in case of t-MN development after anti-cancer treatment exposure. The growing body of evidence in this field allowed the creation of dedicated cancer survivorship programs and “CHIP-Clinics” in order to specifically address the issue of CHIP in patients undergoing anti-cancer treatment and develop measure of early detection possibly guiding tumor surveillance. Abstract Clonal hematopoiesis (CH) has been recognized as a predisposing factor for the development of myeloid malignancies. Its detection has been reported at different frequencies across studies, based on the type of genome scanning approach used and the population studied, but the latest insights recognize its virtual ubiquitous presence in older individuals. The discovery of CH in recent years paved the way for a shift in the paradigm of our understanding of the biology of therapy-related myeloid malignancies (t-MNs). Indeed, we moved from the concept of a treatment-induced lesion to a model where CH precedes the commencement of any cancer-related treatment in patients who subsequently develop a t-MN. Invariant patterns of genes seem to contribute to the arising of t-MN cases, with differences regarding the type of treatment received. Here, we review the principal studies concerning CH, the relationship with myeloid progression and the mechanisms of secondary t-MN development.
Collapse
|
14
|
Husby S, Hjermind Justesen E, Grønbæk K. Protein phosphatase, Mg 2+/Mn 2+-dependent 1D (PPM1D) mutations in haematological cancer. Br J Haematol 2020; 192:697-705. [PMID: 33616916 DOI: 10.1111/bjh.17120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/02/2020] [Indexed: 01/07/2023]
Abstract
Until recently, the protein phosphatase, Mg2+/Mn2+-dependent 1D (PPM1D) gene had not been examined in haematological cancer, but several studies have now explored the functional role of this gene and its aberrations. It is often mutated in the context of clonal haemopoiesis (including in patients with lymphoma, myeloproliferative neoplasms and myelodysplastic syndrome) and mutations have been associated with exposure to cytotoxic and radiation therapy, development of therapy-related neoplasms and inferior survival. The vast majority of PPM1D mutations found in haematopoietic cells are of the nonsense or frameshift type and located within terminal exon 6. These genetic defects are rarely found in the blood of healthy individuals. PPM1D encodes the PPM1D phosphatase [also named wild-type p53-induced phosphatase 1 (WIP1)], which negatively regulates signalling molecules within the DNA damage response pathway, including tumour suppressor p53. Clonal expansion of PPM1D mutant haematopoietic cells can potentially be prevented with inhibitors; however, human trials are awaited. In the present review, we provide a review of the literature regarding PPM1D and its role in haematological cancer.
Collapse
Affiliation(s)
- Simon Husby
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Emma Hjermind Justesen
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Maclachlan K, Diamond B, Maura F, Hillengass J, Turesson I, Landgren CO, Kazandjian D. Second malignancies in multiple myeloma; emerging patterns and future directions. Best Pract Res Clin Haematol 2020; 33:101144. [PMID: 32139010 PMCID: PMC7544243 DOI: 10.1016/j.beha.2020.101144] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 12/20/2022]
Abstract
The changing landscape of treatment options for multiple myeloma has led to a higher proportion of patients achieving deep, long-lasting responses to therapy. With the associated improvement in overall survival, the development of subsequent second malignancies has become of increased significance. The risk of second malignancy after multiple myeloma is affected by a combination of patient-, disease- and therapy-related risk factors. This review discusses recent data refining our knowledge of these contributing factors, including current treatment modalities which increase risk (i.e. high-dose melphalan with autologous stem cell transplant and lenalidomide maintenance therapy). We highlight emerging data towards individualized risk- and response-adapted treatment strategies and discuss key areas requiring future research.
Collapse
Affiliation(s)
- Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Benjamin Diamond
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francesco Maura
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jens Hillengass
- Section of Multiple Myeloma, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ingemar Turesson
- Department of Hematology, Skane University Hospital, Malmo, Sweden
| | - C Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dickran Kazandjian
- Multiple Myeloma Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Park SJ, Bejar R. Clonal hematopoiesis in cancer. Exp Hematol 2020; 83:105-112. [PMID: 32044376 DOI: 10.1016/j.exphem.2020.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/16/2022]
Abstract
Clonal hematopoiesis is a common premalignant condition defined by the abnormal expansion of clonally derived hematopoietic stem cells carrying somatic mutations in leukemia-associated genes. Apart from increasing age, this phenomenon occurs with higher frequency in individuals with lymphoid or solid tumors and is associated with exposures to genotoxic stress. Clonal hematopoiesis in this context confers a greater risk for developing therapy-related myeloid neoplasms and appears to contribute to adverse cancer-related survival through a variety of potential mechanisms. These include alterations of the bone marrow microenvironment, inflammatory changes in clonal effector cells and modulation of immune responses. Understanding how clonal hematopoiesis drives therapy-related myeloid neoplasm initiation and interactions with non-myeloid malignancies will inform screening and surveillance approaches and suggest targeted therapies in this vulnerable population. Here, we examine the clinical implications of clonal hematopoiesis in the cancer setting and discuss potential strategies to mitigate the adverse consequences of clonal expansion.
Collapse
Affiliation(s)
- Soo J Park
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Rafael Bejar
- Moores Cancer Center, University of California San Diego, La Jolla, CA.
| |
Collapse
|
17
|
Palmieri R, Paterno G, De Bellis E, Mercante L, Buzzatti E, Esposito F, Del Principe MI, Maurillo L, Buccisano F, Venditti A. Therapeutic Choice in Older Patients with Acute Myeloid Leukemia: A Matter of Fitness. Cancers (Basel) 2020; 12:cancers12010120. [PMID: 31906489 PMCID: PMC7016986 DOI: 10.3390/cancers12010120] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/19/2019] [Accepted: 12/31/2019] [Indexed: 01/17/2023] Open
Abstract
Acute myeloid leukemia (AML), with an incidence increasing with age, is the most common acute leukemia in adults. Concurrent comorbidities, mild to severe organ dysfunctions, and low performance status (PS) are frequently found in older patients at the onset, conditioning treatment choice and crucially influencing the outcome. Although anthracyclines plus cytarabine-based chemotherapy, also called “7 + 3” regimen, remains the standard of care in young adults, its use in patients older than 65 years should be reserved to selected cases because of higher incidence of toxicity. These adverse features of AML in the elderly underline the importance of a careful patient assessment at diagnosis as a critical tool in the decision-making process of treatment choice. In this review, we will describe selected recently approved drugs as well as examine prognostic algorithms that may be helpful to assign treatment in elderly patients properly.
Collapse
Affiliation(s)
- Raffaele Palmieri
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Giovangiacinto Paterno
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Eleonora De Bellis
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Lisa Mercante
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Elisa Buzzatti
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Fabiana Esposito
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Maria Ilaria Del Principe
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Luca Maurillo
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Francesco Buccisano
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
- Correspondence: ; Tel.: +39-0620903226
| |
Collapse
|
18
|
Clonal Hematopoiesis and risk of Acute Myeloid Leukemia. Best Pract Res Clin Haematol 2019; 32:177-185. [PMID: 31203999 DOI: 10.1016/j.beha.2019.05.007] [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: 05/19/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022]
Abstract
Acute Myeloid Leukemia, the most common form of acute leukemia in adults, is an aggressive hematopoietic stem cell malignancy that is associated with significant morbidity and mortality. Though AML generally presents de novo, risk factors include exposure to chemotherapy and/or radiation, as well as both familial and acquired bone marrow failure syndromes. Clonal Hematopoiesis (CH) refers to an expansion of blood or marrow cells resulting from somatic mutations in leukemia-associated genes detected in individuals without cytopenias or hematological malignancies. While CH is considered part of normal ageing, CH is also significantly associated with cardiovascular disease, solid tumors, and hematological malignancies. In this review, we will discuss evidence linking CH with the development of AML, as well as describe challenges in and strategies for monitoring patients with high risk CH mutations.
Collapse
|