201
|
Hare JM, Beerman I. Potential of Stem Cell-Based Therapy to Restore Function in Aging Systems: Are We There Yet? J Gerontol A Biol Sci Med Sci 2022; 77:1292-1294. [PMID: 34984451 PMCID: PMC9255684 DOI: 10.1093/gerona/glac003] [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: 12/20/2021] [Indexed: 01/07/2023] Open
Abstract
While there is extensive interest in geroscience approaches to health and disease, few basic science discoveries have made their way into clinical trials. Herein, we comment on cell-based therapies, in which supplementing robust stem cell capacity to aged systems theoretically could lead to sustained improvement. This exciting approach has undergone translational development, and we highlight studies targeting a single system and others aimed at treating overall aging frailty by restoring the aged stem cell niches that underly diminished endogenous regenerative capacity.
Collapse
Affiliation(s)
- Joshua M Hare
- The Interdisciplinary Stem Cell Institute, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Isabel Beerman
- Address correspondence to: Isabel Beerman, PhD, Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, BRC, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD 21224, USA. E-mail:
| |
Collapse
|
202
|
Christen F, Hablesreiter R, Hoyer K, Hennch C, Maluck-Böttcher A, Segler A, Madadi A, Frick M, Bullinger L, Briest F, Damm F. Modeling clonal hematopoiesis in umbilical cord blood cells by CRISPR/Cas9. Leukemia 2022; 36:1102-1110. [PMID: 34782715 PMCID: PMC8979818 DOI: 10.1038/s41375-021-01469-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/17/2022]
Abstract
To investigate clonal hematopoiesis associated gene mutations in vitro and to unravel the direct impact on the human stem and progenitor cell (HSPC) compartment, we targeted healthy, young hematopoietic progenitor cells, derived from umbilical cord blood samples, with CRISPR/Cas9 technology. Site-specific mutations were introduced in defined regions of DNMT3A, TET2, and ASXL1 in CD34+ progenitor cells that were subsequently analyzed in short-term as well as long-term in vitro culture assays to assess self-renewal and differentiation capacities. Colony-forming unit (CFU) assays revealed enhanced self-renewal of TET2 mutated (TET2mut) cells, whereas ASXL1mut as well as DNMT3Amut cells did not reveal significant changes in short-term culture. Strikingly, enhanced colony formation could be detected in long-term culture experiments in all mutants, indicating increased self-renewal capacities. While we could also demonstrate preferential clonal expansion of distinct cell clones for all mutants, the clonal composition after long-term culture revealed a mutation-specific impact on HSPCs. Thus, by using primary umbilical cord blood cells, we were able to investigate epigenetic driver mutations without confounding factors like age or a complex mutational landscape, and our findings provide evidence for a direct impact of clonal hematopoiesis-associated mutations on self-renewal and clonal composition of human stem and progenitor cells.
Collapse
Affiliation(s)
- Friederike Christen
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Raphael Hablesreiter
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Kaja Hoyer
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Cornelius Hennch
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Antje Maluck-Böttcher
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Angela Segler
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Gynecology with Center for Oncological Surgery, Berlin, Germany ,grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Berlin, Germany
| | - Annett Madadi
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Obstetrics, Berlin, Germany
| | - Mareike Frick
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Bullinger
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Franziska Briest
- grid.7468.d0000 0001 2248 7639Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Frederik Damm
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
203
|
Kusne Y, Xie Z, Patnaik MM. Clonal Hematopoiesis: Molecular and Clinical Implications. Leuk Res 2022; 113:106787. [DOI: 10.1016/j.leukres.2022.106787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 11/16/2022]
|
204
|
Cobo I, Tanaka T, Glass CK, Yeang C. Clonal hematopoiesis driven by DNMT3A and TET2 mutations: role in monocyte and macrophage biology and atherosclerotic cardiovascular disease. Curr Opin Hematol 2022; 29:1-7. [PMID: 34654019 PMCID: PMC8639635 DOI: 10.1097/moh.0000000000000688] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Clonal hematopoiesis of indeterminate potential (CHIP), defined by the presence of somatic mutations in hematopoietic cells, is associated with advanced age and increased mortality due to cardiovascular disease. Gene mutations in DNMT3A and TET2 are the most frequently identified variants among patients with CHIP and provide selective advantage that spurs clonal expansion and myeloid skewing. Although DNMT3A and TET2 appear to have opposing enzymatic influence on DNA methylation, mounting data has characterized convergent inflammatory pathways, providing insights to how CHIP may mediate atherosclerotic cardiovascular disease (ASCVD). RECENT FINDINGS We review a multitude of studies that characterize aberrant inflammatory signaling as result of DNMT3A and TET2 deficiency in monocytes and macrophages, immune cells with prominent roles in atherosclerosis. Although specific DNA methylation signatures associated with these known epigenetic regulators have been identified, many studies have also characterized diverse modulatory functions of DNTM3A and TET2 that urge cell and context-specific experimental studies to further define how DNMT3A and TET2 may nonenzymatically activate inflammatory pathways with clinically meaningful consequences. SUMMARY CHIP, common in elderly individuals, provides an opportunity understand and potentially modify age-related chronic inflammatory ASCVD risk.
Collapse
Affiliation(s)
- Isidoro Cobo
- Department of Cellular and Molecular Medicine, University of California San Diego
| | - Tiffany Tanaka
- University of California San Diego, Moores Cancer Center
| | - Christopher K. Glass
- Department of Cellular and Molecular Medicine, University of California San Diego
| | - Calvin Yeang
- Sulpizio Cardiovascular Center, Division of Cardiology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
205
|
Visconte V, Maciejewski JP. Clonal dynamics of hematopoietic stem cell compartment in aplastic anemia. Semin Hematol 2022; 59:47-53. [DOI: 10.1053/j.seminhematol.2021.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 11/11/2022]
|
206
|
Genetic Profiles and Risk Stratification in Adult De Novo Acute Myeloid Leukaemia in Relation to Age, Gender, and Ethnicity: A Study from Malaysia. Int J Mol Sci 2021; 23:ijms23010258. [PMID: 35008684 PMCID: PMC8745150 DOI: 10.3390/ijms23010258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Hitherto, no data describing the heterogeneity of genetic profiles and risk stratifications of adult acute myeloid leukaemia (AML) in Southeast Asia are reported. This study assessed genetic profiles, Moorman’s hierarchical classification, and ELN 2017-based risk stratifications in relation to age, gender, and ethnicity in Malaysian adult AML patients. A total of 854 AML patients: male (52%), female (48%) were recruited comprising three main ethnic groups: Malays (59%), Chinese (32%) and Indians (8%). Of 307 patients with abnormal karyotypes: 36% exhibited translocations; 10% deletions and 5% trisomies. The commonest genotype was FLT3-ITD-NPM1wt (276/414; 66.7%). ELN 2017 risk stratification was performed on 494 patients, and 41% were classified as favourable, 39% as intermediate and 20% as adverse groups. More females (47%) were in the favourable risk group compared to males (37%), whereas adverse risk was higher in patients above 60 (24%) of age compared to below 60 (18%) patients. We observed heterogeneity in the distribution of genetic profiles and risk stratifications between the age groups and gender, but not among the ethnic groups. Our study elucidated the diversity of adult AML genetic profiles between Southeast Asians and other regions worldwide.
Collapse
|
207
|
Benard BA, Leak LB, Azizi A, Thomas D, Gentles AJ, Majeti R. Clonal architecture predicts clinical outcomes and drug sensitivity in acute myeloid leukemia. Nat Commun 2021; 12:7244. [PMID: 34903734 PMCID: PMC8669028 DOI: 10.1038/s41467-021-27472-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/17/2021] [Indexed: 12/17/2022] Open
Abstract
The impact of clonal heterogeneity on disease behavior or drug response in acute myeloid leukemia remains poorly understood. Using a cohort of 2,829 patients, we identify features of clonality associated with clinical features and drug sensitivities. High variant allele frequency for 7 mutations (including NRAS and TET2) associate with dismal prognosis; elevated GATA2 variant allele frequency correlates with better outcomes. Clinical features such as white blood cell count and blast percentage correlate with the subclonal abundance of mutations such as TP53 and IDH1. Furthermore, patients with cohesin mutations occurring before NPM1, or transcription factor mutations occurring before splicing factor mutations, show shorter survival. Surprisingly, a branched pattern of clonal evolution is associated with superior clinical outcomes. Finally, several mutations (including NRAS and IDH1) predict drug sensitivity based on their subclonal abundance. Together, these results demonstrate the importance of assessing clonal heterogeneity with implications for prognosis and actionable biomarkers for therapy.
Collapse
Affiliation(s)
- Brooks A Benard
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA
- Cancer Biology Program, Stanford University, Stanford, CA, USA
| | - Logan B Leak
- Cancer Biology Program, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Armon Azizi
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA
| | - Daniel Thomas
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Andrew J Gentles
- Department of Medicine (Biomedical Informatics/Quantitative Sciences unit), Stanford University, Stanford, CA, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
208
|
Porazzi P, Petruk S, Pagliaroli L, De Dominici M, Deming D, Puccetti MV, Kushinsky S, Kumar G, Minieri V, Barbieri E, Deliard S, Grande A, Trizzino M, Gardini A, Canaani E, Palmisiano N, Porcu P, Ertel A, Fortina PM, Eischen CM, Mazo A, Calabretta B. Targeting chemotherapy to de-condensed H3K27me3-marked chromatin of AML cells enhances leukemia suppression. Cancer Res 2021; 82:458-471. [PMID: 34903608 DOI: 10.1158/0008-5472.can-21-1297] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 09/15/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022]
Abstract
Despite treatment with intensive chemotherapy, acute myeloid leukemia (AML) remains an aggressive malignancy with a dismal outcome in most patients. We found that AML cells exhibit an unusually rapid accumulation of the repressive histone mark H3K27me3 on nascent DNA. In cell lines, primary cells and xenograft mouse models, inhibition of the H3K27 histone methyltransferase EZH2 to de-condense the H3K27me3-marked chromatin of AML cells enhanced chromatin accessibility and chemotherapy-induced DNA damage, apoptosis, and leukemia suppression. These effects were further promoted when chromatin de-condensation of AML cells was induced upon S-phase entry after release from a transient G1 arrest mediated by CDK4/6 inhibition. In the p53-null KG-1 and THP-1 AML cell lines, EZH2 inhibitor and doxorubicin co-treatment induced transcriptional reprogramming that was, in part, dependent on de-repression of H3K27me3-marked gene promoters and led to increased expression of cell death-promoting and growth-inhibitory genes. In conclusion, decondensing H3K27me3-marked chromatin by EZH2 inhibition represents a promising approach to improve the efficacy of DNA-damaging cytotoxic agents in AML patients. This strategy might allow for a lowering of chemotherapy doses with a consequent reduction of treatment-related side effects in elderly AML patients or those with significant comorbidities.
Collapse
Affiliation(s)
- Patrizia Porazzi
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University
| | - Svetlana Petruk
- Department of Biochemistry and Molecular Biology and Kimmel Cancer Center,, Thomas Jefferson University
| | - Luca Pagliaroli
- Department of Biochemistry and Molecular Biology and Sidney Kimmel Cancer Center,, Thomas Jefferson University
| | | | - David Deming
- Department of Biochemistry and Molecular Biology and Kimmel Cancer Center,, Thomas Jefferson University
| | - Matthew V Puccetti
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University
| | - Saul Kushinsky
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University
| | - Gaurav Kumar
- Department of Cancer Biology, Thomas Jefferson University
| | - Valentina Minieri
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University
| | - Elisa Barbieri
- Gene Expression and Regulation Program, The Wistar Institute
| | - Sandra Deliard
- Gene Expression and Regulation Program, The Wistar Institute
| | - Alexis Grande
- Department of Life Sciences, University of Modena and Reggio Emilia
| | - Marco Trizzino
- Department of Biochemistry and Molecular Biology and Kimmel Cancer Center,, Thomas Jefferson University
| | | | - Eli Canaani
- The Department of Molecular Cell Biology, Weizmann Institute of Science
| | | | | | - Adam Ertel
- Department of Cancer Biology, Thomas Jefferson University
| | | | | | - Alexander Mazo
- Department of Biochemistry and Molecular Biology and Kimmel Cancer Center,, Thomas Jefferson University
| | - Bruno Calabretta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University
| |
Collapse
|
209
|
Guo X, Li J, Xue J, Fenech M, Wang X. Loss of Y chromosome: An emerging next-generation biomarker for disease prediction and early detection? MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 788:108389. [PMID: 34893154 DOI: 10.1016/j.mrrev.2021.108389] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 12/25/2022]
Abstract
As human life expectancy increases substantially and aging is the primary risk factor for most chronic diseases, there is an urgent need for advancing the development of post-genomic era biomarkers that can be used for disease prediction and early detection (DPED). Mosaic loss of Y chromosome (LOY) is the state of nullisomy Y in sub-groups of somatic cells acquired from different post-zygotic development stages and onwards throughout the lifespan. Multiple large-cohort based epidemiology studies have found that LOY in blood cells is a significant risk factor for future mortality and various diseases in males. Many features intrinsic to LOY analysis may be leveraged to enhance its use as a non-invasive, sensitive, reliable, high throughput-biomarker for DPED. Here, we review the emerging literatures in LOY studies and highlight ten strengths for using LOY as a novel biomarker for genomics-driven DPED diagnostics. Meanwhile, the current limitations in this area are also discussed. We conclude by identifying some important knowledge gaps regarding the consequences of malsegregation of the Y chromosome and propose further steps that are required before clinical implementation of LOY. Taken together, we think that LOY has substantial potential as a biomarker for DPED, despite some hurdles that still need to be addressed before its integration into healthcare becomes acceptable.
Collapse
Affiliation(s)
- Xihan Guo
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan, 650500, China; Yunnan Environmental Mutagen Society, Kunming, Yunnan, 650500, China.
| | - Jianfei Li
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan, 650500, China
| | - Jinglun Xue
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Michael Fenech
- Genome Health Foundation, North Brighton, SA, 5048, Australia; University of South Australia, School of Pharmacy and Medical Sciences, Adelaide, SA, 5000, Australia; Centre of Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Xu Wang
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan, 650500, China; Yunnan Environmental Mutagen Society, Kunming, Yunnan, 650500, China.
| |
Collapse
|
210
|
Mouse Models of Frequently Mutated Genes in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13246192. [PMID: 34944812 PMCID: PMC8699817 DOI: 10.3390/cancers13246192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 01/19/2023] Open
Abstract
Acute myeloid leukemia is a clinically and biologically heterogeneous blood cancer with variable prognosis and response to conventional therapies. Comprehensive sequencing enabled the discovery of recurrent mutations and chromosomal aberrations in AML. Mouse models are essential to study the biological function of these genes and to identify relevant drug targets. This comprehensive review describes the evidence currently available from mouse models for the leukemogenic function of mutations in seven functional gene groups: cell signaling genes, epigenetic modifier genes, nucleophosmin 1 (NPM1), transcription factors, tumor suppressors, spliceosome genes, and cohesin complex genes. Additionally, we provide a synergy map of frequently cooperating mutations in AML development and correlate prognosis of these mutations with leukemogenicity in mouse models to better understand the co-dependence of mutations in AML.
Collapse
|
211
|
Marnell CS, Bick A, Natarajan P. Clonal hematopoiesis of indeterminate potential (CHIP): Linking somatic mutations, hematopoiesis, chronic inflammation and cardiovascular disease. J Mol Cell Cardiol 2021; 161:98-105. [PMID: 34298011 PMCID: PMC8629838 DOI: 10.1016/j.yjmcc.2021.07.004] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is the presence of a clonally expanded hematopoietic stem cell caused by a leukemogenic mutation in individuals without evidence of hematologic malignancy, dysplasia, or cytopenia. CHIP is associated with a 0.5-1.0% risk per year of leukemia. Remarkably, it confers a two-fold increase in cardiovascular risk independent of traditional risk factors. Roughly 80% of patients with CHIP have mutations in epigenetic regulators DNMT3A, TET2, ASXL1, DNA damage repair genes PPM1D, TP53, the regulatory tyrosine kinase JAK2, or mRNA spliceosome components SF3B1, and SRSF2. CHIP is associated with a pro-inflammatory state that has been linked to coronary artery disease, myocardial infarction, and venous thromboembolic disease, as well as prognosis among those with aortic stenosis and heart failure. Heritable and acquired risk factors are associated with increased CHIP prevalence, including germline variation, age, unhealthy lifestyle behaviors (i.e. smoking, obesity), inflammatory conditions, premature menopause, HIV and exposure to cancer therapies. This review aims to summarize emerging research on CHIP, the mechanisms underlying its important role in propagating inflammation and accelerating cardiovascular disease, and new studies detailing the role of associated risk factors and co-morbidities that increase CHIP prevalence.
Collapse
Affiliation(s)
- Christopher S Marnell
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America; Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States of America; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, United States of America
| | - Alexander Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Pradeep Natarajan
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America; Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States of America; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, United States of America.
| |
Collapse
|
212
|
Maillard M, Louveau B, Vilquin P, Goldwirt L, Thomas F, Mourah S. Pharmacogenomics in solid cancers and hematologic malignancies: Improving personalized drug prescription. Therapie 2021; 77:171-183. [PMID: 34922740 DOI: 10.1016/j.therap.2021.11.003] [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: 09/11/2021] [Accepted: 09/29/2021] [Indexed: 11/30/2022]
Abstract
The discovery of molecular alterations involved in oncogenesis is evolving rapidly and has led to the development of new innovative targeted therapies in oncology. High-throughput sequencing techniques help to identify genomic targets and to provide predictive molecular biomarkers of response to guide alternative therapeutic strategies. Besides the emergence of these theranostic markers for the new targeted treatments, pharmacogenetic markers (corresponding to genetic variants existing in the constitutional DNA, i.e., the host genome) can help to optimize the use of chemotherapy. In this review, we present the current clinical applications of constitutional PG and the recent concepts and advances in pharmacogenomics, a rapidly evolving field that focuses on various molecular alterations identified on constitutional or somatic (tumor) genome.
Collapse
Affiliation(s)
- Maud Maillard
- Institut Claudius-Regaud, Institut universitaire du cancer de Toulouse, IUCT-Oncopole, 31059 Toulouse, France; Centre de recherches en cancérologie de Toulouse CRCT, 31037 Toulouse, France; Université Paul-Sabatier Toulouse III, 31062 Toulouse, France
| | - Baptiste Louveau
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - Paul Vilquin
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - Lauriane Goldwirt
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - Fabienne Thomas
- Institut Claudius-Regaud, Institut universitaire du cancer de Toulouse, IUCT-Oncopole, 31059 Toulouse, France; Centre de recherches en cancérologie de Toulouse CRCT, 31037 Toulouse, France; Université Paul-Sabatier Toulouse III, 31062 Toulouse, France
| | - Samia Mourah
- Inserm, UMR_S976, 75475 Paris, France; Université de Paris, 75010 Paris, France; Pharmacogenomics department, Hôpital Saint-Louis, AP-HP, 75010 Paris, France.
| |
Collapse
|
213
|
Kusne Y, Fernandez J, Patnaik MM. Clonal hematopoiesis and VEXAS syndrome: survival of the fittest clones? Semin Hematol 2021; 58:226-229. [PMID: 34802544 DOI: 10.1053/j.seminhematol.2021.10.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/19/2021] [Accepted: 10/01/2021] [Indexed: 01/13/2023]
Abstract
Clonal hematopoiesis (CH) is defined by the acquisition of somatic mutations in hematopoietic stem cells (HSC) leading to enhanced cellular fitness and proliferation under positive clonal selection pressures. CH most frequently involves epigenetic regulator genes (DNMT3A, TET2 and ASXL1), with these mutations being associated with enhanced inflammation and increased all-cause mortality largely from cardiovascular disease and endothelial dysfunction. These mutations also increase the risk for hematological neoplasms. Somatic mutations in UBA1, encoding the E1 ubiquitin ligase in HSC, cause a severe adult-onset autoinflammatory disease that can be associated with myeloid and plasma cell neoplasms, termed VEXAS (vacuoles, X-linked, autoinflammatory, somatic) syndrome. Given the degree of inflammation seen, one would have expected this to be a fertile ground for CH development and propagation, however, preliminary data doesn't support this. Here in, we review the current data on CH, inflammation and VEXAS syndrome.
Collapse
Affiliation(s)
- Yael Kusne
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ
| | - Jenna Fernandez
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Mrinal M Patnaik
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
214
|
Leukemia Stem Cells as a Potential Target to Achieve Therapy-Free Remission in Chronic Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13225822. [PMID: 34830976 PMCID: PMC8616035 DOI: 10.3390/cancers13225822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Leukemia stem cells (LSCs, also known as leukemia-initiating cells) not only drive leukemia initiation and progression, but also contribute to drug resistance and/or disease relapse. Therefore, eradication of every last LSC is critical for a patient's long-term cure. Chronic myeloid leukemia (CML) is a myeloproliferative disorder that arises from multipotent hematopoietic stem and progenitor cells. Tyrosine kinase inhibitors (TKIs) have dramatically improved long-term outcomes and quality of life for patients with CML in the chronic phase. Point mutations of the kinase domain of BCR-ABL1 lead to TKI resistance through a reduction in drug binding, and as a result, several new generations of TKIs have been introduced to the clinic. Some patients develop TKI resistance without known mutations, however, and the presence of LSCs is believed to be at least partially associated with resistance development and CML relapse. We previously proposed targeting quiescent LSCs as a therapeutic approach to CML, and a number of potential strategies for targeting insensitive LSCs have been presented over the last decade. The identification of specific markers distinguishing CML-LSCs from healthy HSCs, and the potential contributions of the bone marrow microenvironment to CML pathogenesis, have also been explored. Nonetheless, 25% of CML patients are still expected to switch TKIs at least once, and various TKI discontinuation studies have shown a wide range in the incidence of molecular relapse (from 30% to 60%). In this review, we revisit the current knowledge regarding the role(s) of LSCs in CML leukemogenesis and response to pharmacological treatment and explore how durable treatment-free remission may be achieved and maintained after discontinuing TKI treatment.
Collapse
|
215
|
Alarcón-Sánchez BR, Pérez-Carreón JI, Villa-Treviño S, Arellanes-Robledo J. Molecular alterations that precede the establishment of the hallmarks of cancer: An approach on the prevention of hepatocarcinogenesis. Biochem Pharmacol 2021; 194:114818. [PMID: 34757033 DOI: 10.1016/j.bcp.2021.114818] [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: 09/22/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023]
Abstract
Chronic liver injury promotes the molecular alterations that precede the establishment of cancer. Usually, several decades of chronic insults are needed to develop the most common primary liver tumor known as hepatocellular carcinoma. As other cancer types, liver cancer cells are governed by a common set of rules collectively called the hallmarks of cancer. Although those rules have provided a conceptual framework for understanding the complex pathophysiology of established tumors, therapeutic options are still ineffective in advanced stages. Thus, the molecular alterations that precede the establishment of cancer remain an attractive target for therapeutic interventions. Here, we first summarize the chemopreventive interventions targeting the early liver carcinogenesis stages. After an integrative analysis on the plethora of molecular alterations regulated by anticancer agents, we then underline and discuss that two critical processes namely oxidative stress and genetic alterations, play the role of 'dirty work laborer' in the initial cell damage and drive the transformation of preneoplastic into neoplastic cells, respectively; besides, the activation of cellular senescence works as a key mechanism in attempting to prevent the onset and establishment of liver cancer. Whereas the detrimental effects of the binomial made up of oxidative stress and genetic alterations are either eliminated or reduced, senescence activation is promoted by anticancer agents. We argue that collectively, oxidative stress, genetic alterations, and senescence are key events that influence the fate of initiated cells and the establishment of the hallmarks of cancer.
Collapse
Affiliation(s)
- Brisa Rodope Alarcón-Sánchez
- Laboratory of Liver Diseases, National Institute of Genomic Medicine - INMEGEN, CDMX, Mexico; Departament of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute - CINVESTAV-IPN, CDMX, Mexico
| | | | - Saúl Villa-Treviño
- Departament of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute - CINVESTAV-IPN, CDMX, Mexico
| | - Jaime Arellanes-Robledo
- Laboratory of Liver Diseases, National Institute of Genomic Medicine - INMEGEN, CDMX, Mexico; Directorate of Cátedras, National Council of Science and Technology - CONACYT, CDMX, Mexico.
| |
Collapse
|
216
|
Awada H, Durmaz A, Gurnari C, Kishtagari A, Meggendorfer M, Kerr CM, Kuzmanovic T, Durrani J, Shreve J, Nagata Y, Radivoyevitch T, Advani AS, Ravandi F, Carraway HE, Nazha A, Haferlach C, Saunthararajah Y, Scott J, Visconte V, Kantarjian H, Kadia T, Sekeres MA, Haferlach T, Maciejewski JP. Machine learning integrates genomic signatures for subclassification beyond primary and secondary acute myeloid leukemia. Blood 2021; 138:1885-1895. [PMID: 34075412 PMCID: PMC8767789 DOI: 10.1182/blood.2020010603] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/04/2021] [Indexed: 11/20/2022] Open
Abstract
Although genomic alterations drive the pathogenesis of acute myeloid leukemia (AML), traditional classifications are largely based on morphology, and prototypic genetic founder lesions define only a small proportion of AML patients. The historical subdivision of primary/de novo AML and secondary AML has shown to variably correlate with genetic patterns. The combinatorial complexity and heterogeneity of AML genomic architecture may have thus far precluded genomic-based subclassification to identify distinct molecularly defined subtypes more reflective of shared pathogenesis. We integrated cytogenetic and gene sequencing data from a multicenter cohort of 6788 AML patients that were analyzed using standard and machine learning methods to generate a novel AML molecular subclassification with biologic correlates corresponding to underlying pathogenesis. Standard supervised analyses resulted in modest cross-validation accuracy when attempting to use molecular patterns to predict traditional pathomorphologic AML classifications. We performed unsupervised analysis by applying the Bayesian latent class method that identified 4 unique genomic clusters of distinct prognoses. Invariant genomic features driving each cluster were extracted and resulted in 97% cross-validation accuracy when used for genomic subclassification. Subclasses of AML defined by molecular signatures overlapped current pathomorphologic and clinically defined AML subtypes. We internally and externally validated our results and share an open-access molecular classification scheme for AML patients. Although the heterogeneity inherent in the genomic changes across nearly 7000 AML patients was too vast for traditional prediction methods, machine learning methods allowed for the definition of novel genomic AML subclasses, indicating that traditional pathomorphologic definitions may be less reflective of overlapping pathogenesis.
Collapse
MESH Headings
- Bayes Theorem
- Cytogenetics
- Gene Expression Regulation, Leukemic
- Genomics
- Humans
- Leukemia, Myeloid, Acute/classification
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Machine Learning
- Mutation
- Neoplasms, Second Primary/classification
- Neoplasms, Second Primary/diagnosis
- Neoplasms, Second Primary/genetics
- Translocation, Genetic
Collapse
Affiliation(s)
- Hassan Awada
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Arda Durmaz
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Ashwin Kishtagari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Cassandra M Kerr
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Teodora Kuzmanovic
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Jibran Durrani
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Jacob Shreve
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Yasunobu Nagata
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Anjali S Advani
- Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Hetty E Carraway
- Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Aziz Nazha
- Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Center for Clinical Artificial Intelligence, Cleveland Clinic, Cleveland, OH
| | | | - Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Jacob Scott
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Mikkael A Sekeres
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
217
|
Richart L, Lapi E, Pancaldi V, Cuenca-Ardura M, Pau ECDS, Madrid-Mencía M, Neyret-Kahn H, Radvanyi F, Rodríguez JA, Cuartero Y, Serra F, Le Dily F, Valencia A, Marti-Renom MA, Real FX. STAG2 loss-of-function affects short-range genomic contacts and modulates the basal-luminal transcriptional program of bladder cancer cells. Nucleic Acids Res 2021; 49:11005-11021. [PMID: 34648034 PMCID: PMC8565347 DOI: 10.1093/nar/gkab864] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 09/08/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022] Open
Abstract
Cohesin exists in two variants containing STAG1 or STAG2. STAG2 is one of the most mutated genes in cancer and a major bladder tumor suppressor. Little is known about how its inactivation contributes to tumorigenesis. Here, we analyze the genomic distribution of STAG1 and STAG2 and perform STAG2 loss-of-function experiments using RT112 bladder cancer cells; we then analyze the genomic effects by integrating gene expression and chromatin interaction data. Functional compartmentalization exists between the cohesin complexes: cohesin-STAG2 displays a distinctive genomic distribution and mediates short and mid-ranged interactions that engage genes at higher frequency than those established by cohesin-STAG1. STAG2 knockdown results in down-regulation of the luminal urothelial signature and up-regulation of the basal transcriptional program, mirroring differences between STAG2-high and STAG2-low human bladder tumors. This is accompanied by rewiring of DNA contacts within topological domains, while compartments and domain boundaries remain refractive. Contacts lost upon depletion of STAG2 are assortative, preferentially occur within silent chromatin domains, and are associated with de-repression of lineage-specifying genes. Our findings indicate that STAG2 participates in the DNA looping that keeps the basal transcriptional program silent and thus sustains the luminal program. This mechanism may contribute to the tumor suppressor function of STAG2 in the urothelium.
Collapse
Affiliation(s)
- Laia Richart
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Eleonora Lapi
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain.,Center for Biomedical Research Network (CIBERONC), 28029 Madrid, Spain
| | - Vera Pancaldi
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain.,Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 Inserm, ERL5294 CNRS, 31037 Toulouse, France.,University Paul Sabatier III, Toulouse, France
| | - Mirabai Cuenca-Ardura
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | | | - Miguel Madrid-Mencía
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain.,Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 Inserm, ERL5294 CNRS, 31037 Toulouse, France.,University Paul Sabatier III, Toulouse, France
| | - Hélène Neyret-Kahn
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France.,Sorbonne Université, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - François Radvanyi
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France.,Sorbonne Université, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Juan Antonio Rodríguez
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Yasmina Cuartero
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - François Serra
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
| | - François Le Dily
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Marc A Marti-Renom
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.,Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08003 Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain.,Center for Biomedical Research Network (CIBERONC), 28029 Madrid, Spain.,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| |
Collapse
|
218
|
Liu Y, Gu Z, Cao H, Kaphle P, Lyu J, Zhang Y, Hu W, Chung SS, Dickerson KE, Xu J. Convergence of oncogenic cooperation at single-cell and single-gene levels drives leukemic transformation. Nat Commun 2021; 12:6323. [PMID: 34732703 PMCID: PMC8566485 DOI: 10.1038/s41467-021-26582-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/13/2021] [Indexed: 12/17/2022] Open
Abstract
Cancers develop from the accumulation of somatic mutations, yet it remains unclear how oncogenic lesions cooperate to drive cancer progression. Using a mouse model harboring NRasG12D and EZH2 mutations that recapitulates leukemic progression, we employ single-cell transcriptomic profiling to map cellular composition and gene expression alterations in healthy or diseased bone marrows during leukemogenesis. At cellular level, NRasG12D induces myeloid lineage-biased differentiation and EZH2-deficiency impairs myeloid cell maturation, whereas they cooperate to promote myeloid neoplasms with dysregulated transcriptional programs. At gene level, NRasG12D and EZH2-deficiency independently and synergistically deregulate gene expression. We integrate results from histopathology, leukemia repopulation, and leukemia-initiating cell assays to validate transcriptome-based cellular profiles. We use this resource to relate developmental hierarchies to leukemia phenotypes, evaluate oncogenic cooperation at single-cell and single-gene levels, and identify GEM as a regulator of leukemia-initiating cells. Our studies establish an integrative approach to deconvolute cancer evolution at single-cell resolution in vivo.
Collapse
Affiliation(s)
- Yuxuan Liu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zhimin Gu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hui Cao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pranita Kaphle
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Junhua Lyu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yuannyu Zhang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Stephen S Chung
- Division of Hematology Oncology, Department of Internal Medicine, and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kathryn E Dickerson
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jian Xu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
219
|
Ganini C, Amelio I, Bertolo R, Bove P, Buonomo OC, Candi E, Cipriani C, Di Daniele N, Juhl H, Mauriello A, Marani C, Marshall J, Melino S, Marchetti P, Montanaro M, Natale ME, Novelli F, Palmieri G, Piacentini M, Rendina EA, Roselli M, Sica G, Tesauro M, Rovella V, Tisone G, Shi Y, Wang Y, Melino G. Global mapping of cancers: The Cancer Genome Atlas and beyond. Mol Oncol 2021; 15:2823-2840. [PMID: 34245122 PMCID: PMC8564642 DOI: 10.1002/1878-0261.13056] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/04/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer genomes have been explored from the early 2000s through massive exome sequencing efforts, leading to the publication of The Cancer Genome Atlas in 2013. Sequencing techniques have been developed alongside this project and have allowed scientists to bypass the limitation of costs for whole-genome sequencing (WGS) of single specimens by developing more accurate and extensive cancer sequencing projects, such as deep sequencing of whole genomes and transcriptomic analysis. The Pan-Cancer Analysis of Whole Genomes recently published WGS data from more than 2600 human cancers together with almost 1200 related transcriptomes. The application of WGS on a large database allowed, for the first time in history, a global analysis of features such as molecular signatures, large structural variations and noncoding regions of the genome, as well as the evaluation of RNA alterations in the absence of underlying DNA mutations. The vast amount of data generated still needs to be thoroughly deciphered, and the advent of machine-learning approaches will be the next step towards the generation of personalized approaches for cancer medicine. The present manuscript wants to give a broad perspective on some of the biological evidence derived from the largest sequencing attempts on human cancers so far, discussing advantages and limitations of this approach and its power in the era of machine learning.
Collapse
Affiliation(s)
- Carlo Ganini
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- IDI‐IRCCSRomeItaly
| | - Ivano Amelio
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Riccardo Bertolo
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Pierluigi Bove
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Oreste Claudio Buonomo
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Eleonora Candi
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- IDI‐IRCCSRomeItaly
| | - Chiara Cipriani
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Nicola Di Daniele
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | | | - Alessandro Mauriello
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Carla Marani
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - John Marshall
- Medstar Georgetown University HospitalGeorgetown UniversityWashingtonDCUSA
| | - Sonia Melino
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | | | - Manuela Montanaro
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Maria Emanuela Natale
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Flavia Novelli
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Giampiero Palmieri
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Mauro Piacentini
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | | | - Mario Roselli
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Giuseppe Sica
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Manfredi Tesauro
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Valentina Rovella
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Giuseppe Tisone
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Yufang Shi
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthShanghai Institutes for Biological SciencesUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
- The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSoochow UniversityChina
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthShanghai Institutes for Biological SciencesUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Gerry Melino
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| |
Collapse
|
220
|
Récher C. The beginning of a new therapeutic era in acute myeloid leukemia. EJHAEM 2021; 2:823-833. [PMID: 35845213 PMCID: PMC9175720 DOI: 10.1002/jha2.252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/17/2022]
Abstract
In the field of AML, the early 2000s were shaped by the advent of novel molecular biology technologies including high-throughput sequencing that improved prognostic classification, response evaluation through the quantification of minimal residual disease, and the launch of research on targeted therapies. Our knowledge of leukemogenesis, AML genetic diversity, gene-gene interactions, clonal evolution, and treatment response assessment has also greatly improved. New classifications based on chromosomal abnormalities and gene mutations are now integrated on a routine basis. These considerable efforts contributed to the discovery and development of promising drugs which specifically target gene mutations, apoptotic pathways and cell surface antigens as well as reformulate classical cytotoxic agents. In less than 2 years, nine novels drugs have been approved for the treatment of AML patients, and many others are being intensively investigated, in particular immune therapies. There are now numerous clinical research opportunities offered to clinicians, thanks to these new treatment options. We are only at the start of a new era which should see major disruptions in the way we understand, treat, and monitor patients with AML.
Collapse
Affiliation(s)
- Christian Récher
- Service d'HématologieCentre Hospitalier Universitaire de ToulouseInstitut Universitaire du Cancer de Toulouse OncopoleUniversité Toulouse III Paul SabatierCentre de Recherches en Cancérologie de ToulouseToulouseFrance
| |
Collapse
|
221
|
Patel AA, Rojek AE, Drazer MW, Weiner H, Godley LA, Le Beau MM, Larson RA. Therapy-related myeloid neoplasms in 109 patients after radiation monotherapy. Blood Adv 2021; 5:4140-4148. [PMID: 34492705 PMCID: PMC8945635 DOI: 10.1182/bloodadvances.2021004964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/09/2021] [Indexed: 11/20/2022] Open
Abstract
Therapy-related myeloid neoplasms (t-MNs) are a late complication of cytotoxic therapy and are defined as a distinct entity by the World Health Organization. Although the link between chemotherapy exposure and risk of subsequent t-MN is well described, the association between radiation monotherapy (RT) and t-MN risk is less definitive. We analyzed 109 consecutive patients who developed t-MNs after RT and describe latencies, cytogenetic profile, mutation analyses, and clinical outcomes. The most common cytogenetic abnormality was a clonal abnormality in chromosome 5 and/or 7, which was present in 45% of patients. The median latency from RT to t-MN diagnosis was 6.5 years, with the shortest latency in patients with balanced translocations. One-year overall survival (OS) was 52% and 5-year OS was 22% for the entire cohort. Patients with chromosome 5 and/or 7 abnormalities experienced worse 1-year OS (37%) and 5-year OS (2%) compared with other cytogenetic groups (P < .0001). Sixteen patients underwent net-generation sequencing; ASXL1 and TET2 were the most commonly mutated genes (n = 4). In addition, 17 patients underwent germline variant testing and 3 carried pathogenic or likely pathogenic germline variants. In conclusion, patients with t-MN after RT monotherapy have increased frequencies of chromosome 5 and/or 7 abnormalities, which are associated with poor OS. In addition, pathogenic germline variants may be common in patients with t-MN after RT monotherapy.
Collapse
Affiliation(s)
| | | | - Michael W Drazer
- Section of Hematology/Oncology, Department of Medicine
- Department of Human Genetics; and
| | - Howard Weiner
- Section of Hematology/Oncology, Department of Medicine
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine
- Department of Human Genetics; and
- Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Michelle M Le Beau
- Section of Hematology/Oncology, Department of Medicine
- Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Richard A Larson
- Section of Hematology/Oncology, Department of Medicine
- Comprehensive Cancer Center, University of Chicago, Chicago, IL
| |
Collapse
|
222
|
Rosendahl Huber A, Van Hoeck A, Van Boxtel R. The Mutagenic Impact of Environmental Exposures in Human Cells and Cancer: Imprints Through Time. Front Genet 2021; 12:760039. [PMID: 34745228 PMCID: PMC8565797 DOI: 10.3389/fgene.2021.760039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
During life, the DNA of our cells is continuously exposed to external damaging processes. Despite the activity of various repair mechanisms, DNA damage eventually results in the accumulation of mutations in the genomes of our cells. Oncogenic mutations are at the root of carcinogenesis, and carcinogenic agents are often highly mutagenic. Over the past decade, whole genome sequencing data of healthy and tumor tissues have revealed how cells in our body gradually accumulate mutations because of exposure to various mutagenic processes. Dissection of mutation profiles based on the type and context specificities of the altered bases has revealed a variety of signatures that reflect past exposure to environmental mutagens, ranging from chemotherapeutic drugs to genotoxic gut bacteria. In this review, we discuss the latest knowledge on somatic mutation accumulation in human cells, and how environmental mutagenic factors further shape the mutation landscapes of tissues. In addition, not all carcinogenic agents induce mutations, which may point to alternative tumor-promoting mechanisms, such as altered clonal selection dynamics. In short, we provide an overview of how environmental factors induce mutations in the DNA of our healthy cells and how this contributes to carcinogenesis. A better understanding of how environmental mutagens shape the genomes of our cells can help to identify potential preventable causes of cancer.
Collapse
Affiliation(s)
- Axel Rosendahl Huber
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Arne Van Hoeck
- Oncode Institute, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Ruben Van Boxtel
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
223
|
Krasnov GS, Ghukasyan LG, Abramov IS, Nasedkina TV. Determination of the Subclonal Tumor Structure in Childhood Acute Myeloid Leukemia and Acral Melanoma by Next-Generation Sequencing. Mol Biol 2021. [DOI: 10.1134/s0026893321040051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
224
|
Singh VK, Thakral D, Gupta R. Regulatory noncoding RNAs: potential biomarkers and therapeutic targets in acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:504-519. [PMID: 34824883 PMCID: PMC8610797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
The noncoding RNAs (ncRNA) comprise a substantial segment of the human transcriptome and have emerged as key elements of cellular homeostasis and disease pathogenesis. Dysregulation of these ncRNAs by alterations in the primary RNA motifs and/or aberrant expression levels is relevant in various diseases, especially cancer. The recent research advances indicate that ncRNAs regulate vital oncogenic processes, including hematopoietic cell differentiation, proliferation, apoptosis, migration, and angiogenesis. The ever-expanding role of ncRNAs in cancer progression and metastasis has sparked interest as potential diagnostic and prognostic biomarkers in acute myeloid leukemia. Moreover, advances in antisense oligonucleotide technologies and pharmacologic discoveries of small molecule inhibitors in targeting RNA structures and RNA-protein complexes have opened newer avenues that may help develop the next generation anti-cancer therapeutics. In this review, we have discussed the role of ncRNA in acute myeloid leukemia and their utility as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Vivek Kumar Singh
- Laboratory Oncology, Dr B.R.A, IRCH, All India Institute of Medical Sciences New Delhi 110029, India
| | - Deepshi Thakral
- Laboratory Oncology, Dr B.R.A, IRCH, All India Institute of Medical Sciences New Delhi 110029, India
| | - Ritu Gupta
- Laboratory Oncology, Dr B.R.A, IRCH, All India Institute of Medical Sciences New Delhi 110029, India
| |
Collapse
|
225
|
Goel H, Rahul E, Gupta I, Chopra A, Ranjan A, Gupta AK, Meena JP, Viswanathan GK, Bakhshi S, Misra A, Hussain S, Kumar R, Singh A, Rath GK, Sharma A, Mittan S, Tanwar P. Molecular and genomic landscapes in secondary & therapy related acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:472-497. [PMID: 34824881 PMCID: PMC8610791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/04/2021] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a complex, aggressive myeloid neoplasm characterized by frequent somatic mutations that influence different functional categories' genes, resulting in maturational arrest and clonal expansion. AML can arise de novo (dn-AML) or can be secondary AML (s-AML) refers to a leukemic process which may arise from an antecedent hematologic disorder (AHD-AML), mostly from a myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN) or can be the result of an antecedent cytotoxic chemotherapy or radiation therapy (therapy-related AML, t-AML). Clinical and biological features in secondary and therapy-related AML are distinct from de novo AML. Secondary and therapy-related AML occurs mainly in the elderly population and responds worse to therapy with higher relapse rates due to resistance to cytotoxic chemotherapy. Over the last decade, advances in molecular genetics have disclosed the sub-clonal architecture of secondary and therapy-related AML. Recent investigations have revealed that cytogenetic abnormalities and underlying genetic aberrations (mutations) are likely to be significant factors dictating prognosis and critical impacts on treatment outcome. Secondary and therapy-related AML have a poorer outcome with adverse cytogenetic abnormalities and higher recurrences of unfavorable mutations compared to de novo AML. In this review, we present an overview of the clinical features of secondary and therapy-related AML and address the function of genetic mutations implicated in the pathogenesis of secondary leukemia. Detailed knowledge of the pathogenetic mechanisms gives an overview of new prognostic markers, including targetable mutations that will presumably lead to the designing and developing novel molecular targeted therapies for secondary and therapy-related AML. Despite significant advances in knowing the genetic aspect of secondary and therapy-related AML, its influence on the disease's pathophysiology, standard treatment prospects have not significantly evolved during the past three decades. Thus, we conclude this review by summarizing the modern and developing treatment strategies in secondary and therapy-related acute myeloid leukemia.
Collapse
Affiliation(s)
- Harsh Goel
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ekta Rahul
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ishan Gupta
- All India Institute of Medical SciencesNew Delhi 110029, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Amar Ranjan
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Ganesh Kumar Viswanathan
- Department of Hematology, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Aroonima Misra
- National Institute of Pathology, ICMRNew Delhi 110029, India
| | - Showket Hussain
- Division Of Molecular Oncology, National Institute of Cancer Prevention & Research I-7, Sector-39Noida 201301, India
| | - Ritesh Kumar
- Department of Radiation Oncology, Rudgers Cancer Institute of New JerseyNJ 07103, United States
| | - Archana Singh
- Department of Pathology, College of Medical Sciences, Rajasthan University of Health SciencesJaipur 302033, India
| | - GK Rath
- Department of Radiotherapy, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Sandeep Mittan
- Department of Cardiology, Ichan School of Medicine, Mount Sinai Hospital1468 Madison Avenue, New York 10028, United States
| | - Pranay Tanwar
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| |
Collapse
|
226
|
Zhang T, Bao X, Qiu H, Tang X, Han Y, Fu C, Sun A, Ruan C, Wu D, Chen S, Xu Y. Development of a Nomogram for Predicting the Cumulative Incidence of Disease Recurrence of AML After Allo-HSCT. Front Oncol 2021; 11:732088. [PMID: 34646774 PMCID: PMC8503644 DOI: 10.3389/fonc.2021.732088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/25/2021] [Indexed: 11/23/2022] Open
Abstract
Using targeted exome sequencing, we studied correlations between mutations at diagnosis and transplant outcomes in 332 subjects with acute myeloid leukemia (AML) receiving allotransplantation. A total of 299 patients (299/332, 90.1%) had at least one oncogenic point mutation. In multivariable analyses, pretransplant disease status, minimal residual disease (MRD) before transplantation (pre-MRD), cytogenetic risk classification, and TP53 and FLT3-ITDhigh ratio mutations were independent risk factors for AML recurrence after allotransplantation (p < 0.05). A nomogram for the cumulative incidence of relapse (CIR) that integrated all the predictors in the multivariable model was then constructed, and the concordance index (C-index) values at 6, 12, 18, and 24 months for CIR prediction were 0.754, 0.730, 0.715, and 0.690, respectively. Moreover, calibration plots showed good agreements between the actual observation and the nomogram prediction for the 6, 12, 18, and 24 months posttransplantation CIR in the internal validation. The integrated calibration index (ICI) values were 0.008, 0.055, 0.094, and 0.136 at 6, 12, 18, and 24 months posttransplantation, respectively. With a median cutoff score of 9.73 from the nomogram, all patients could be divided into two groups, and the differences in 2-year CIR, disease-free survival (DFS), and overall survival (OS) between these two groups were significant (p < 0.05). Taken together, the results of our study indicate that gene mutations could help to predict the outcomes of patients with AML receiving allotransplantation.
Collapse
Affiliation(s)
- Tongtong Zhang
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiebing Bao
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huiying Qiu
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China
| | - Xiaowen Tang
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China
| | - Yue Han
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China
| | - Chengcheng Fu
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China
| | - Aining Sun
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China
| | - Changgeng Ruan
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China
| | - Depei Wu
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Centre of Haematology, Soochow University, Suzhou, China
| | - Suning Chen
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Centre of Haematology, Soochow University, Suzhou, China
| | - Yang Xu
- Jiangsu Institute of Haematology, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,National Clinical Research Centre for Haematological Diseases, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Centre of Haematology, Soochow University, Suzhou, China
| |
Collapse
|
227
|
Brandsma AM, Bertrums EJM, van Roosmalen MJ, Hofman DA, Oka R, Verheul M, Manders F, Ubels J, Belderbos ME, van Boxtel R. Mutation signatures of pediatric acute myeloid leukemia and normal blood progenitors associated with differential patient outcomes. Blood Cancer Discov 2021; 2:484-499. [PMID: 34642666 PMCID: PMC7611805 DOI: 10.1158/2643-3230.bcd-21-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A subset of pediatric AML cases harbors more somatic mutations in their genomes compared to normal blood progenitors. This subset displays expression profiles that resemble more committed progenitors and associates with better patient survival. Acquisition of oncogenic mutations with age is believed to be rate limiting for carcinogenesis. However, the incidence of leukemia in children is higher than in young adults. Here we compare somatic mutations across pediatric acute myeloid leukemia (pAML) patient-matched leukemic blasts and hematopoietic stem and progenitor cells (HSPC), as well as HSPCs from age-matched healthy donors. HSPCs in the leukemic bone marrow have limited genetic relatedness and share few somatic mutations with the cell of origin of the malignant blasts, suggesting polyclonal hematopoiesis in patients with pAML. Compared with normal HSPCs, a subset of pAML cases harbored more somatic mutations and a distinct composition of mutational process signatures. We hypothesize that these cases might have arisen from a more committed progenitor. This subset had better outcomes than pAML cases with mutation burden comparable with age-matched healthy HSPCs. Our study provides insights into the etiology and patient stratification of pAML.
Collapse
Affiliation(s)
- Arianne M Brandsma
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Eline J M Bertrums
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Markus J van Roosmalen
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Damon A Hofman
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Rurika Oka
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Mark Verheul
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Freek Manders
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Joske Ubels
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Mirjam E Belderbos
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| |
Collapse
|
228
|
Zhang HX, Pang AM, Chen X, Zhang RL, Zhai WH, Ma QL, Yang DL, Wei JL, He Y, Feng SZ, Han MZ, Jiang EL. [Allo-HSCT for acute myeloid leukemia with myelodysplastic-related changes: a clinical analysis]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:814-822. [PMID: 34788920 PMCID: PMC8607026 DOI: 10.3760/cma.j.issn.0253-2727.2021.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Indexed: 11/18/2022]
Abstract
Objective: To evaluate the outcomes and prognostic factors of adults with acute myeloid leukemia with myelodysplastic-related changes (AML-MRC) who received allogeneic hematopoietic stem cell transplantation (allo-HSCT) . The genetic mutation lineage of patients with AML-MRC and the molecular mutation affecting the transplantation prognosis was discussed. Methods: The clinical data of 75 patients with AML-MRC who underwent allo-HACT from 2006 to 2020 were retrospectively analyzed for clinical characteristics, survival, relapse-related indicators, and risk factors affecting transplantation prognosis. Additionally, the clinical characteristics and prognosis of multilineage dysplasia (M) group, history of myelodysplastic syndrome (MDS) or myelodysplastic syndrome/myelodysplastic proliferative tumor (MDS/MPN) (H) group, and MDS related cytogenetic abnormalities (C) group were compared. The bone marrow of 43 patients underwent targeting second-generation sequencing (137 genes) . Results: ①There were 41 males and 34 females with a median age of 41 (18-56) years, a median follow-up time of 35 (95%CI 30-49) months, and a median survival time (OS) of 78 (95%CI 23-) months. Three-year OS and event-free survival (EFS) were 57.1% (95%CI 45.6%-71.4%) and 52.0% (95%CI 40.8%-66.1%) . Also, the three-year cumulative recurrence rate (CIR) and transplant-related mortality rate (TRM) were 26.8% (95%CI 16.6%-30.0%) and 22.7% (95%CI 13.2%-33.8%) , respectively. Furthermore, multivariate analysis revealed that pre-transplant non-CR1 status was an independent risk factor for OS and EFS. Other independent risk factors for OS included abnormal karyotype of -5/5q- chromosome and the absence of chronic graft-versus-host disease (cGVHD) after transplantation. ②Among the 75 patients, 59 (78.7%) were in group H, 20 had received demethylation drugs before turning to AML and nine cases (12.0%) in group C and seven cases (9.3%) in group M. There was no significant difference in the three-year OS and EFS among the three groups[group M vs H vs C: OS: 71.4% (95%CI 44.7%-100.0%) vs 55.0% (95%CI 41.8%-72.5%) vs 55.6% (95%CI 31.0%-99.7%) , P=0.700; EFS: 71.4% (95%CI 44.7%-100.0%) vs 46.5% (95%CI 34.0%-63.8%) vs 55.6% (95%CI 31.0%-99.7%) , P=0.600]. Compared with primary and secondary AML-MRC, there was no statistically significant difference in the three-year OS and EFS[61.9% (95%CI 41.9%-91.4%) vs 55.0% (95%CI 41.8%-72.5%) , P=0.600; 61.9% (95%CI 41.9%-91.4%) vs 46.5% (95%CI 34.0%-63.8%) , P=0.400]. Furthermore, there was no significant difference in the time to AML between patients who received demethylation treatment before (20 cases) and those who did not (39 cases) [195 (16-937) d vs 162 (9-3167) d, P=0.804]. Moreover, there were no statistically significant differences in the three-year OS and EFS between the two groups (P=0.400, P=0.700) . ③ NGS test was performed on bone marrow samples of 43 patients (57.3%) , and 73 mutation types were found. Additionally, U2AF1 had the highest mutation incidence (11 cases, 25.6%) , and more than 10% were found: RUNX1 (ten cases, 23.3%) , NRAS (ten cases, 23.3%) , ASXL1 (six cases, 14.0%) , PTPN11 (five cases, 11.6%) , TET2 (five cases, 11.6%) . Univariate analysis showed U2AF1[P=0.875, HR=1.110 (95%CI 0.295-4.195) ], RUNX1[P=0.685, HR=0.728 (95%CI 0.157-3.375) ], NRAS[P=0.919, HR=0.923 (95%CI 0.196-4.334) ] mutation did not affect OS. Conclusion: Chromosome abnormality of -5/5q-, cGVHD, and non-CR1 status before transplantation were independent risk factors for OS in patients with allo-HSCT and AML-MRC. Additionally, the MHC subgroup classification was not a factor affecting the prognosis of transplantation. Treatment with demethylated drugs may not delay MDS turning to AML and prolong the OS after transplantation.
Collapse
Affiliation(s)
- H X Zhang
- 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
| | - A M Pang
- 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
| | - X Chen
- 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
| | - R L Zhang
- 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
| | - W H Zhai
- 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
| | - Q L Ma
- 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
| | - D L Yang
- 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
| | - J L Wei
- 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
| | - Y He
- 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
| | - S Z Feng
- 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
| | - M Z Han
- 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
| | - E L Jiang
- 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
|
229
|
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
|
230
|
Aleem A, Haque AR, Roloff GW, Griffiths EA. Application of Next-Generation Sequencing-Based Mutational Profiling in Acute Lymphoblastic Leukemia. Curr Hematol Malig Rep 2021; 16:394-404. [PMID: 34613552 DOI: 10.1007/s11899-021-00641-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Recent efforts to characterize hematologic cancers with genetic and molecular detail have largely relied on mutational profiling via next-generation sequencing (NGS). The application of NGS-guided disease prognostication and clinical decision making requires a basic understanding of sequencing advantages, pitfalls, and areas where clinical care might be enhanced by the knowledge generated. This article identifies avenues within the landscape of adult acute lymphoblastic leukemia (ALL) where mutational data hold the opportunity to enhance understanding of disease biology and patient care. RECENT FINDINGS NGS-based assessment of measurable residual disease (MRD) after ALL treatment allows for a sensitive and specific molecular survey that is at least comparable, if not superior, to existing techniques. Mutational assessment by NGS has unraveled complex signaling networks that drive pathogenesis of T-cell ALL. Sequencing of patients with familial clustering of ALL has also identified novel germline mutations whose inheritance predisposes to disease development in successive generations. While NGS-based assessment of hematopoietic malignancies often provides actionable information to clinicians, patients with acute lymphoblastic leukemia are left underserved due to a lack of disease classification and prognostication schema that integrate molecular data. Ongoing research is positioned to enrich the molecular toolbox available to clinicians caring for adult ALL patients and deliver new insights to guide therapeutic selection, monitor clinical response, and detect relapse.
Collapse
Affiliation(s)
- Ahmed Aleem
- Department of Medicine, Loyola University Medical Center, 2160 S. 1st Ave, Maywood, IL, 60153, USA
| | - Ali R Haque
- Department of Medicine, Loyola University Medical Center, 2160 S. 1st Ave, Maywood, IL, 60153, USA
| | - Gregory W Roloff
- Department of Medicine, Loyola University Medical Center, 2160 S. 1st Ave, Maywood, IL, 60153, USA.
| | - Elizabeth A Griffiths
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
231
|
Lit BMW, Guo BB, Malherbe JAJ, Kwong YL, Erber WN. Mutation profile of acute myeloid leukaemia in a Chinese cohort by targeted next-generation sequencing. Cancer Rep (Hoboken) 2021; 5:e1573. [PMID: 34617422 PMCID: PMC9575498 DOI: 10.1002/cnr2.1573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 11/07/2022] Open
Abstract
Background Acute myeloid leukaemia (AML) results from the clonal expansion of blast cells of myeloid origin driven by genomic defects. The advances in next‐generation sequencing (NGS) have allowed the identification of many mutated genes important in the pathogenesis of AML. Aims In this study, we aimed to assess the mutation types and frequency in a Chinese cohort presenting with de novo AML cohort using a targeted NGS strategy. Methods In total, we studied samples from 87 adult patients with de novo AML who had no prior history of cytotoxic chemotherapy. Samples were evaluated using a 120‐gene targeted NGS panel to assess the mutation profile. Results Of the 87 AML patients, there were 60 (69%) with a normal karyotype. 89.7% of patients had variants, with an average of 1.9 mutations per patient (range: 0–5 mutations per patient). DNMT3A variants were the most common, being detected in 33 patients (37.9%). NPM1 (34.5%), IDH1/2 (24.1%) and FLT3‐ITD (20.7%) mutations was the next most common. Of the patients with DNMT3A mutations, 24.2% also had mutations NPM1 and FLT3‐ITD and 6.1% NPM1, FLT3‐ITD and IDH mutations. Conclusion Both DNMT3A and NPM1 mutations were more common than in other Chinese and Western AML cohorts that have been studied. DNMT3A mutations tended to co‐occur with NPM1 and FLT3‐ITD mutations and were most commonly seen with a normal karyotype.
Collapse
Affiliation(s)
| | - Belinda B Guo
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | | | - Yok Lam Kwong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Wendy N Erber
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,PathWest Laboratory Medicine, Nedlands, WA, Australia
| |
Collapse
|
232
|
TP53 in Acute Myeloid Leukemia: Molecular Aspects and Patterns of Mutation. Int J Mol Sci 2021; 22:ijms221910782. [PMID: 34639121 PMCID: PMC8509740 DOI: 10.3390/ijms221910782] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 01/10/2023] Open
Abstract
Mutation of the tumor suppressor gene, TP53, is associated with abysmal survival outcomes in acute myeloid leukemia (AML). Although it is the most commonly mutated gene in cancer, its occurrence is observed in only 5–10% of de novo AML, and in 30% of therapy related AML (t-AML). TP53 mutation serves as a prognostic marker of poor response to standard-of-care chemotherapy, particularly in t-AML and AML with complex cytogenetics. In light of a poor response to traditional chemotherapy and only a modest improvement in outcome with hypomethylation-based interventions, allogenic stem cell transplant is routinely recommended in these cases, albeit with a response that is often short lived. Despite being frequently mutated across the cancer spectrum, progress and enthusiasm for the development of p53 targeted therapeutic interventions is lacking and to date there is no approved drug that mitigates the effects of TP53 mutation. There is a mounting body of evidence indicating that p53 mutants differ in functionality and form from typical AML cases and subsequently display inconsistent responses to therapy at the cellular level. Understanding this pathobiological activity is imperative to the development of effective therapeutic strategies. This review aims to provide a comprehensive understanding of the effects of TP53 on the hematopoietic system, to describe its varying degree of functionality in tumor suppression, and to illustrate the need for the adoption of personalized therapeutic strategies to target distinct classes of the p53 mutation in AML management.
Collapse
|
233
|
Beltrame MP, Souto EX, Yamamoto M, Furtado FM, da Costa ES, Sandes AF, Pimenta G, Cavalcanti Júnior GB, Santos-Silva MC, Lorand-Metze I, Ikoma-Colturato MRV. Updating recommendations of the Brazilian Group of Flow Cytometry (GBCFLUX) for diagnosis of acute leukemias using four-color flow cytometry panels. Hematol Transfus Cell Ther 2021; 43:499-506. [PMID: 34127423 PMCID: PMC8573049 DOI: 10.1016/j.htct.2021.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/14/2021] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Flow cytometry has become an increasingly important tool in the clinical laboratory for the diagnosis and monitoring of many hematopoietic neoplasms. This method is ideal for immunophenotypic identification of cellular subpopulations in complex samples, such as bone marrow and peripheral blood. In general, 4-color panels appear to be adequate, depending on the assay. In acute leukemias (ALs), it is necessary identify and characterize the population of abnormal cells in order to recognize the compromised lineage and classify leukemia according to the WHO criteria. Although the use of eight- to ten-color immunophenotyping panels is wellestablished, many laboratories do not have access to this technology. OBJECTIVE AND METHOD In 2015, the Brazilian Group of Flow Cytometry (Grupo Brasileiro de Citometria de Fluxo, GBCFLUX) proposed antibody panels designed to allow the precise diagnosis and characterization of AL within available resources. As many Brazilian flow cytometry laboratories use four-color immunophenotyping, the GBCFLUX has updated that document, according to current leukemia knowledge and after a forum of discussion and validation of antibody panels. RESULTS Recommendations for morphological analysis of bone marrow smears and performing screening panel for lineage (s) identification of AL were maintained from the previous publication. The lineage-oriented proposed panels for B and T cell acute lymphoblastic leukemia (ALL) and for acute myeloid leukemia (AML) were constructed for an appropriate leukemia classification. CONCLUSION Three levels of recommendations (i.e., mandatory, recommended, and optional) were established to enable an accurate diagnosis with some flexibility, considering local laboratory resources and patient-specific needs.
Collapse
Affiliation(s)
- Míriam P Beltrame
- Hospital Erasto Gaertner, Laboratório de Citometria de Fluxo, Curitiba, PR, Brazil.
| | - Elizabeth Xisto Souto
- Hospital do Câncer de Barretos, Barretos, SP, Brazil; Hospital Brigadeiro, São Paulo, SP, Brazil
| | - Mihoko Yamamoto
- Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Felipe M Furtado
- Sabin Medicina Diagnóstica, Brasília, DF, Brazil; Hospital da Criança de Brasília José Alencar, Brasilia, DF, Brazil
| | - Elaine Sobral da Costa
- Instituto de Puericultura e Pediatria Margatão Gesteira, Universidade Federal do Rio de Janeiro (IPPMG/UFRJ), Rio de Janeiro, RJ, Brazil
| | - Alex Freire Sandes
- Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil; Grupo Fleury - Divisão de Hematologia e Citometria de Fluxo, São Paulo, SP, Brazil
| | - Glicínia Pimenta
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | | | - Irene Lorand-Metze
- Faculdade de Ciências Médicas, Universidade Estadual de Campinas (FCM Unicamp), Campinas, SP, Brazil
| | | |
Collapse
|
234
|
Duchmann M, Laplane L, Itzykson R. Clonal Architecture and Evolutionary Dynamics in Acute Myeloid Leukemias. Cancers (Basel) 2021; 13:4887. [PMID: 34638371 PMCID: PMC8507870 DOI: 10.3390/cancers13194887] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemias (AML) results from the accumulation of genetic and epigenetic alterations, often in the context of an aging hematopoietic environment. The development of high-throughput sequencing-and more recently, of single-cell technologies-has shed light on the intratumoral diversity of leukemic cells. Taking AML as a model disease, we review the multiple sources of genetic, epigenetic, and functional heterogeneity of leukemic cells and discuss the definition of a leukemic clone extending its definition beyond genetics. After introducing the two dimensions contributing to clonal diversity, namely, richness (number of leukemic clones) and evenness (distribution of clone sizes), we discuss the mechanisms at the origin of clonal emergence (mutation rate, number of generations, and effective size of the leukemic population) and the causes of clonal dynamics. We discuss the possible role of neutral drift, but also of cell-intrinsic and -extrinsic influences on clonal fitness. After reviewing available data on the prognostic role of genetic and epigenetic diversity of leukemic cells on patients' outcome, we discuss how a better understanding of AML as an evolutionary process could lead to the design of novel therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Matthieu Duchmann
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université de Paris, 75010 Paris, France;
- Laboratoire d’Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
| | - Lucie Laplane
- Institut d’Histoire et Philosophie des Sciences et des Techniques UMR 8590, CNRS, Université Paris 1 Panthéon-Sorbonne, 75010 Paris, France;
- Gustave Roussy Cancer Center, UMR1287, 94805 Villejuif, France
| | - Raphael Itzykson
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université de Paris, 75010 Paris, France;
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
| |
Collapse
|
235
|
Mat Yusoff Y, Ahid F, Abu Seman Z, Abdullah J, Kamaluddin NR, Esa E, Zakaria Z. Comprehensive analysis of mutations and clonal evolution patterns in a cohort of patients with cytogenetically normal acute myeloid leukemia. Mol Cytogenet 2021; 14:45. [PMID: 34560908 PMCID: PMC8464159 DOI: 10.1186/s13039-021-00561-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
Background Relapsed acute myeloid leukemia (AML) is associated with the acquisition of additional somatic mutations which are thought to drive phenotypic adaptability, clonal selection and evolution of leukemic clones during treatment. We performed high throughput exome sequencing of matched presentation and relapsed samples from 6 cytogenetically normal AML (CN-AML) patients treated with standard remission induction chemotherapy in order to contribute with the investigation of the mutational landscape of CN-AML and clonal evolution during AML treatment. Result A total of 24 and 32 somatic variants were identified in presentation and relapse samples respectively with an average of 4.0 variants per patient at presentation and 5.3 variants per patient at relapse, with SNVs being more frequent than indels at both disease stages. All patients have somatic variants in at least one gene that is frequently mutated in AML at both disease presentation and relapse, with most of these variants are classic AML and recurrent hotspot mutations including NPM1 p.W288fs, FLT3-ITD, NRAS p.G12D and IDH2 p.R140Q. In addition, we found two distinct clonal evolution patterns of relapse: (1) a leukemic clone at disease presentation acquires additional mutations and evolves into the relapse clone after the chemotherapy; (2) a leukemic clone at disease presentation persists at relapse without the addition of novel somatic mutations. Conclusions The findings of this study suggest that the relapse-initiating clones may pre-exist prior to therapy, which harbor or acquire mutations that confer selective advantage during chemotherapy, resulting in clonal expansion and eventually leading to relapse.
Collapse
Affiliation(s)
- Yuslina Mat Yusoff
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| | - Fadly Ahid
- Centre for Medical Laboratory Technology Studies, Faculty of Health Sciences, Universiti Teknologi MARA, 42300, Puncak Alam, Selangor, Malaysia.
| | - Zahidah Abu Seman
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| | - Julia Abdullah
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| | - Nor Rizan Kamaluddin
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| | - Ezalia Esa
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| | - Zubaidah Zakaria
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| |
Collapse
|
236
|
Rivas MA, Durmaz C, Kloetgen A, Chin CR, Chen Z, Bhinder B, Koren A, Viny AD, Scharer CD, Boss JM, Elemento O, Mason CE, Melnick AM. Cohesin Core Complex Gene Dosage Contributes to Germinal Center Derived Lymphoma Phenotypes and Outcomes. Front Immunol 2021; 12:688493. [PMID: 34621263 PMCID: PMC8490713 DOI: 10.3389/fimmu.2021.688493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/24/2021] [Indexed: 01/10/2023] Open
Abstract
The cohesin complex plays critical roles in genomic stability and gene expression through effects on 3D architecture. Cohesin core subunit genes are mutated across a wide cross-section of cancers, but not in germinal center (GC) derived lymphomas. In spite of this, haploinsufficiency of cohesin ATPase subunit Smc3 was shown to contribute to malignant transformation of GC B-cells in mice. Herein we explored potential mechanisms and clinical relevance of Smc3 deficiency in GC lymphomagenesis. Transcriptional profiling of Smc3 haploinsufficient murine lymphomas revealed downregulation of genes repressed by loss of epigenetic tumor suppressors Tet2 and Kmt2d. Profiling 3D chromosomal interactions in lymphomas revealed impaired enhancer-promoter interactions affecting genes like Tet2, which was aberrantly downregulated in Smc3 deficient lymphomas. Tet2 plays important roles in B-cell exit from the GC reaction, and single cell RNA-seq profiles and phenotypic trajectory analysis in Smc3 mutant mice revealed a specific defect in commitment to the final steps of plasma cell differentiation. Although Smc3 deficiency resulted in structural abnormalities in GC B-cells, there was no increase of somatic mutations or structural variants in Smc3 haploinsufficient lymphomas, suggesting that cohesin deficiency largely induces lymphomas through disruption of enhancer-promoter interactions of terminal differentiation and tumor suppressor genes. Strikingly, the presence of the Smc3 haploinsufficient GC B-cell transcriptional signature in human patients with GC-derived diffuse large B-cell lymphoma (DLBCL) was linked to inferior clinical outcome and low expression of cohesin core subunits. Reciprocally, reduced expression of cohesin subunits was an independent risk factor for worse survival int DLBCL patient cohorts. Collectively, the data suggest that Smc3 functions as a bona fide tumor suppressor for lymphomas through non-genetic mechanisms, and drives disease by disrupting the commitment of GC B-cells to the plasma cell fate.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/immunology
- Cell Cycle Proteins/metabolism
- Cell Differentiation
- Cells, Cultured
- Chondroitin Sulfate Proteoglycans/genetics
- Chondroitin Sulfate Proteoglycans/immunology
- Chondroitin Sulfate Proteoglycans/metabolism
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/immunology
- Chromosomal Proteins, Non-Histone/metabolism
- Coculture Techniques
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Databases, Genetic
- Dioxygenases/genetics
- Dioxygenases/metabolism
- Gene Dosage
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Germinal Center/immunology
- Germinal Center/metabolism
- Haploinsufficiency
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Mice, Knockout
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Phenotype
- Plasma Cells/immunology
- Plasma Cells/metabolism
- Transcription, Genetic
- Mice
Collapse
Affiliation(s)
- Martin A. Rivas
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Ceyda Durmaz
- Graduate Program on Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, New York, NY, United States
| | - Andreas Kloetgen
- Department of Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Cristopher R. Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Al-Saud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
| | - Zhengming Chen
- Division of Biostatistics and Epidemiology, Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, United States
| | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Al-Saud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Amnon Koren
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States
| | - Aaron D. Viny
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University, New York, NY, United States
| | - Christopher D. Scharer
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, United States
| | - Jeremy M. Boss
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, United States
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Al-Saud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Christopher E. Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Al-Saud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, United States
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Ari M. Melnick
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
237
|
Stasik S, Eckardt JN, Kramer M, Röllig C, Krämer A, Scholl S, Hochhaus A, Crysandt M, Brümmendorf TH, Naumann R, Steffen B, Kunzmann V, Einsele H, Schaich M, Burchert A, Neubauer A, Schäfer-Eckart K, Schliemann C, Krause S, Herbst R, Hänel M, Frickhofen N, Noppeney R, Kaiser U, Baldus CD, Kaufmann M, Rácil Z, Platzbecker U, Berdel WE, Mayer J, Serve H, Müller-Tidow C, Ehninger G, Bornhäuser M, Schetelig J, Middeke JM, Thiede C. Impact of PTPN11 mutations on clinical outcome analyzed in 1529 patients with acute myeloid leukemia. Blood Adv 2021; 5:3279-3289. [PMID: 34459887 PMCID: PMC8525221 DOI: 10.1182/bloodadvances.2021004631] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/13/2021] [Indexed: 12/18/2022] Open
Abstract
The tyrosine-protein phosphatase nonreceptor type 11 (PTPN11) is an important regulator of RAS signaling and frequently affected by mutations in patients with acute myeloid leukemia (AML). Despite the relevance for leukemogenesis and as a potential therapeutic target, the prognostic role is controversial. To investigate the prognostic impact of PTPN11 mutations, we analyzed 1529 adult AML patients using next-generation sequencing. PTPN11 mutations were detected in 106 of 1529 (6.93%) patients (median VAF: 24%) in dominant (36%) and subclonal (64%) configuration. Patients with PTPN11 mutations were associated with concomitant mutations in NPM1 (63%), DNMT3A (37%), and NRAS (21%) and had a higher rate of European LeukemiaNet (ELN) favorable cytogenetics (57.8% vs 39.1%; P < .001) and higher white blood cell counts (P = .007) compared with PTPN11 wild-type patients. In a multivariable analysis, PTPN11 mutations were independently associated with poor overall survival (hazard ratio [HR]: 1.75; P < .001), relapse-free survival (HR: 1.52; P = .013), and a lower rate of complete remission (odds ratio: 0.46; P = .008). Importantly, the deleterious effect of PTPN11 mutations was confined predominantly to the ELN favorable-risk group and patients with subclonal PTPN11 mutations (HR: 2.28; P < .001) but not found with dominant PTPN11 mutations (HR: 1.07; P = .775), presumably because of significant differences within the rate and spectrum of associated comutations. In conclusion, our data suggest an overall poor prognostic impact of PTPN11 mutations in AML, which is significantly modified by the underlying cytogenetics and the clonal context in which they occur.
Collapse
Affiliation(s)
- Sebastian Stasik
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Jan-Niklas Eckardt
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Michael Kramer
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Christoph Röllig
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Alwin Krämer
- Medizinische Klinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Sebastian Scholl
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Andreas Hochhaus
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Martina Crysandt
- Klinik für Hämatologie, Onkologie, Hämostasiologie und Stammzelltransplantation , Uniklinik RWTH Aachen, Aachen, Germany
| | - Tim H Brümmendorf
- Klinik für Hämatologie, Onkologie, Hämostasiologie und Stammzelltransplantation , Uniklinik RWTH Aachen, Aachen, Germany
| | - Ralph Naumann
- Medizinische Klinik III, St. Marien-Krankenhaus Siegen, Siegen, Germany
| | - Björn Steffen
- Medizinische Klinik II, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| | - Volker Kunzmann
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Markus Schaich
- Klinik für Hämatologie, Onkologie und Palliativmedizin, Rems-Murr-Klinikum Winnenden, Winnenden, Germany
| | - Andreas Burchert
- Klinik für Hämatologie, Onkologie, Immunologie, Philipps Universität Marburg, Marburg, Germany
| | - Andreas Neubauer
- Klinik für Hämatologie, Onkologie, Immunologie, Philipps Universität Marburg, Marburg, Germany
| | - Kerstin Schäfer-Eckart
- Klinik für Innere Medizin V, Paracelsus Medizinische Privatuniversität, Klinikum Nürnberg Nord, Nürnberg, Germany
| | | | - Stefan Krause
- Medizinische Klinik V, Paracelsus Medizinische Privatuniversität, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Regina Herbst
- Medizinische Klinik III, Klinikum Chemnitz, Chemnitz, Germany
| | - Mathias Hänel
- Medizinische Klinik III, Klinikum Chemnitz, Chemnitz, Germany
| | | | - Richard Noppeney
- Klinik für Hämatologie, Universitätsklinikum Essen, Essen, Germany
| | - Ulrich Kaiser
- Medizinische Klinik II, St. Bernward Krankenhaus, Hildesheim, Germany
| | - Claudia D Baldus
- Hämatologie und Onkologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Kaufmann
- Abteilung für Hämatologie, Onkologie und Palliativmedizin, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Zdenek Rácil
- Masaryk University and University Hospital, Department of Internal Medicine, Hematology and Oncology, Brno, Czech Republic
| | - Uwe Platzbecker
- Medizinische Klinik und Poliklinik I, Hämatologie und Zelltherapie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Wolfgang E Berdel
- Medizinische Klinik A, Universitätsklinikum Münster, Münster, Germany
| | - Jiri Mayer
- Masaryk University and University Hospital, Department of Internal Medicine, Hematology and Oncology, Brno, Czech Republic
| | - Hubert Serve
- Medizinische Klinik II, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| | | | - Gerhard Ehninger
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Martin Bornhäuser
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany; and
| | - Johannes Schetelig
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
- DKMS Clinical Trials Unit, Dresden, Germany
| | - Jan M Middeke
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Christian Thiede
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
238
|
Parinet V, Chapiro E, Bidet A, Gaillard B, Maarek O, Simon L, Lefebvre C, Defasque S, Mozziconacci MJ, Quinquenel A, Decamp M, Lifermann F, Ali-Ammar N, Maillon A, Baron M, Martin M, Struski S, Penther D, Micol JB, Auger N, Bilhou-Nabera C, Martignoles JA, Tondeur S, Nguyen-Khac F, Hirsch P, Roos-Weil D. Myeloid malignancies with translocation t(4;12)(q11-13;p13): molecular landscape, clonal hierarchy and clinical outcomes. J Cell Mol Med 2021; 25:9557-9566. [PMID: 34492730 PMCID: PMC8505829 DOI: 10.1111/jcmm.16895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Translocation t(4;12)(q11‐13;p13) is a recurrent but very rare chromosomal aberration in acute myeloid leukaemia (AML) resulting in the non‐constant expression of a CHIC2/ETV6 fusion transcript. We report clinico‐biological features, molecular characteristics and outcomes of 21 cases of t(4;12) including 19 AML and two myelodysplastic syndromes (MDS). Median age at the time of t(4;12) was 78 years (range, 56–88). Multilineage dysplasia was described in 10 of 19 (53%) AML cases and CD7 and/or CD56 expression in 90%. FISH analyses identified ETV6 and CHIC2 region rearrangements in respectively 18 of 18 and 15 of 17 studied cases. The t(4;12) was the sole cytogenetic abnormality in 48% of cases. The most frequent associated mutated genes were ASXL1 (n = 8/16, 50%), IDH1/2 (n = 7/16, 44%), SRSF2 (n = 5/16, 31%) and RUNX1 (n = 4/16, 25%). Interestingly, concurrent FISH and molecular analyses showed that t(4;12) can be, but not always, a founding oncogenic event. Median OS was 7.8 months for the entire cohort. In the 16 of 21 patients (76%) who received antitumoral treatment, overall response and first complete remission rates were 37% and 31%, respectively. Median progression‐free survival in responders was 13.7 months. Finally, t(4;12) cases harboured many characteristics of AML with myelodysplasia‐related changes (multilineage dysplasia, MDS‐related cytogenetic abnormalities, frequent ASXL1 mutations) and a poor prognosis.
Collapse
Affiliation(s)
- Vincent Parinet
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Elise Chapiro
- Sorbonne Université, Unité de Cytogénétique, Hôpital Pitié-Salpêtrière, APHP, Paris, France.,Centre de Recherche des Cordeliers, Inserm, Université de Paris, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Sorbonne Université, Paris, France
| | - Audrey Bidet
- Laboratoire d'Hématologie Biologique, CHU Bordeaux, Bordeaux, France
| | - Baptiste Gaillard
- Laboratoire d'Hématologie, Hôpital Robert Debré, Reims, France.,Laboratoire de cytogénétique, Centre Hospitalier de Troyes, Troyes, France
| | - Odile Maarek
- Hematology Laboratory, Hôpital Saint-Louis, APHP, University of Paris, Paris, France
| | - Laurence Simon
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Christine Lefebvre
- Laboratoire de Génétique des Hémopathies, CHU Grenoble Alpes, Grenoble, France
| | - Sabine Defasque
- Secteur cytogénétique hématologique, Laboratoire CERBA, Saint-Ouen l'Aumône, France
| | | | - Anne Quinquenel
- CHU de Reims, Hôpital Robert Debré, Reims, France.,Unité de Formation et de recherche (UFR) Médecine, Université Reims Champagne-Ardenne, Reims, France
| | | | | | - Nadia Ali-Ammar
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Agathe Maillon
- Sorbonne Université, Unité de Cytogénétique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Marine Baron
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Mélanie Martin
- Laboratoire de Cytogénétique, CHU Caremeau, Nîmes, France
| | - Stéphanie Struski
- Laboratoire d'hématologie/Plateau Technique Hématologie-Oncologie, IUCT Oncopole, Toulouse, France
| | - Dominique Penther
- Laboratoire de Génétique Oncologique, CLCC Henri Becquerel & INSERM U1245, Rouen, France
| | - Jean-Baptiste Micol
- Hematology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Nathalie Auger
- Laboratoire de Cytogénétique, Institut Gustave Roussy, Villejuif, France
| | - Chrystèle Bilhou-Nabera
- Service d'Hématologie Biologique, Unité de Cytogénétique onco-hématologique, Hôpital Saint-Antoine, APHP, Sorbonne Université, Paris, France.,Département d'hématologie biologique, INSERM, Centre de Recherche Saint-Antoine Sorbonne, Université, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Jean-Alain Martignoles
- Département d'hématologie biologique, INSERM, Centre de Recherche Saint-Antoine Sorbonne, Université, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Sylvie Tondeur
- Laboratoire de Génétique des Hémopathies, CHU Grenoble Alpes, Grenoble, France
| | - Florence Nguyen-Khac
- Sorbonne Université, Unité de Cytogénétique, Hôpital Pitié-Salpêtrière, APHP, Paris, France.,Centre de Recherche des Cordeliers, Inserm, Université de Paris, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Sorbonne Université, Paris, France
| | - Pierre Hirsch
- Département d'hématologie biologique, INSERM, Centre de Recherche Saint-Antoine Sorbonne, Université, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Damien Roos-Weil
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France.,Centre de Recherche des Cordeliers, Inserm, Université de Paris, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Sorbonne Université, Paris, France
| | | |
Collapse
|
239
|
Onecha E, Rapado I, Luz Morales M, Carreño-Tarragona G, Martinez-Sanchez P, Gutierrez X, Sáchez Pina JM, Linares M, Gallardo M, Martinez-López J, Ayala R. Monitoring of clonal evolution of acute myeloid leukemia identifies the leukemia subtype, clinical outcome and potential new drug targets for post-remission strategies or relapse. Haematologica 2021; 106:2325-2333. [PMID: 32732356 PMCID: PMC8409047 DOI: 10.3324/haematol.2020.254623] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Indexed: 12/16/2022] Open
Abstract
In cases of treatment failure in acute myeloid leukemia (AML), the utility of mutational profiling in primary refractoriness and relapse is not established. We undertook a perspective study using next-generation sequencing (NGS) of clinical follow-up samples (n=91) from 23 patients with AML with therapeutic failure to cytarabine plus idarubicin or fludarabine. Cases of primary refractoriness to treatment were associated with a lower number of DNA variants at diagnosis than cases of relapse (median 1.67 and 3.21, respectively, P=0.029). The most frequently affected pathways in patients with primary refractoriness were signaling, transcription and tumor suppression, whereas methylation and splicing pathways were mainly implicated in relapsed patients. New therapeutic targets, either by an approved drug or within clinical trials, were not identified in any of the cases of refractoriness (zero of ten); however, eight potential new targets were found in five relapsed patients (five of 13, P=0.027): one IDH2, three SF3B1, two KRAS, one KIT and one JAK2. Sixty-five percent of all variants detected at diagnosis were not detected at complete response. Specifically, 100% of variants in EZH2, RUNX1, VHL, FLT3, ETV6, U2AF1, PHF6 and SF3B1 disappeared at complete response, indicating their potential use as markers to evaluate minimal residual disease for follow-up of AML. Molecular follow-up using a custom NGS myeloid panel of 32 genes in the post-treatment evaluation of AML can help in the stratification of prognostic risk, the selection of minimal residual disease markers to monitor the response to treatment and guide post-remission strategies targeting AML, and the selection of new drugs for leukemia relapse.
Collapse
Affiliation(s)
- Esther Onecha
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid
| | | | | | | | | | - Xabier Gutierrez
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid
| | | | - María Linares
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid
| | - Miguel Gallardo
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid
| | | | - Rosa Ayala
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid
| |
Collapse
|
240
|
Abstract
Clonal haematopoiesis (CH) is a common, age-related expansion of blood cells with somatic mutations that is associated with an increased risk of haematological malignancies, cardiovascular disease and all-cause mortality. CH may be caused by point mutations in genes associated with myeloid neoplasms, chromosomal copy number changes and loss of heterozygosity events. How inherited and environmental factors shape the incidence of CH is incompletely understood. Even though the several varieties of CH may have distinct phenotypic consequences, recent research points to an underlying genetic architecture that is highly overlapping. Moreover, there are numerous commonalities between the inherited variation associated with CH and that which has been linked to age-associated biomarkers and diseases. In this Review, we synthesize what is currently known about how inherited variation shapes the risk of CH and how this genetic architecture intersects with the biology of diseases that occur with ageing.
Collapse
Affiliation(s)
- Alexander J Silver
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alexander G Bick
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Michael R Savona
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
241
|
Aung MMK, Mills ML, Bittencourt‐Silvestre J, Keeshan K. Insights into the molecular profiles of adult and paediatric acute myeloid leukaemia. Mol Oncol 2021; 15:2253-2272. [PMID: 33421304 PMCID: PMC8410545 DOI: 10.1002/1878-0261.12899] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a clinically and molecularly heterogeneous disease characterised by uncontrolled proliferation, block in differentiation and acquired self-renewal of hematopoietic stem and myeloid progenitor cells. This results in the clonal expansion of myeloid blasts within the bone marrow and peripheral blood. The incidence of AML increases with age, and in childhood, AML accounts for 20% of all leukaemias. Whilst there are many clinical and biological similarities between paediatric and adult AML with continuum across the age range, many characteristics of AML are associated with age of disease onset. These include chromosomal aberrations, gene mutations and differentiation lineage. Following chemotherapy, AML cells that survive and result in disease relapse exist in an altered chemoresistant state. Molecular profiling currently represents a powerful avenue of experimentation to study AML cells from adults and children pre- and postchemotherapy as a means of identifying prognostic biomarkers and targetable molecular vulnerabilities that may be age-specific. This review highlights recent advances in our knowledge of the molecular profiles with a focus on transcriptomes and metabolomes, leukaemia stem cells and chemoresistant cells in adult and paediatric AML and focus on areas that hold promise for future therapies.
Collapse
Affiliation(s)
- Myint Myat Khine Aung
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| | - Megan L. Mills
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| | | | - Karen Keeshan
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| |
Collapse
|
242
|
Alshemmari SH, Pandita R, Ram M, Rajan R, Aouda K, Samuel L. Prognosis and Outcome of Fit Patients with Acute Myeloid Leukemia in Kuwait. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2021; 21:e736-e743. [PMID: 34154982 DOI: 10.1016/j.clml.2021.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Acute myeloid leukemia (AML) data from the Middle East are limited to single-center studies. We report leukemia-free survival (LFS) and overall survival (OS) of young (≤70 years) patients with AML treated in Kuwait. PATIENTS AND METHODS This study investigated prognostic markers among 172 young and fit patients with de novo nonacute promyelocytic leukemia AML treated with intensive induction protocols from a tertiary cancer center. RESULTS The median age was 44 years (interquartile range, 32-51) and 67% of cases were Arab. A greater proportion of males was found in the 2017 European Leukemia Net-defined unfavorable-risk group (20% vs 9%, respectively; P = .02). Most patients (94%) were treated by a standard 7 × 3 regimen; 72.5% of cases achieved complete remission. The 24-month LFS was 44% (95% confidence interval, 30-65), 36% (95% confidence interval, 26-50), and 23% (95% confidence interval, 10-53) for the favorable-, intermediate-, and adverse-risk groups, respectively (P = .018). The 24-month OS was 70% (95% confidence interval, 60-90), 65% (95% confidence interval, 53-79), and 49% (95% confidence interval, 31-78), respectively (P = .05). Multivariable factor analysis identified male gender (hazard ratio [HR], 1.66; P = .029) and older age (HR, 1.02; P = .05) with poor LFS outcome, whereas favorable-risk classification predicated better outcome (HR, 0.49; P = .03). Favorable-risk classification was the only predictor of OS (HR, 0.39; P = .029). CONCLUSION Fit patients with AML in the favorable-risk group treated with intensive chemotherapy fare well, whereas patients in the adverse-risk group have poor survival.
Collapse
Affiliation(s)
- Salem H Alshemmari
- Department of Hematology, Kuwait Cancer Control Center, Kuwait City, Kuwait; Department of Medicine, Faculty of Medicine, Kuwait University, Jabriya, Kuwait.
| | - Ramesh Pandita
- Department of Hematology, Kuwait Cancer Control Center, Kuwait City, Kuwait
| | - Mohan Ram
- Department of Hematology, Kuwait Cancer Control Center, Kuwait City, Kuwait
| | - Reshmi Rajan
- Department of Hematology, Kuwait Cancer Control Center, Kuwait City, Kuwait
| | - Kloud Aouda
- Department of Hematology, Kuwait Cancer Control Center, Kuwait City, Kuwait
| | - Lovely Samuel
- Cytogenetics Laboratory, Kuwait Cancer Center, Kuwait
| |
Collapse
|
243
|
Sadigh S, Kim AS. Molecular Pathology of Myeloid Neoplasms: Molecular Pattern Recognition. Surg Pathol Clin 2021; 14:517-528. [PMID: 34373100 DOI: 10.1016/j.path.2021.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Despite the apparent complexity of the molecular genetic underpinnings of myeloid neoplasms, most myeloid mutational profiles can be understood within a simple framework. Somatic mutations accumulate in hematopoietic stem cells with aging and toxic insults, termed clonal hematopoiesis. These "old stem cells" mutations, predominantly in the epigenetic and RNA spliceosome pathways, act as "founding" driver mutations leading to a clonal myeloid neoplasm when sufficient in number and clone size. Subsequent mutations can create the genetic flavor of the myeloid neoplasm ("backseat" drivers) due to their enrichment in certain entities or act as progression events ("aggressive" drivers) during clonal evolution.
Collapse
Affiliation(s)
- Sam Sadigh
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
244
|
Brunner AM, Newcomb RA, Chen YB, Crabbe AM. Case 26-2021: A 49-Year-Old Man with Relapsed Acute Myeloid Leukemia. N Engl J Med 2021; 385:834-843. [PMID: 34437787 DOI: 10.1056/nejmcpc2103458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Andrew M Brunner
- From the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Massachusetts General Hospital, and the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Harvard Medical School - both in Boston
| | - Richard A Newcomb
- From the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Massachusetts General Hospital, and the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Harvard Medical School - both in Boston
| | - Yi-Bin Chen
- From the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Massachusetts General Hospital, and the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Harvard Medical School - both in Boston
| | - Andrew M Crabbe
- From the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Massachusetts General Hospital, and the Departments of Medicine (A.M.B., R.A.N., Y.-B.C.) and Pathology (A.M.C.), Harvard Medical School - both in Boston
| |
Collapse
|
245
|
López-Cortés A, Abarca E, Silva L, Velastegui E, León-Sosa A, Karolys G, Cabrera F, Caicedo A. Identification of key proteins in the signaling crossroads between wound healing and cancer hallmark phenotypes. Sci Rep 2021; 11:17245. [PMID: 34446793 PMCID: PMC8390472 DOI: 10.1038/s41598-021-96750-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Wound healing (WH) and cancer seem to share common cellular and molecular processes that could work in a tight balance to maintain tissue homeostasis or, when unregulated, drive tumor progression. The "Cancer Hallmarks" comprise crucial biological properties that mediate the advancement of the disease and affect patient prognosis. These hallmarks have been proposed to overlap with essential features of the WH process. However, common hallmarks and proteins actively participating in both processes have yet to be described. In this work we identify 21 WH proteins strongly linked with solid tumors by integrated TCGA Pan-Cancer and multi-omics analyses. These proteins were associated with eight of the ten described cancer hallmarks, especially avoiding immune destruction. These results show that WH and cancer's common proteins are involved in the microenvironment modification of solid tissues and immune system regulation. This set of proteins, between WH and cancer, could represent key targets for developing therapies.
Collapse
Affiliation(s)
- Andrés López-Cortés
- grid.412257.70000 0004 0485 6316Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador ,Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain ,grid.8073.c0000 0001 2176 8535RNASA-IMEDIR, Computer Science Faculty, Universidad of A Coruna, A Coruña, Spain
| | - Estefanía Abarca
- grid.442129.8Carrera de Biotecnología, Universidad Politécnica Salesiana UPS, Quito, Ecuador
| | - Leonardo Silva
- grid.442129.8Carrera de Biotecnología, Universidad Politécnica Salesiana UPS, Quito, Ecuador
| | - Erick Velastegui
- grid.442129.8Carrera de Biotecnología, Universidad Politécnica Salesiana UPS, Quito, Ecuador
| | - Ariana León-Sosa
- grid.412251.10000 0000 9008 4711Instituto de Investigaciones en Biomedicina iBioMed, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Germania Karolys
- grid.442129.8Carrera de Biotecnología, Universidad Politécnica Salesiana UPS, Quito, Ecuador ,grid.442129.8Grupo de Investigación y Desarrollo en Ciencias Aplicadas a los Recursos Biológicos, Universidad Politécnica Salesiana, Quito, Ecuador
| | - Francisco Cabrera
- grid.412251.10000 0000 9008 4711Instituto de Investigaciones en Biomedicina iBioMed, Universidad San Francisco de Quito USFQ, Quito, Ecuador ,grid.412251.10000 0000 9008 4711Colegio de Ciencias de la Salud, Escuela de Medicina Veterinaria, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Andrés Caicedo
- grid.412251.10000 0000 9008 4711Instituto de Investigaciones en Biomedicina iBioMed, Universidad San Francisco de Quito USFQ, Quito, Ecuador ,grid.412251.10000 0000 9008 4711Colegio de Ciencias de la Salud, Escuela de Medicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador ,Mito-Act Research Consortium, Quito, Ecuador ,grid.412251.10000 0000 9008 4711Sistemas Médicos SIME, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| |
Collapse
|
246
|
Abstract
Indirect somatic genetic rescue (SGR) of a germline mutation is thought to be rare in inherited Mendelian disorders. Here, we establish that acquired mutations in the EIF6 gene are a frequent mechanism of SGR in Shwachman-Diamond syndrome (SDS), a leukemia predisposition disorder caused by a germline defect in ribosome assembly. Biallelic mutations in the SBDS or EFL1 genes in SDS impair release of the anti-association factor eIF6 from the 60S ribosomal subunit, a key step in the translational activation of ribosomes. Here, we identify diverse mosaic somatic genetic events (point mutations, interstitial deletion, reciprocal chromosomal translocation) in SDS hematopoietic cells that reduce eIF6 expression or disrupt its interaction with the 60S subunit, thereby conferring a selective advantage over non-modified cells. SDS-related somatic EIF6 missense mutations that reduce eIF6 dosage or eIF6 binding to the 60S subunit suppress the defects in ribosome assembly and protein synthesis across multiple SBDS-deficient species including yeast, Dictyostelium and Drosophila. Our data suggest that SGR is a universal phenomenon that may influence the clinical evolution of diverse Mendelian disorders and support eIF6 suppressor mimics as a therapeutic strategy in SDS.
Collapse
|
247
|
Abbas HA, Reville PK, Geppner A, Rausch CR, Pemmaraju N, Ohanian M, Sasaki K, Borthakur G, Daver N, DiNardo C, Bueso-Ramos C, Pierce S, Jabbour E, Garcia-Manero G, Konopleva M, Ravandi F, Kantarjian H, Kadia TM. Clinical and molecular characterization of myeloid sarcoma without medullary leukemia. Leuk Lymphoma 2021; 62:3402-3410. [PMID: 34380367 DOI: 10.1080/10428194.2021.1961235] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myeloid sarcoma (MS) in the setting of concomitant medullary AML is relatively well described, while much less is known about patients presenting with MS with <20% bone marrow blasts. We conducted a retrospective analysis of 56 patients with MS with <20% marrow blasts seen at MD Anderson between 2005 and 2020. The prevalence of MS without medullary AML was 1.4% among all newly diagnosed AML patients. The majority (75%) of patients had a single known anatomic site involved, with the skin (34%) being the most frequent. The most common histologic subtype was monocytic, and 11% of patients had a known history of an antecedent hematologic disorder. The majority of patients (70%) received frontline intensive chemotherapy induction, with 75% of those evaluable attaining complete or partial responses. The median overall survival (OS) and event-free survival (EFS) were 3.41 and 3.07 years, respectively. Patients with bone marrow blasts of ≥5% or medullary relapse had inferior outcomes, while age (>60 years) was not associated with outcomes. There was a suggestion that patients with isolated leukemia cutis may have had better outcomes compared to patients with other organ involvement, but this did not reach statistical significance. Most patients who had cytogenetic analysis had a diploid karyotype within their MS and bone marrow. RAS pathway mutations were enriched in MS at diagnosis, and at time of medullary relapse. Our study provides a large dataset summarizing the clinical and molecular analysis of patients with MS with <20% BM blasts and suggests that monitoring for medullary leukemia is important for early detection of relapse.
Collapse
Affiliation(s)
- Hussein A Abbas
- Division of Cancer Medicine, Medical Oncology Fellowship, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick K Reville
- Division of Cancer Medicine, Medical Oncology Fellowship, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexis Geppner
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caitlin R Rausch
- Division of Pharmacy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maro Ohanian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney DiNardo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos Bueso-Ramos
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sherry Pierce
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Marina Konopleva
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan M Kadia
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
248
|
Fuster JJ. Clonal Hematopoiesis and Incident Heart Failure Risk: The Clone Wars Reach the Myocardium. J Am Coll Cardiol 2021; 78:53-55. [PMID: 34210414 DOI: 10.1016/j.jacc.2021.04.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 11/18/2022]
Affiliation(s)
- José J Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
249
|
Yamauchi T, Miyawaki K, Semba Y, Takahashi M, Izumi Y, Nogami J, Nakao F, Sugio T, Sasaki K, Pinello L, Bauer DE, Bamba T, Akashi K, Maeda T. Targeting leukemia-specific dependence on the de novo purine synthesis pathway. Leukemia 2021; 36:383-393. [PMID: 34344987 DOI: 10.1038/s41375-021-01369-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/16/2021] [Accepted: 07/22/2021] [Indexed: 01/02/2023]
Abstract
Acute myeloid leukemia (AML) is a devastating disease, and clinical outcomes are still far from satisfactory. Here, to identify novel targets for AML therapy, we performed a genome-wide CRISPR/Cas9 screen using AML cell lines, followed by a second screen in vivo. We show that PAICS, an enzyme involved in de novo purine biosynthesis, is a potential target for AML therapy. AML cells expressing shRNA-PAICS exhibited a proliferative disadvantage, indicating a toxic effect of shRNA-PAICS. Treatment of human AML cells with a PAICS inhibitor suppressed their proliferation by inhibiting DNA synthesis and promoting apoptosis and had anti-leukemic effects in AML PDX models. Furthermore, CRISPR/Cas9 screens using AML cells in the presence of the inhibitor revealed genes mediating resistance or synthetic lethal to PAICS inhibition. Our findings identify PAICS as a novel therapeutic target for AML and further define components of de novo purine synthesis pathway and its downstream effectors essential for AML cell survival.
Collapse
Affiliation(s)
- Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan.,Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Kohta Miyawaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Yuichiro Semba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Jumpei Nogami
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Fumihiko Nakao
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Takeshi Sugio
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan.,Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Kensuke Sasaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Luca Pinello
- Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, 02129, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Takahiro Maeda
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, 812-8582, Japan. .,Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan.
| |
Collapse
|
250
|
Accurate SNV detection in single cells by transposon-based whole-genome amplification of complementary strands. Proc Natl Acad Sci U S A 2021; 118:2013106118. [PMID: 33593904 PMCID: PMC7923680 DOI: 10.1073/pnas.2013106118] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The boom of single-cell sequencing technologies in the past decade has profoundly expanded our understanding of fundamental biology. Today, tens of thousands of cells can be measured by single-cell RNA-seq in one experiment. However, single-cell DNA-sequencing studies have been limited by false positives and cost. Here we report META-CS, a single-cell whole-genome amplification method that takes advantage of the complementary strands of double-stranded DNA to filter out false positives and reduce sequencing cost. META-CS achieved the highest accuracy in terms of detecting single-nucleotide variations, and provided potential solutions for the identification of other genomic variants, such as insertions, deletions, and structural variations in single cells. Single-nucleotide variants (SNVs), pertinent to aging and disease, occur sporadically in the human genome, hence necessitating single-cell measurements. However, detection of single-cell SNVs suffers from false positives (FPs) due to intracellular single-stranded DNA damage and the process of whole-genome amplification (WGA). Here, we report a single-cell WGA method termed multiplexed end-tagging amplification of complementary strands (META-CS), which eliminates nearly all FPs by virtue of DNA complementarity, and achieved the highest accuracy thus far. We validated META-CS by sequencing kindred cells and human sperm, and applied it to other human tissues. Investigation of mature single human neurons revealed increasing SNVs with age and potentially unrepaired strand-specific oxidative guanine damage. We determined SNV frequencies along the genome in differentiated single human blood cells, and identified cell type-dependent mutational patterns for major types of lymphocytes.
Collapse
|