1
|
Chiu CY, Matsuo T, Wurster S, Gerstein Y, Hammond DE, Chien KS, DiNardo C, Kontoyiannis DP. Invasive mucorales sinusitis in a young patient with Emberger syndrome and newly diagnosed AML: A case report and literature review of invasive fungal infections in GATA2 deficiency. Mycoses 2023; 66:1029-1034. [PMID: 37550272 DOI: 10.1111/myc.13638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023]
Abstract
Germline pathogenic variants (PVs) in the gene encoding the GATA2 transcription factor can result in profound reductions of monocytes, dendritic cells, natural killer cells and B cells. GATA2 PVs are associated with an increased risk of myeloid malignancies and a predisposition to nontuberculous mycobacterial and human papillomavirus infections. Additionally, invasive fungal infections (IFIs) have been reported in individuals with GATA2 PVs, even in the absence of myeloid malignancies. In this report, we present the case of a 40-year-old man with Emberger syndrome (GATA2 mutation, recently diagnosed acute myeloid leukaemia [AML] and history of lymphedema with hearing loss) who developed Mucorales sinusitis while receiving his first course of remission induction chemotherapy. Additionally, we review the literature on all published cases of proven IFIs in patients with GATA2 PVs. Clinicians should be aware that patients with GATA2 PVs could be vulnerable to opportunistic IFIs, even in the absence of AML and antineoplastic therapy. Furthermore, the distinctly unusual occurrence of mucormycosis during the first course of induction chemotherapy for AML in our patient indicates that patients with germline GATA2 PVs receiving induction chemotherapy for AML might be at high risk for early onset of IFIs due to aggressive, opportunistic moulds.
Collapse
Affiliation(s)
- Chia-Yu Chiu
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Takahiro Matsuo
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Sebastian Wurster
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Yoheved Gerstein
- Department of Clinical Cancer Genetics, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Danielle E Hammond
- Department of Leukaemia, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Kelly S Chien
- Department of Leukaemia, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Courtney DiNardo
- Department of Leukaemia, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| |
Collapse
|
2
|
West RR, Bauer TR, Tuschong LM, Embree LJ, Calvo KR, Tillo D, Davis J, Holland SM, Hickstein DD. A novel GATA2 distal enhancer mutation results in MonoMAC syndrome in 2 second cousins. Blood Adv 2023; 7:6351-6363. [PMID: 37595058 PMCID: PMC10587712 DOI: 10.1182/bloodadvances.2023010458] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/24/2023] [Accepted: 08/12/2023] [Indexed: 08/20/2023] Open
Abstract
Mutations in the transcription factor GATA2 can cause MonoMAC syndrome, a GATA2 deficiency disease characterized by several findings, including disseminated nontuberculous mycobacterial infections, severe deficiencies of monocytes, natural killer cells, and B lymphocytes, and myelodysplastic syndrome. GATA2 mutations are found in ∼90% of patients with a GATA2 deficiency phenotype and are largely missense mutations in the conserved second zinc-finger domain. Mutations in an intron 5 regulatory enhancer element are also well described in GATA2 deficiency. Here, we present a multigeneration kindred with the clinical features of GATA2 deficiency but lacking an apparent GATA2 mutation. Whole genome sequencing revealed a unique adenine-to-thymine variant in the GATA2 -110 enhancer 116,855 bp upstream of the GATA2 ATG start site. The mutation creates a new E-box consensus in position with an existing GATA-box to generate a new hematopoietic regulatory composite element. The mutation segregates with the disease in several generations of the family. Cell type-specific allelic imbalance of GATA2 expression was observed in the bone marrow of a patient with higher expression from the mutant-linked allele. Allele-specific overexpression of GATA2 was observed in CRISPR/Cas9-modified HL-60 cells and in luciferase assays with the enhancer mutation. This study demonstrates overexpression of GATA2 resulting from a single nucleotide change in an upstream enhancer element in patients with MonoMAC syndrome. Patients in this study were enrolled in the National Institute of Allergy and Infectious Diseases clinical trial and the National Cancer Institute clinical trial (both trials were registered at www.clinicaltrials.gov as #NCT01905826 and #NCT01861106, respectively).
Collapse
Affiliation(s)
- Robert R. West
- Immune Deficiency–Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Thomas R. Bauer
- Immune Deficiency–Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Laura M. Tuschong
- Immune Deficiency–Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa J. Embree
- Immune Deficiency–Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Katherine R. Calvo
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD
| | - Desiree Tillo
- Genomics Core, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Joie Davis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Dennis D. Hickstein
- Immune Deficiency–Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
3
|
Lawrence L, Wang A, Charville G, Liu CL, Garofalo A, Alizadeh A, Jangam D, Pinsky BA, Sahoo M, Gratzinger D, Khodadoust M, Kim Y, Novoa R, Stehr H. Identification and confirmation via in situ hybridization of Merkel cell polyomavirus in rare cases of posttransplant cutaneous T-cell lymphoma. J Cutan Pathol 2023; 50:835-844. [PMID: 37394808 DOI: 10.1111/cup.14486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Viral infection is an oncogenic factor in many hematolymphoid malignancies. We sought to determine the diagnostic yield of aligning off-target reads incidentally obtained during targeted hematolymphoid next-generation sequencing to a large database of viral genomes to screen for viral sequences within tumor specimens. METHODS Alignment of off-target reads to viral genomes was performed using magicBLAST. Localization of Merkel cell polyomavirus (MCPyV) RNA was confirmed by RNAScope in situ hybridization. Integration analysis was performed using Virus-Clip. RESULTS Four cases of post-cardiac-transplant folliculotropic mycosis fungoides (fMF) and one case of peripheral T-cell lymphoma (PTCL) were positive in off-target reads for MCPyV DNA. Two of the four cases of posttransplant fMF and the case of PTCL showed localization of MCPyV RNA to malignant lymphocytes, whereas the remaining two cases of posttransplant fMF showed MCPyV RNA in keratinocytes. CONCLUSIONS Our findings raise the question of whether MCPyV may play a role in rare cases of T-lymphoproliferative disorders, particularly in the skin and in the heavily immunosuppressed posttransplant setting.
Collapse
Affiliation(s)
| | - Aihui Wang
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Chih Long Liu
- Stanford University School of Medicine, Stanford, California, USA
| | - Andrea Garofalo
- Stanford University School of Medicine, Stanford, California, USA
| | - Ash Alizadeh
- Stanford University School of Medicine, Stanford, California, USA
| | | | | | - Malaya Sahoo
- Stanford University School of Medicine, Stanford, California, USA
| | - Dita Gratzinger
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Youn Kim
- Stanford University School of Medicine, Stanford, California, USA
| | - Roberto Novoa
- Stanford University School of Medicine, Stanford, California, USA
| | - Henning Stehr
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
4
|
Santiago M, Liquori A, Such E, Zúñiga Á, Cervera J. The Clinical Spectrum, Diagnosis, and Management of GATA2 Deficiency. Cancers (Basel) 2023; 15:cancers15051590. [PMID: 36900380 PMCID: PMC10000430 DOI: 10.3390/cancers15051590] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Hereditary myeloid malignancy syndromes (HMMSs) are rare but are becoming increasingly significant in clinical practice. One of the most well-known syndromes within this group is GATA2 deficiency. The GATA2 gene encodes a zinc finger transcription factor essential for normal hematopoiesis. Insufficient expression and function of this gene as a result of germinal mutations underlie distinct clinical presentations, including childhood myelodysplastic syndrome and acute myeloid leukemia, in which the acquisition of additional molecular somatic abnormalities can lead to variable outcomes. The only curative treatment for this syndrome is allogeneic hematopoietic stem cell transplantation, which should be performed before irreversible organ damage happens. In this review, we will examine the structural characteristics of the GATA2 gene, its physiological and pathological functions, how GATA2 genetic mutations contribute to myeloid neoplasms, and other potential clinical manifestations. Finally, we will provide an overview of current therapeutic options, including recent transplantation strategies.
Collapse
Affiliation(s)
- Marta Santiago
- Hematology Department, Hospital La Fe, 46026 Valencia, Spain; (M.S.); (E.S.); (J.C.)
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Alessandro Liquori
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence:
| | - Esperanza Such
- Hematology Department, Hospital La Fe, 46026 Valencia, Spain; (M.S.); (E.S.); (J.C.)
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ángel Zúñiga
- Genetics Unit, Hospital La Fe, 46026 Valencia, Spain;
| | - José Cervera
- Hematology Department, Hospital La Fe, 46026 Valencia, Spain; (M.S.); (E.S.); (J.C.)
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Genetics Unit, Hospital La Fe, 46026 Valencia, Spain;
| |
Collapse
|
5
|
Tabaja H, Jensen KL, Rivera CG, Misra A, Pruthi RK, Vergidis P. Multiple Simultaneous Infections with Nontuberculous Mycobacteria in the Setting of GATA2 Mutation and Myelodysplastic Syndrome. Open Forum Infect Dis 2022; 9:ofac309. [PMID: 35891688 PMCID: PMC9308453 DOI: 10.1093/ofid/ofac309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
GATA2 mutation can result in profoundly reduced monocytes, dendritic cells, natural killer cells, and B cells, and is associated with a predisposition for recurrent and disseminated nontuberculous mycobacterial (NTM) infections and myelodysplasias. Herein, we describe a unique case of 3 simultaneous disseminated NTM infections in a patient with GATA2 mutations.
Collapse
Affiliation(s)
- Hussam Tabaja
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic Rochester , MN , USA
| | - Kelsey L Jensen
- Department of Pharmacy Services, Mayo Clinic Health System – Austin , MN , USA
| | | | - Anisha Misra
- Division of Clinical Microbiology, Mayo Clinic , Rochester, MN , USA
| | - Rajiv K Pruthi
- Division of Hematology, Mayo Clinic , Rochester, MN , USA
| | - Paschalis Vergidis
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic Rochester , MN , USA
| |
Collapse
|
6
|
West RR, Calvo KR, Embree LJ, Wang W, Tuschong LM, Bauer TR, Tillo D, Lack J, Droll S, Hsu AP, Holland SM, Hickstein DD. ASXL1 and STAG2 are common mutations in GATA2 deficiency patients with bone marrow disease and myelodysplastic syndrome. Blood Adv 2022; 6:793-807. [PMID: 34529785 PMCID: PMC8945308 DOI: 10.1182/bloodadvances.2021005065] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/29/2021] [Indexed: 11/20/2022] Open
Abstract
Patients with GATA2 deficiencyharbor de novo or inherited germline mutations in the GATA2 transcription factor gene, predisposing them to myeloid malignancies. There is considerable variation in disease progression, even among family members with the same mutation in GATA2. We investigated somatic mutations in 106 patients with GATA2 deficiency to identify acquired mutations that are associated with myeloid malignancies. Myelodysplastic syndrome (MDS) was the most common diagnosis (∼44%), followed by GATA2 bone marrow immunodeficiency disorder (G2BMID; ∼37%). Thirteen percent of the cohort had GATA2 mutations but displayed no disease manifestations. There were no correlations between age or sex with disease progression or survival. Cytogenetic analyses showed a high incidence of abnormalities (∼43%), notably trisomy 8 (∼23%) and monosomy 7 (∼12%), but the changes did not correlate with lower survival. Somatic mutations in ASXL1 and STAG2 were detected in ∼25% of patients, although the mutations were rarely concomitant. Mutations in DNMT3A were found in ∼10% of patients. These somatic mutations were found similarly in G2BMID and MDS, suggesting clonal hematopoiesis in early stages of disease, before the onset of MDS. ASXL1 mutations conferred a lower survival probability and were more prevalent in female patients. STAG2 mutations also conferred a lower survival probability, but did not show a statistically significant sex bias. There was a conspicuous absence of many commonly mutated genes associated with myeloid malignancies, including TET2, IDH1/2, and the splicing factor genes. Notably, somatic mutations in chromatin-related genes and cohesin genes characterized disease progression in GATA2 deficiency.
Collapse
Affiliation(s)
- Robert R. West
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda MD
| | | | - Lisa J. Embree
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda MD
| | - Weixin Wang
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD
| | - Laura M. Tuschong
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda MD
| | - Thomas R. Bauer
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda MD
| | - Desiree Tillo
- Genomics Core, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD
| | - Justin Lack
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD; and
| | - Stephenie Droll
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD
| | - Amy P. Hsu
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD
| | - Steven M. Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD
| | - Dennis D. Hickstein
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda MD
| |
Collapse
|
7
|
Koyunlar C, de Pater E. From Basic Biology to Patient Mutational Spectra of GATA2 Haploinsufficiencies: What Are the Mechanisms, Hurdles, and Prospects of Genome Editing for Treatment. Front Genome Ed 2021; 2:602182. [PMID: 34713225 PMCID: PMC8525360 DOI: 10.3389/fgeed.2020.602182] [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: 09/02/2020] [Accepted: 10/30/2020] [Indexed: 12/23/2022] Open
Abstract
Inherited bone marrow failure syndromes (IBMFS) are monogenetic disorders that result in a reduction of mature blood cell formation and predisposition to leukemia. In children with myeloid leukemia the gene most often mutated is Gata binding protein 2 (GATA2) and 80% of patients with GATA2 mutations develop myeloid malignancy before the age of forty. Although GATA2 is established as one of the key regulators of embryonic and adult hematopoiesis, the mechanisms behind the leukemia predisposition in GATA2 haploinsufficiencies is ambiguous. The only curative treatment option currently available is allogeneic hematopoietic stem cell transplantation (allo-SCT). However, allo-SCT can only be applied at a relatively late stage of the disease as its applicability is compromised by treatment related morbidity and mortality (TRM). Alternatively, autologous hematopoietic stem cell transplantation (auto-SCT), which is associated with significantly less TRM, might become a treatment option if repaired hematopoietic stem cells would be available. Here we discuss the recent literature on leukemia predisposition syndromes caused by GATA2 mutations, current knowledge on the function of GATA2 in the hematopoietic system and advantages and pitfalls of potential treatment options provided by genome editing.
Collapse
Affiliation(s)
- Cansu Koyunlar
- Department of Hematology, Erasmus MC, Rotterdam, Netherlands
| | - Emma de Pater
- Department of Hematology, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
8
|
Abbas HA, Mohanty V, Wang R, Huang Y, Liang S, Wang F, Zhang J, Qiu Y, Hu CW, Qutub AA, Dail M, Bolen CR, Daver N, Konopleva M, Futreal A, Chen K, Wang L, Kornblau SM. Decoupling Lineage-Associated Genes in Acute Myeloid Leukemia Reveals Inflammatory and Metabolic Signatures Associated With Outcomes. Front Oncol 2021; 11:705627. [PMID: 34422660 PMCID: PMC8372368 DOI: 10.3389/fonc.2021.705627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/07/2021] [Indexed: 12/28/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with variable responses to therapy. Cytogenetic and genomic features are used to classify AML patients into prognostic and treatment groups. However, these molecular characteristics harbor significant patient-to-patient variability and do not fully account for AML heterogeneity. RNA-based classifications have also been applied in AML as an alternative approach, but transcriptomic grouping is strongly associated with AML morphologic lineages. We used a training cohort of newly diagnosed AML patients and conducted unsupervised RNA-based classification after excluding lineage-associated genes. We identified three AML patient groups that have distinct biological pathways associated with outcomes. Enrichment of inflammatory pathways and downregulation of HOX pathways were associated with improved outcomes, and this was validated in 2 independent cohorts. We also identified a group of AML patients who harbored high metabolic and mTOR pathway activity, and this was associated with worse clinical outcomes. Using a comprehensive reverse phase protein array, we identified higher mTOR protein expression in the highly metabolic group. We also identified a positive correlation between degree of resistance to venetoclax and mTOR activation in myeloid and lymphoid cell lines. Our approach of integrating RNA, protein, and genomic data uncovered lineage-independent AML patient groups that share biologic mechanisms and can inform outcomes independent of commonly used clinical and demographic variables; these groups could be used to guide therapeutic strategies.
Collapse
Affiliation(s)
- Hussein A Abbas
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ruiping Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yuefan Huang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Biostatistics & Data Science, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shaoheng Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Computer Science, Rice University, Houston, TX, United States
| | - Feng Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yihua Qiu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Chenyue W Hu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amina A Qutub
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Monique Dail
- Oncology Biomarker Development, Genentech Inc, South San Francisco, CA, United States
| | - Christopher R Bolen
- Oncology Bioinformatics, Genentech Inc, South San Francisco, CA, United States
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven M Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
9
|
Fang F, Xu J, Kang Y, Ren H, Muyey DM, Chen X, Tan Y, Xu Z, Wang H. GATA2 rs2335052 and GATA2 rs78245253 single-nucleotide polymorphisms in Chinese patients with acute myelocytic leukemia. Int J Lab Hematol 2021; 43:1491-1500. [PMID: 34374210 DOI: 10.1111/ijlh.13649] [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: 01/21/2021] [Revised: 05/09/2021] [Accepted: 06/09/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION GATA binding protein 2 (GATA2) gene, involved in progression of hematologic malignancies and various solid tumors, is a susceptibility gene for inherited acute myeloid leukemia (AML). However, the influence of its single-nucleotide polymorphisms (SNPs) on AML remains unknown. METHODS We used allele-specific PCR to genotype GATA2 rs2335052 and rs78245253 in 159 newly diagnosed AML (non-M3) patients and 300 healthy volunteers, and all of participants came from China. And 34 common hematological tumor gene mutations in 159 AML patients were detected by next-generation sequencing. Kaplan-Meier survival analysis and Cox proportional hazard regression were used to analyze the association between the two SNPs and the prognosis of AML. RESULTS We found GATA2 rs2335052 C/T genotype, rs2335052 T/T genotype and rs78245253 G/C genotype in 51.6%, 13.8% and 11.3% AML patients. Our results demonstrated that GATA2 rs2335052 and rs78245253 were associated with certain laboratory features in AML patients, which had no effect on the pathogeny, chemotherapy response and recurrence of patients. Nevertheless, Kaplan-Meier survival analysis showed that, compared with rs78245253 G/G genotype, rs78245253 G/C genotype was significantly related to a decrease in overall survival (OS) (P = .020). Additionally, multivariate cox regression analysis showed that GATA2 rs78245253 was an independent risk factor for OS of AML patients in China. CONCLUSION GATA2 rs78245253 was an independent predictor for prognosis of AML patients in China and may be used as a potential indicator to predict the survival of AML patients in China. Further studies are needed to validate these findings and clarify the underlying mechanism.
Collapse
Affiliation(s)
- Fang Fang
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Xu
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yefang Kang
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huanying Ren
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Daniel Muteb Muyey
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiuhua Chen
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanhong Tan
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhifang Xu
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongwei Wang
- Institute of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
10
|
Human GATA2 mutations and hematologic disease: how many paths to pathogenesis? Blood Adv 2021; 4:4584-4592. [PMID: 32960960 DOI: 10.1182/bloodadvances.2020002953] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/21/2020] [Indexed: 01/19/2023] Open
Abstract
The surge of human genetic information, enabled by increasingly facile and economically feasible genomic technologies, has accelerated discoveries on the relationship of germline genetic variation to hematologic diseases. For example, germline variation in GATA2, encoding a vital transcriptional regulator of multilineage hematopoiesis, creates a predisposition to bone marrow failure and acute myeloid leukemia termed GATA2 deficiency syndrome. More than 300 GATA2 variants representing missense, truncating, and noncoding enhancer mutations have been documented. Although these variants can diminish GATA2 expression and/or function, the functional ramifications of many variants are unknown. Studies using genetic rescue and knockin mouse systems have established that GATA2 mutations differentially affect molecular processes in distinct target genes and within a single target cell. Considering that target genes for a transcription factor can differ in sensitivity to altered levels of the factor, and transcriptional mechanisms are often cell type specific, the context-dependent consequences of GATA2 mutations in experimental systems portend the complex phenotypes and interindividual variation of GATA2 deficiency syndrome. This review documents GATA2 human genetics and the state of efforts to traverse from physiological insights to pathogenic mechanisms.
Collapse
|
11
|
Vedula RS, Cheng MP, Ronayne CE, Farmakiotis D, Ho VT, Koo S, Marty FM, Lindsley RC, Bold TD. Somatic GATA2 mutations define a subgroup of myeloid malignancy patients at high risk for invasive fungal disease. Blood Adv 2021; 5:54-60. [PMID: 33570623 PMCID: PMC7805332 DOI: 10.1182/bloodadvances.2020002854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022] Open
Abstract
Invasive fungal disease (IFD) can be a severe treatment complication in patients with myeloid malignancies, but current risk models do not incorporate disease-specific factors, such as somatic gene mutations. Germline GATA2 deficiency is associated with a susceptibility to IFD. To determine whether myeloid gene mutations were associated with IFD risk, we identified 2 complementary cohorts of patients with myeloid malignancy, based on (1) the diagnosis of invasive aspergillosis (IA), or (2) the presence of GATA2 mutations identified during standard clinical sequencing. We found somatic GATA2 mutations in 5 of 27 consecutive patients who had myeloid malignancy and developed IA. Among 51 consecutive patients with GATA2 mutations identified in the evaluation of myeloid malignancy, we found that IFD was diagnosed and treated in 21 (41%), all of whom had received chemotherapy or had undergone an allogeneic stem cell transplant. Pulmonary infections and disseminated candidiasis were most common. The 90-day mortality was 52% among patients with IFD. Our results indicate that patients with somatic GATA2 mutations are a vulnerable subgroup of patients with myeloid malignancy who have high risk for treatment-associated IFD and suggest that a focused approach to antifungal prophylaxis be considered.
Collapse
Affiliation(s)
- Rahul S Vedula
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Matthew P Cheng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA
| | - Christine E Ronayne
- Division of Infectious Diseases and International Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| | - Dimitrios Farmakiotis
- Division of Infectious Diseases, Warren Alpert Medical School, Brown University, Providence, RI
| | - Vincent T Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sophia Koo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA
| | - Francisco M Marty
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA
| | - R Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Tyler D Bold
- Division of Infectious Diseases and International Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| |
Collapse
|
12
|
Wu WE, Zhou X, Xu N, Huang JX, Liu L, Tan YX, Luo J, Qin JY, Yin CX, Zhou LL, Liu XL. [Targeted next-generation sequencing for the molecular diagnosis of patients with chronic myeloid leukemia with resistance or intolerance to tyrosine kinase inhibitor]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41:848-852. [PMID: 33190443 PMCID: PMC7656074 DOI: 10.3760/cma.j.issn.0253-2727.2020.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 11/17/2022]
MESH Headings
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- High-Throughput Nucleotide Sequencing
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Mutation/drug effects
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- W E Wu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - X Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - N Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - J X Huang
- Department of Hematology, Yuebei People's Hospital, Shantou University, Shaoguan 512025, China
| | - L Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y X Tan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - J Luo
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - J Y Qin
- Annoroad Gene Technology Co. Ltd, Beijing 100176, China
| | - C X Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - L L Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - X L Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
13
|
Germline predisposition in myeloid neoplasms: Unique genetic and clinical features of GATA2 deficiency and SAMD9/SAMD9L syndromes. Best Pract Res Clin Haematol 2020; 33:101197. [PMID: 33038986 PMCID: PMC7388796 DOI: 10.1016/j.beha.2020.101197] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Increasing awareness about germline predisposition and the widespread application of unbiased whole exome sequencing contributed to the discovery of new clinical entities with high risk for the development of haematopoietic malignancies. The revised 2016 WHO classification introduced a novel category of "myeloid neoplasms with germline predisposition" with GATA2, CEBPA, DDX41, RUNX1, ANKRD26 and ETV6 genes expanding the spectrum of hereditary myeloid neoplasms (MN). Since then, more germline causes of MN were identified, including SAMD9, SAMD9L, and ERCC6L2. This review describes the genetic and clinical spectrum of predisposition to MN. The main focus lies in delineation of phenotypes, genetics and management of GATA2 deficiency and the novel SAMD9/SAMD9L-related disorders. Combined, GATA2 and SAMD9/SAMD9L (SAMD9/9L) syndromes are recognized as most frequent causes of primary paediatric myelodysplastic syndromes, particularly in setting of monosomy 7. To date, ~550 cases with germline GATA2 mutations, and ~130 patients with SAMD9/9L mutations had been reported in literature. GATA2 deficiency is a highly penetrant disorder with a progressive course that often rapidly necessitates bone marrow transplantation. In contrast, SAMD9/9L disorders show incomplete penetrance with various clinical outcomes ranging from spontaneous haematological remission observed in young children to malignant progression.
Collapse
|
14
|
Wu W, Xu N, Zhou X, Liu L, Tan Y, Luo J, Huang J, Qin J, Wang J, Li Z, Yin C, Zhou L, Liu X. Integrative Genomic Analysis Reveals Cancer-Associated Gene Mutations in Chronic Myeloid Leukemia Patients with Resistance or Intolerance to Tyrosine Kinase Inhibitor. Onco Targets Ther 2020; 13:8581-8591. [PMID: 32943879 PMCID: PMC7468532 DOI: 10.2147/ott.s257661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction While the acquisition of mutations in the ABL1 kinase domain (KD) has been identified as a common mechanism behind tyrosine kinase inhibitor (TKI) resistance, recent genetic studies have revealed that patients with TKI resistance or intolerance frequently harbor one or more genetic alterations implicated in myeloid malignancies. This suggests that additional mutations other than ABL1 KD mutations might contribute to disease progression. Methods We performed targeted-capture sequencing of 127 known and putative cancer-related genes of 63 patients with CML using next-generation sequencing (NGS), including 42 patients with TKI resistance and 21 with TKI intolerance. Results The differences in the number of mutations between groups had no statistical significance. This could be explained in part by not all of the patients having achieved major molecular remission in the early period as expected. Overall, 66 mutations were identified in 96.8% of the patients, most frequently in the KTM2C (31.82%), ABL1 (31.82%), FAT1 (25.76%), and ASXL1 (22.73%) genes. CUX1, KIT, and GATA2 were associated with TKI intolerance, and two of them (CUX1, GATA2) are transcription factors in which mutations were identified in 82.61% of patients with TKI intolerance. ASXL1 mutations were found more frequently in patients with ABL1 KD mutations (38.1% vs 15.21%, P=0.041). Although the number of mutations was low, pairwise interaction between mutated genes showed that ABL1 KD mutations cooccurred with SH2B3 mutations (P<0.05). In Kaplan-Meier analyses, only TET2 mutations were associated with shorter progression-free survival (P=0.026). Conclusion Our data suggested that the CUX1, KIT, and GATA2 genes may play important roles in TKI intolerance. ASXL1 and TET2 mutations may be associated with poor patient prognosis. NGS helps improving the clinical risk stratification, which enables the identification of patients with TKI resistance or intolerance in the era of TKI therapy.
Collapse
Affiliation(s)
- Waner Wu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Xuan Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Liang Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Yaxian Tan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Jie Luo
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Jixian Huang
- Department of Hematology, Yuebei People's Hospital, Shantou University, Shaoguan 512025, Guangdong, People's Republic of China
| | - Jiayue Qin
- Yiwu Cancer Research Center, Fudan University Shanghai Cancer Center, Yiwu, Zhejiang 322000, People's Republic of China
| | - Juan Wang
- Yiwu Cancer Research Center, Fudan University Shanghai Cancer Center, Yiwu, Zhejiang 322000, People's Republic of China
| | - Zhimin Li
- Yiwu Cancer Research Center, Fudan University Shanghai Cancer Center, Yiwu, Zhejiang 322000, People's Republic of China
| | - Changxin Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Lingling Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Xiaoli Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| |
Collapse
|