1
|
Lux S, Milsom MD. EVI1-mediated Programming of Normal and Malignant Hematopoiesis. Hemasphere 2023; 7:e959. [PMID: 37810550 PMCID: PMC10553128 DOI: 10.1097/hs9.0000000000000959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/14/2023] [Indexed: 10/10/2023] Open
Abstract
Ecotropic viral integration site 1 (EVI1), encoded at the MECOM locus, is an oncogenic zinc finger transcription factor with diverse roles in normal and malignant cells, most extensively studied in the context of hematopoiesis. EVI1 interacts with other transcription factors in a context-dependent manner and regulates transcription and chromatin remodeling, thereby influencing the proliferation, differentiation, and survival of cells. Interestingly, it can act both as a transcriptional activator as well as a transcriptional repressor. EVI1 is expressed, and fulfills important functions, during the development of different tissues, including the nervous system and hematopoiesis, demonstrating a rigid spatial and temporal expression pattern. However, EVI1 is regularly overexpressed in a variety of cancer entities, including epithelial cancers such as ovarian and pancreatic cancer, as well as in hematologic malignancies like myeloid leukemias. Importantly, EVI1 overexpression is generally associated with a very poor clinical outcome and therapy-resistance. Thus, EVI1 is an interesting candidate to study to improve the prognosis and treatment of high-risk patients with "EVI1high" hematopoietic malignancies.
Collapse
Affiliation(s)
- Susanne Lux
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael D. Milsom
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
- DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
2
|
El Hussein S, Loghavi S. The Impact of Clonal Hierarchy and Heterogeneity on Phenotypic Manifestations of Myelodysplastic Neoplasms. Cancers (Basel) 2022; 14:5690. [PMID: 36428782 PMCID: PMC9688198 DOI: 10.3390/cancers14225690] [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: 09/07/2022] [Revised: 10/30/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Until recently, conventional prognostication of myelodysplastic neoplasms (MDS) was performed using the revised International Prognostic Scoring System (IPSS-R), with additional adverse prognoses conferred by select mutations. Nonetheless, the clonal diversity and dynamics of coexisting mutations have been shown to alter the prognosis and treatment response in patients with MDS. Often in the process of clonal evolution, various initial hits are preferentially followed by a specific spectrum of secondary alterations, shaping the phenotypic and biologic features of MDS. Our ability to recapitulate the clonal ontology of MDS is a necessary step toward personalized therapy and the conceptualization of a better classification system, which ideally would take into consideration all genomic aberrations and their inferred clonal architecture in individual cases. In this review, we summarize our current understanding of the molecular landscape of MDS and the role of mutational combinations, clonal burden, and clonal hierarchy in defining the clinical fate of the disease.
Collapse
Affiliation(s)
- Siba El Hussein
- Department of Pathology, University of Rochester Medical Center, Rochester, NY 14607, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
3
|
Wang Y, Yao C, Lin C, Chen C, Hsu C, Tsai C, Hou H, Chou W, Tien H. Higher RUNX1 expression levels are associated with worse overall and leukaemia-free survival in myelodysplastic syndrome patients. EJHAEM 2022; 3:1209-1219. [PMID: 36467848 PMCID: PMC9713038 DOI: 10.1002/jha2.547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 06/17/2023]
Abstract
RUNX1 mutations are frequently detected in various myeloid neoplasms and implicate unfavourable clinical outcomes in patients with myelodysplastic syndrome (MDS) and acute myeloid leukaemia (AML). On the other hand, high expression of RUNX1 is also correlated with poor prognosis in AML patients. However, the clinical relevancy of RUNX1 expression in MDS patients remains elusive. This study aimed to investigate the prognostic and biologic impacts of RUNX1 expression in MDS patients. We recruited 341 MDS patients who had sufficient bone marrow samples for next-generation sequencing. Higher RUNX1 expression occurred more frequently in the patients with Revised International Prognostic Scoring System (IPSS-R) higher-risk MDS than the lower-risk group. It was closely associated with poor-risk cytogenetics and mutations in ASXL1, NPM1, RUNX1, SRSF2, STAG2, TET2 and TP53. Furthermore, patients with higher RUNX1 expression had significantly shorter leukaemia-free survival (LFS) and overall survival (OS) than those with lower expression. Subgroups analysis revealed that higher-RUNX1 group consistently had shorter LFS and OS than the lower-RUNX1 group, no matter RUNX1 was mutated or not. The same findings were observed in IPSS-R subgroups. In multivariable analysis, higher RUNX1 expression appeared as an independent adverse risk factor for survival. The prognostic significance of RUNX1 expression was validated in two external public cohorts, GSE 114922 and GSE15061. In summary, we present the characteristics and prognosis of MDS patients with various RUNX1 expressions and propose that RUNX1 expression complement RUNX1 mutation in MDS prognostication, wherein patients with wild RUNX1 but high expression may need more proactive treatment.
Collapse
Affiliation(s)
- Yu‐Hung Wang
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Stem Cell and Leukaemia Proteomics LaboratoryUniversity of ManchesterManchesterUK
| | - Chi‐Yuan Yao
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Graduate Institute of Clinical MedicineCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chien‐Chin Lin
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chi‐Ling Chen
- Graduate Institute of Clinical MedicineCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Chia‐Lang Hsu
- Department of Medical ResearchNational Taiwan University HospitalTaipeiTaiwan
| | - Cheng‐Hong Tsai
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Hsin‐An Hou
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Wen‐Chien Chou
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Hwei‐Fang Tien
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| |
Collapse
|
4
|
Olkhovskiy IA, Gorbenko AS, Stolyar MA, Bakhtina VI, Mikhalev MA, Olkhovik TI, Sudarikov AB, Sidorova YS, Pospelova TI, Kolesnikova MA, Kaporskaya TS, Lyskova VA. Study of mRNA of WT1, BAALC, EVI1, PRAME and HMGA2 genes in whole blood samples. Klin Lab Diagn 2022; 67:613-620. [PMID: 36315178 DOI: 10.51620/0869-2084-2022-67-10-613-620] [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] [Indexed: 06/16/2023]
Abstract
Simultaneous quantitative measurement of mRNA of the WT1, BAALC, EVI1, PRAME and HMGA2 genes in whole blood samples reflects the specific pathological proliferative activity in acute leukemia and their ratio is promising as a diagnostic marker. The transcriptome profile of acute leukemia cells is usually assessed using NGS or microarray techniques after a preliminary procedure for isolation of mononuclear cells. However, the results of using the multiplex PCR reaction for the simultaneous determination of all above mRNAs in whole blood samples have not been published so far. Determination of mRNA of WT1, BAALC, EVI1, PRAME and HMGA2 genes in venous blood level samples by multiplex RT-PCR. The study included 127 blood samples from patients who diagnosis of acute leukemia was subsequently confirmed. In the comparison group, 87 samples of patients without oncohematological diagnosis were selected, including 31 samples (K1) with a normal blood formula and 56 samples (K2) with a violation of the cellular composition - anemia, leukocytosis and thrombocytopenia. RNA isolation and reverse transcription were performed using the Ribozol-D and Reverta-L kits (TsNIIE, Russia). Determination of the mRNA expression level of the WT1, BAALC, EVI1, PRAME and HMGA2 genes by multiplex real-time PCR using a homemade multiplex PCR kit. The mRNA level was characterized by high interindividual variation and did not correlate with the rate of circulating leukocytes or blood blasts. Expression of WT1 mRNA was observed in whole blood only in one patient from the control group and in 112 (88%) patients with leukemia and was combined with a decrease in the level of HMGA2 mRNA expression and BAALC mRNA values. In contrast to the control groups, patients with leukemia had higher levels of BAALC mRNA in AML and ALL, increased PRAME mRNA in AML and APL, but lower levels of HMGA2 in APL.
Collapse
Affiliation(s)
- I A Olkhovskiy
- Krasnoyarsk branch of the «National Research Center for Hematology» Department of Health
- Federal Research Center Krasnoyarsk Scientific Center of the Siberian Branch of the Russian Academy of Sciences
| | - A S Gorbenko
- Krasnoyarsk branch of the «National Research Center for Hematology» Department of Health
- Federal Research Center Krasnoyarsk Scientific Center of the Siberian Branch of the Russian Academy of Sciences
| | - M A Stolyar
- Krasnoyarsk branch of the «National Research Center for Hematology» Department of Health
- Federal Research Center Krasnoyarsk Scientific Center of the Siberian Branch of the Russian Academy of Sciences
| | - V I Bakhtina
- Krasnoyarsk regional clinic Hospital
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University
| | | | | | - A B Sudarikov
- «National Research Center for Hematology» Department of Health
| | - Yu S Sidorova
- «National Research Center for Hematology» Department of Health
| | | | | | - T S Kaporskaya
- State-financed health care institution Irkutsk regional clinical hospital
| | - V A Lyskova
- State-financed health care institution Irkutsk regional clinical hospital
| |
Collapse
|
5
|
Verma D, Kumar R, Ali MS, Singh J, Arora M, Singh I, Kumari S, Bakhshi S, Sharma A, Palanichamy JK, Tanwar P, Singh AR, Chopra A. BAALC gene expression tells a serious patient outcome tale in NPM1-wild type/FLT3-ITD negative cytogenetically normal-acute myeloid leukemia in adults. Blood Cells Mol Dis 2022; 95:102662. [PMID: 35429905 DOI: 10.1016/j.bcmd.2022.102662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/21/2022] [Accepted: 04/05/2022] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia with normal cytogenetics (CN-AML) is the largest group of AML patients which is associated with a variegated patient outcome. Multiple molecular markers have been used to risk-stratify these patients. Estimation of expression of BAALC gene (Brain and Acute Leukemia, Cytoplasmic) mRNA level is one of the predictive markers which has been identified in multiple studies. In this study, we examined the clinical and prognostic value of BAALC gene expression in 149 adult CN-AML patients. We also utilized multi-omics databases to ascertain the association of BAALC gene expression with comprehensive molecular and clinicopathologic features in AML. BAALC overexpression was associated with CD34 positivity on leukemic blasts (p = 0.0026) and the absence of NPM1 gene mutation (p < 0.0001), presence of RUNX1 gene mutation (p < 0.001) and poor patient outcomes, particularly in NPM1-wild type/FLT3-ITD negative adult CN-AML patients. Additionally, BAALC expression was associated with the alteration of methylation of its promoter. Further, pathway analysis revealed that BAALC expression is correlated with MYC targets and Ras signalling. We conclude that high BAALC expression associates with poor patient outcome in NPM1-wild type/FLT3-ITD negative adult CN-AML patients.
Collapse
Affiliation(s)
| | | | | | - Jay Singh
- Laboratory Oncology, AIIMS, New Delhi, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Yuen KY, Lin XY, Zhou YZ, Luo H, Liu Y, Xu LH. Optimal time-points for detecting expression levels of BAALC, EVI1, and WT1 genes in patients with acute myeloid leukemia: a meta-analysis. Hematology 2021; 26:995-1006. [PMID: 34871539 DOI: 10.1080/16078454.2021.2006409] [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: 10/19/2022] Open
Abstract
OBJECTIVES This meta-analysis examined the prognostic role of brain and acute leukemia, cytoplasmic (BAALC), Ecotropic virus integration site-1 (EVI1) and Wilms' tumor 1 (WT1) genes at different time-points during conventional chemotherapy. METHODS A systematic search of publications indexed in the electronic databases from January 1988 to October 2020 was performed. Over 7525 cases of AML from 25 studies were involved. RESULTS At diagnosis, overexpression of either BAALC or EVI1 had a negative impact on complete remission achievement (Summary Odds ratios [SORs] for BAALC = 0.32; SORs for EVI1 = 0.49) and survival outcome. The summary hazard ratios of overall survival (OS) and disease-free survival (DFS) were 1.97 and 2.04 for BAALC and 1.33 and 1.86 for EVI1, respectively. The prognostic value of pretreatment WT1 levels was heterogeneous while subgroup analyses unveiled that overexpressed WT1 may correlate with a favorable outcome (summary hazard ratio [SHR] for OS = 0.42). Both WT1 and BAALC played a role in prognosis assessment at post-induction and the diagnostic performance of WT1 transcript reduction was superior to the absolute WT1 level. Post-consolidation WT1 overexpression consistently indicated an increased risk of relapse, while the combined HR for RFS was statistically insignificant (SHR = 4.22). CONCLUSION These findings confirm the application of BAALC and EVI1 at diagnosis, WT1 after induction chemotherapy in AML patients throughout conventional chemotherapy.
Collapse
Affiliation(s)
- Ka-Yuk Yuen
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiao-Ying Lin
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yong-Zhuo Zhou
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Hua Luo
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yong Liu
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Lu-Hong Xu
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
7
|
Schwind S, Jentzsch M, Kubasch AS, Metzeler KH, Platzbecker U. Myelodysplastic syndromes: Biological and therapeutic consequences of the evolving molecular aberrations landscape. Neoplasia 2021; 23:1101-1109. [PMID: 34601234 PMCID: PMC8495032 DOI: 10.1016/j.neo.2021.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 11/29/2022]
Abstract
Myelodysplastic syndromes (MDS) are clonal hematopoietic disorders with heterogeneous presentation, ranging from indolent disease courses to aggressive diseases similar to acute myeloid leukemia (AML). Approximately 90% of MDS patients harbor recurrent mutations , which – with the exception of mutated SF3B1 –have not (yet) been included into the diagnostic criteria or risk stratification for MDS. Accumulating evidence suggests their utility for diagnostic workup, treatment indication and prognosis. Subsequently, in patients with unexplained cytopenia or dysplasia identification of these mutations may lead to earlier diagnosis. The acquisition and expansion of additional driver mutations usually antecedes further disease progression to higher risk MDS or secondary AML and thus, can be clinically helpful to detect individuals that may benefit from aggressive treatment approaches. Here, we review our current understanding of somatic gene mutations, gene expression patterns and flow cytometry regarding their relevance for disease evolution from pre-neoplastic states to MDS and potentially AML.
Collapse
Affiliation(s)
- Sebastian Schwind
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Madlen Jentzsch
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Anne Sophie Kubasch
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Klaus H Metzeler
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany; German MDS Study Group (G-MDS), Leipzig, Germany; European Myelodysplastic Syndromes Cooperative Group, Leipzig, Germany.
| |
Collapse
|
8
|
Intrinsically disordered Meningioma-1 stabilizes the BAF complex to cause AML. Mol Cell 2021; 81:2332-2348.e9. [PMID: 33974912 DOI: 10.1016/j.molcel.2021.04.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/05/2021] [Accepted: 04/15/2021] [Indexed: 02/07/2023]
Abstract
Meningioma-1 (MN1) overexpression in AML is associated with poor prognosis, and forced expression of MN1 induces leukemia in mice. We sought to determine how MN1 causes AML. We found that overexpression of MN1 can be induced by translocations that result in hijacking of a downstream enhancer. Structure predictions revealed that the entire MN1 coding frame is disordered. We identified the myeloid progenitor-specific BAF complex as the key interaction partner of MN1. MN1 over-stabilizes BAF on enhancer chromatin, a function directly linked to the presence of a long polyQ-stretch within MN1. BAF over-stabilization at binding sites of transcription factors regulating a hematopoietic stem/progenitor program prevents the developmentally appropriate decommissioning of these enhancers and results in impaired myeloid differentiation and leukemia. Beyond AML, our data detail how the overexpression of a polyQ protein, in the absence of any coding sequence mutation, can be sufficient to cause malignant transformation.
Collapse
|
9
|
Cui W, Liu Y, Tan Y, Peng X, Cui L, Cheng Z, Dai Y, Fu L, Zeng T, Liu Y. Prognostic value of HMGN family expression in acute myeloid leukemia. Future Oncol 2021; 17:541-548. [PMID: 33467898 DOI: 10.2217/fon-2020-0555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aim: The objective of this work was to investigate the prognostic role of the HMGN family in acute myeloid leukemia (AML). Methods: A total of 155 AML patients with HMGN1-5 expression data from the Cancer Genome Atlas database were enrolled in this study. Results: In the chemotherapy-only group, patients with high HMGN2 expression had significantly longer event-free survival (EFS) and overall survival (OS) than those with low expression (all p < 0.05), whereas high HMGN5 expressers had shorter EFS and OS than the low expressers (all p < 0.05). Multivariate analysis identified that high HMGN2 expression was an independent favorable prognostic factor for patients who only received chemotherapy (all p < 0.05). HMGN family expression had no impact on EFS and OS in AML patients receiving allogeneic hematopoietic stem cell transplantation. Conclusion: High HMGN2/5 expression is a potential prognostic indicator for AML.
Collapse
Affiliation(s)
- Wei Cui
- Department of Clinical Laboratory, Beijing Haidian Hospital, Beijing Haidian Section of Peking University Third Hospital, Beijing, 100080, China
| | - Yuna Liu
- Department of Clinical Laboratory Medicine, Beijing Hospital of Integrated Chinese & Western Medicine, Beijing, 100039, China
| | - Yanan Tan
- Department of Clinical Laboratory, Beijing Haidian Hospital, Beijing Haidian Section of Peking University Third Hospital, Beijing, 100080, China
| | - Xingyue Peng
- Department of Clinical Laboratory, Beijing Haidian Hospital, Beijing Haidian Section of Peking University Third Hospital, Beijing, 100080, China
| | - Longzhen Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Zhiheng Cheng
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, 999025, Netherlands
| | - Yifeng Dai
- Immunoendocrinology, Division of Medical Biology, Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, 999025, Netherlands
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Tiansheng Zeng
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yan Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| |
Collapse
|
10
|
Molecular-Based Score inspired on metabolic signature improves prognostic stratification for myelodysplastic syndrome. Sci Rep 2021; 11:1675. [PMID: 33462268 PMCID: PMC7814118 DOI: 10.1038/s41598-020-80918-6] [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: 07/02/2020] [Accepted: 12/29/2020] [Indexed: 01/29/2023] Open
Abstract
Deregulated cellular energetics is formally incorporated as an emerging hallmark of cancer, however little is known about its processes in myelodysplastic syndromes (MDS). Using transcriptomic data of CD34+ cells from 159 MDS patients and 17 healthy donors, we selected 37 genes involved in cellular energetics and interrogated about its clinical and prognostic functions. Based on the low expression of ACLY, ANPEP, and PANK1, as well as high expression of PKM and SLC25A5, we constructed our Molecular-Based Score (MBS), that efficiently discriminated patients at three risks groups: favourable risk (n = 28; 3-year overall survival (OS): 100%); intermediate (n = 60; 76% [62-93%]) and adverse (n = 71; 35% [17-61%]). Adverse MBS risk was independently associated with inferior OS (HR = 10.1 [95% CI 1.26-81]; P = 0.029) in multivariable analysis using age, gender and the revised international prognostic score system as confounders. Transcriptional signature revealed that Favourable- and intermediate-risk patients presented enriched molecular programs related to mature myeloid progenitors, cell cycle progression, and oxidative phosphorylation, indicating that this cells differs in their origin, metabolic state, and cell cycle regulation, in comparison to the adverse-risk. Our study provides the first evidence that cellular energetics is transcriptionally deregulated in MDS CD34+ cells and establishes a new useful prognostic score based on the expression of five genes.
Collapse
|
11
|
Acute Myeloid Leukemia (AML): Upregulation of BAALC/MN1/MLLT11/EVI1 Gene Cluster Relate With Poor Overall Survival and a Possible Linkage With Coexpression of MYC/BCL2 Proteins. Appl Immunohistochem Mol Morphol 2019; 26:483-488. [PMID: 28362701 DOI: 10.1097/pai.0000000000000452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Molecular heterogeneity accounts for the variable and often poor prognosis in acute myeloid leukemia (AML). The current risk stratification strategy in clinical practice is limited to karyotyping and limited molecular studies screening for genetic mutations such as FLT-3 and NPM1. There is opportunity to identify further molecular prognostic markers, which may also lay the groundwork for the development of novel targeted therapies. Complex molecular technologies require transition into widely available laboratory platforms, for better integration into routine clinical practice. METHOD In a defined subset (MYC/BCL2 or MYC/BCL2) of AML patients (n=20), we examined expression signature of several genes (n=12) of established prognostic value in AML. RNA expression and MYC/BCL2 protein pattern was correlated with 3 cytogenetic risk groups and overall survival. RESULTS K-means++ unsupervised clustering defined 2 distinct groups with high and low transcript levels of BAALC/MN1/MLLT11/EVI1/SOCS2 genes (>2.5-fold difference; P<0.001). This mRNA signature trended with higher prevalence of MYC/BCL2 coexpression (P<0.057) and poor overall survival (P<0.036), but did not correlate with conventional cytogenetic risk groups (P<0.084). CONCLUSIONS This pilot study provides useful data, which may help further refine the prognostic scheme of AML patients outside conventional cytogenetic risk groups. It also presents some biological rationale for future studies to explore the use of novel agents targeting MYC and/or BCL2 genes in combination with conventional chemotherapy protocols for AML.
Collapse
|
12
|
Combining gene mutation with gene expression analysis improves outcome prediction in acute promyelocytic leukemia. Blood 2019; 134:951-959. [PMID: 31292112 DOI: 10.1182/blood.2019000239] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/23/2019] [Indexed: 12/15/2022] Open
Abstract
By combining the analysis of mutations with aberrant expression of genes previously related to poorer prognosis in both acute promyelocytic leukemia (APL) and acute myeloid leukemia, we arrived at an integrative score in APL (ISAPL) and demonstrated its relationship with clinical outcomes of patients treated with all-trans retinoic acid (ATRA) in combination with anthracycline-based chemotherapy. Based on fms-like tyrosine kinase-3-internal tandem duplication mutational status; the ΔNp73/TAp73 expression ratio; and ID1, BAALC, ERG, and KMT2E gene expression levels, we modeled ISAPL in 159 patients (median ISAPL score, 3; range, 0-10). ISAPL modeling identified 2 distinct groups of patients, with significant differences in early mortality (P < .001), remission (P = .004), overall survival (P < .001), cumulative incidence of relapse (P = .028), disease-free survival (P = .03), and event-free survival (P < .001). These data were internally validated by using a bootstrap resampling procedure. At least for patients treated with ATRA and anthracycline-based chemotherapy, ISAPL modeling may identify those who need to be treated differently to maximize their chances for a cure.
Collapse
|
13
|
Sakhdari A, Tang Z, Ok CY, Bueso-Ramos CE, Medeiros LJ, Huh YO. Homogeneously staining region (hsr) on chromosome 11 is highly specific for KMT2A amplification in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Cancer Genet 2019; 238:18-22. [PMID: 31425921 DOI: 10.1016/j.cancergen.2019.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 01/01/2023]
Abstract
AML and MDS are most common myeloid neoplasms that affect mainly older patients. Overexpression of certain proto-oncogenes plays an indispensable role in tumorigenesis and overexpression can be a consequence of gene rearrangement, amplification and/or mutation. Rearrangement and amplification of KMT2A located at chromosome band 11q23 is a well-characterized genetic driver in a subset of AML/MDS cases and is associated with a poor prognosis. The presence of homogeneously staining regions (hsr) also has been correlated with amplification of specific proto-oncogenes. In this study, we correlated hsr(11)(q23) with KMT2A in a large cohort of AML/MDS (n = 54) patients. We identified 37 patients with hsr(11)(q23) in the setting of AML (n = 27) and MDS (n = 10). All patients showed a complex karyotype including 12 cases with monosomy 17. KMT2A FISH analysis was available for 35 patients which showed KMT2A amplification in all patients. Among control cases with hsr involving chromosomes other than 11q [non-11q hsr, n = 17], FISH analysis for KMT2A was available in 10 cases and none of these cases showed KMT2A amplification (p = 0.0001, Fisher's exact test, two-tailed). Mutational analysis was performed in 32 patients with hsr(11)(q23). The most common mutated gene was TP53 (n = 29), followed by DNMT3A (n = 4), NF1 (n = 4), and TET2 (n = 3). Thirty (83%) patients died over a median follow-up of 7.6 months (range, 0.4-33.4). In summary, hsr(11)(q23) in AML/MDS cases is associated with a complex karyotype, monosomy 17, KMT2A amplification, and TP53 mutation.
Collapse
Affiliation(s)
- Ali Sakhdari
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, United States.
| | - Zhenya Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, United States
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, United States
| | - Carlos E Bueso-Ramos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, United States
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, United States
| | - Yang O Huh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, United States
| |
Collapse
|
14
|
High IFITM3 expression predicts adverse prognosis in acute myeloid leukemia. Cancer Gene Ther 2019; 27:38-44. [PMID: 30923336 DOI: 10.1038/s41417-019-0093-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is a malignancy caused by the uncontrolled and dysregulated clonal expansion of abnormal myeloid primordial cells. In general, the prognosis of AML remains poor despite new discoveries in its pathogenesis and treatment. It is crucial to find early and sensitive biomarkers and continue to explore active targeted treatments. Interferon-induced transmembrane protein (IFITM) family is an important part of the interferon signaling pathway and participate in the regulation of immune cell signaling, adhesion, cancer, and liver cell migration. However, the clinical and prognostic value of the IFITM family in AML has rarely been studied. We screened The Cancer Genome Atlas database and found 155 AML patients with IFITM family (IFITM1-5) expression data. In patients who only received chemotherapy, those with high IFITM3 expression had significantly shorter event-free survival (EFS) and overall survival (OS) than patients with low expression (all P < 0.05). Multivariate analysis demonstrated that high IFITM3 expression was an independent risk factor for EFS and OS in patients only received chemotherapy (all P < 0.05). In patients who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT), however, all IFITM members had no impact on either EFS or OS. In conclusion, our study elucidated that high IFITM3 expression could be an adverse prognostic factor for AML, whose effect might be overcome by allo-HSCT.
Collapse
|
15
|
Gene expression and risk of leukemic transformation in myelodysplasia. Blood 2017; 130:2642-2653. [DOI: 10.1182/blood-2017-05-783050] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/11/2017] [Indexed: 01/08/2023] Open
Abstract
Key Points
Through a comprehensive transcriptomic analysis, we discovered 2 major subgroups of myelodysplasia defined by gene expression profiles. The gene expression–based subgroups had independent prognostic value, which was validated in an external cohort.
Collapse
|
16
|
Almeida A, Fenaux P, List AF, Raza A, Platzbecker U, Santini V. Recent advances in the treatment of lower-risk non-del(5q) myelodysplastic syndromes (MDS). Leuk Res 2017; 52:50-57. [DOI: 10.1016/j.leukres.2016.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/27/2016] [Accepted: 11/11/2016] [Indexed: 12/12/2022]
|
17
|
Xu B, Naughton D, Busam K, Pulitzer M. ERG Is a Useful Immunohistochemical Marker to Distinguish Leukemia Cutis From Nonneoplastic Leukocytic Infiltrates in the Skin. Am J Dermatopathol 2016; 38:672-7. [PMID: 26909589 PMCID: PMC5026187 DOI: 10.1097/dad.0000000000000491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Leukemia cutis (LC) and reactive myeloid infiltrates in the skin may be difficult to distinguish pathologically, sometimes even after an extensive immunohistochemical work-up. This poses a serious clinical dilemma, as the prognosis and treatment of either condition are markedly different. Although most reactive myeloid infiltrates require a simple course of corticosteroids before the symptoms regress, the development of LC may require chemotherapeutic or transplant-variant interventions. Erythroblast transformation specific regulated gene-1 (ERG) is a member of the erythroblast transformation specific family of transcription factors, which are downstream effectors of mitogenic signaling transduction pathways. ERG is a key regulator of cell proliferation, differentiation, angiogenesis, inflammation, and apoptosis and has recently been found to be overexpressed in acute myeloid and lymphoblastic leukemia. In this study, the authors aimed to explore the diagnostic utility of ERG immunohistochemistry in LC by comparing the frequency and expression level of ERG immunostain in 32 skin biopsies, 16 with LC and 16 with reactive leukocytic infiltrates. A significantly higher frequency of ERG positivity was detected in LC (13/16, 81.4%), compared with reactive conditions (0/16). In addition, the expression level of ERG in LC, calculated using H score (mean ± standard error of mean, 188 ± 24), was significantly higher than that in nonneoplastic leukocytic infiltrate (28 ± 8). Our results strongly suggest that ERG expression is potentially an extremely useful marker to distinguish between cases of LC from those of reactive myeloid infiltrates in the skin with a positive predictive value of 100% and negative predictive value of 84.2%.
Collapse
Affiliation(s)
- Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | |
Collapse
|
18
|
Lewen M, Gresh R, Queenan M, Paessler M, Pillai V, Hexner E, Frank D, Bagg A, Aplenc R, Caywood E, Wertheim G. Pediatric chronic myeloid leukemia with inv(3)(q21q26.2) and T lymphoblastic transformation: a case report. Biomark Res 2016; 4:14. [PMID: 27453784 PMCID: PMC4957483 DOI: 10.1186/s40364-016-0069-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/07/2016] [Indexed: 12/11/2022] Open
Abstract
Background Chronic myeloid leukemia (CML) comprises ~3 % of pediatric leukemia. Although therapy with tyrosine kinase inhibitors (TKIs) is highly effective for CML, multiple factors have been identified as predictive of treatment failure. Chromosomal abnormalities involving the MECOM locus at 3q26 portend therapy resistant disease in adults, yet have never been described in pediatric patients and have not been associated with T lymphoblastic progression. Case presentation We present a case of an 11-year-old boy with CML possessing the unique combination of T lymphoblastic transformation and a subclone harboring inv(3)(q21q26.2) at diagnosis. This is the first reported case of pediatric CML with inv(3)(q21q26.2) and the first case of T lymphoblastic progression associated with this karyotype. The patient was treated with single agent TKI therapy with robust initial response. Marrow histology at one month showed restoration of trilineage hematopoiesis and BCR-ABL RT-PCR at three months showed a 1.4 log reduction in transcript levels. Conclusions The karyotypic abnormality of inv(3)(q21q26.2) in CML is not restricted to adult patients. Moreover, while chromosome 3 abnormalities are markers of TKI resistance in adults, our patient showed a robust early response to single agent TKI therapy. This finding suggests pediatric CML with inv(3)(q21q26.2) may have distinct features and more favorable treatment responses than those described in adults.
Collapse
Affiliation(s)
- Margaret Lewen
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts USA
| | - Renee Gresh
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware USA
| | - Maria Queenan
- Department of Pathology and Laboratory Medicine, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware USA
| | - Michele Paessler
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania USA ; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Vinodh Pillai
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania USA ; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Elizabeth Hexner
- Department of Medicine, Division of Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Dale Frank
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Richard Aplenc
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania USA
| | - Emi Caywood
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware USA
| | - Gerald Wertheim
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania USA ; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania USA
| |
Collapse
|
19
|
Harnan S, Ren S, Gomersall T, Everson-Hock ES, Sutton A, Dhanasiri S, Kulasekararaj A. Association between Transfusion Status and Overall Survival in Patients with Myelodysplastic Syndromes: A Systematic Literature Review and Meta-Analysis. Acta Haematol 2016; 136:23-42. [PMID: 27160308 DOI: 10.1159/000445163] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 03/02/2016] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Multiple studies show that transfusion independence (TI) in myelodysplastic syndrome (MDS) has a positive impact on overall survival (OS). To assess this, a systematic review and meta-analysis of the association between TI and OS in patients with MDS was conducted (PROSPERO ID: CRD42014007264). METHODS Comprehensive searches of 5 key bibliographic databases were conducted and supplemented with additional search techniques. Included were studies that had recruited adults aged >18 years with MDS and had examined the impact of transfusion status on OS. RESULTS Fifty-five studies (89 citations) were included. The vast majority reported a statistically significant hazard ratio (HR) for OS in favor of TI patients or in patients who acquired TI after treatment. A random-effects meta-analysis was conducted. Patients classed as TI at baseline showed a 59% decrease in the risk of death compared with transfusion-dependent (TD) patients [HR 0.41; 95% credible interval (CrI) 0.29-0.56], and this effect did not appear to interact significantly with illness severity (interaction coefficient HR 1.38; 95% CrI 0.62-3.41). A meta-analysis of studies where patients acquired TI was not possible, but those studies consistently reported a survival benefit for those who acquired TI. CONCLUSION The findings revealed a 59% pooled reduction in mortality among TI patients when compared with TD patients.
Collapse
Affiliation(s)
- Sue Harnan
- Health Economics and Decision Science, School of Health and Related Research, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | |
Collapse
|
20
|
Weber S, Haferlach C, Jeromin S, Nadarajah N, Dicker F, Noël L, Zenger M, Alpermann T, Kern W, Haferlach T, Schnittger S. Gain of chromosome 21 or amplification of chromosome arm 21q is one mechanism for increased ERG expression in acute myeloid leukemia. Genes Chromosomes Cancer 2015; 55:148-57. [PMID: 26542308 DOI: 10.1002/gcc.22321] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 01/19/2023] Open
Abstract
In acute myeloid leukemia (AML), acquired genomic gains and losses are common and lead to altered expression of genes located within or nearby the affected regions. Increased expression of the ETS-related transcription factor gene ERG has been described in myeloid malignancies with chromosomal rearrangements involving chromosome band 21q22, but also in cytogenetically normal AML, where it is associated with adverse prognosis. In this study, fluorescence in situ hybridization on interphase nuclei disclosed an amplification of the ERG gene (more than six copies) in 33 AML patients with structural rearrangements of 21q22. Array comparative genomic hybridization of these cases disclosed a minimal amplified region at the position 39.6-40.0 Mbp from pter that harbors ERG as the only gene. Analysis by quantitative real-time reverse transcription polymerase chain reaction revealed significantly higher ERG mRNA expression in these patients and in a group of 95 AML patients with complete or partial gain of chromosome 21 (three to six copies) compared with 351 AML patients without gain of chromosome 21. Quantification of ERG DNA copy numbers revealed a strong correlation with ERG mRNA expression. Furthermore, in patients with gain of chromosome 21, higher ERG expression was found to be associated with RUNX1 mutations. Our results suggest that acquired gain of chromosome 21 or amplification of chromosome arm 21q is one mechanism contributing to increased ERG expression in AML.
Collapse
Affiliation(s)
| | | | | | | | | | - Louisa Noël
- MLL Munich Leukemia Laboratory, Munich, Germany
| | | | | | | | | | | |
Collapse
|
21
|
Hecht A, Nowak D, Nowak V, Hanfstein B, Büchner T, Spiekermann K, Weiß C, Hofmann WK, Lengfelder E, Nolte F. A molecular risk score integrating BAALC, ERG and WT1 expression levels for risk stratification in acute promyelocytic leukemia. Leuk Res 2015; 39:S0145-2126(15)30363-5. [PMID: 26344466 DOI: 10.1016/j.leukres.2015.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/15/2015] [Indexed: 11/23/2022]
Abstract
To date risk stratification in acute promyelocytic leukemia (APL) is based on highly dynamic leukocyte and platelet counts only. To identify a more robust risk stratification model, a molecular risk score was developed based on expression levels of the genes BAALC, ERG and WT1. Hereby, the main focus was on prediction of relapse. The integrative risk score divided patients into two groups with highly significant differences in outcome. It discriminated a high risk group with a high incidence of relapse successfully from a low risk group with no APL-related events after achievement of first remission. Especially the concurrent presence of molecular risk factors showed to be a negative prognostic factor in APL. The molecular risk score might be a promising approach to guide monitoring of APL patients and therapeutic decisions in the future.
Collapse
Affiliation(s)
- Anna Hecht
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Germany.
| | - Daniel Nowak
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Germany
| | - Benjamin Hanfstein
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Germany
| | - Thomas Büchner
- Department of Hematology/Oncology, University of Münster, Münster, Germany
| | | | - Christel Weiß
- Department for Medical Statistics and Biomathematics, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Germany
| | - Eva Lengfelder
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Germany
| | - Florian Nolte
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Germany
| |
Collapse
|
22
|
Steinmetz B, Hackl H, Slabáková E, Schwarzinger I, Smějová M, Spittler A, Arbesu I, Shehata M, Souček K, Wieser R. The oncogene EVI1 enhances transcriptional and biological responses of human myeloid cells to all-trans retinoic acid. Cell Cycle 2015; 13:2931-43. [PMID: 25486480 PMCID: PMC4613657 DOI: 10.4161/15384101.2014.946869] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The product of the ecotropic virus integration site 1 (EVI1) gene, whose overexpression is associated with a poor prognosis in myeloid leukemias and some epithelial tumors, regulates gene transcription both through direct DNA binding and through modulation of the activity of other sequence specific transcription factors. Previous results from our laboratory have shown that EVI1 influenced transcription regulation in response to the myeloid differentiation inducing agent, all-trans retinoic acid (ATRA), in a dual manner: it enhanced ATRA induced transcription of the RARβ gene, but repressed the ATRA induction of the EVI1 gene itself. In the present study, we asked whether EVI1 would modulate the ATRA regulation of a larger number of genes, as well as biological responses to this agent, in human myeloid cells. U937 and HL-60 cells ectopically expressing EVI1 through retroviral transduction were subjected to microarray based gene expression analysis, and to assays measuring cellular proliferation, differentiation, and apoptosis. These experiments showed that EVI1 modulated the ATRA response of several dozens of genes, and in fact reinforced it in the vast majority of cases. A particularly strong synergy between EVI1 and ATRA was observed for GDF15, which codes for a member of the TGF-β superfamily of cytokines. In line with the gene expression results, EVI1 enhanced cell cycle arrest, differentiation, and apoptosis in response to ATRA, and knockdown of GDF15 counteracted some of these effects. The potential clinical implications of these findings are discussed.
Collapse
Key Words
- AML, acute myeloid leukemia
- APL, acute promyelocytic leukemia
- ATRA, all-trans retinoic acid
- Ar, ATRA regulation
- DMSO, dimethyl sulfoxide
- EVI1
- Em, EVI1 modulation
- Er, EVI1 regulation
- FBS, fetal bovine serum
- FC, fold change
- FDR, false discovery rate
- GDF15
- GFP, green fluorescent protein
- MDS, myelodysplastic syndrome
- PSG, penicillin streptomycin glutamine
- RAR, retinoic acid receptor
- RARE, retinoic acid response element
- SE, standard error
- all-trans retinoic acid
- apoptosis
- cell cycle
- gene expression profiling
- mcoEvi1, murine codon optimized Evi1
- myeloid differentiation
Collapse
Affiliation(s)
- Birgit Steinmetz
- a Department of Medicine I ; Medical University of Vienna ; Währinger Gürtel, Vienna , Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Minetto P, Guolo F, Clavio M, De Astis E, Colombo N, Grasso R, Fugazza G, Sessarego M, Lemoli RM, Gobbi M, Miglino M. Combined assessment of WT1 and BAALC gene expression at diagnosis may improve leukemia-free survival prediction in patients with myelodysplastic syndromes. Leuk Res 2015; 39:866-73. [DOI: 10.1016/j.leukres.2015.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/08/2015] [Accepted: 04/19/2015] [Indexed: 11/24/2022]
|
24
|
Abstract
Myelodysplastic syndromes are a collection of clonal hematopoietic disorders with a wide range of clinical manifestations and eventual outcomes. Accurate prediction of a patient's prognosis is useful to define the risk posed by the disease and which treatment options are most appropriate. Several models have been created to help predict the prognosis for patients with myelodysplastic syndromes. The International Prognostic Scoring System (IPSS) has been the standard tool used to risk stratify MDS patients since its publication in 1997. Other models have since been created to improve upon the IPSS, including the recent Revised International Prognostic Scoring System. Most models include the presence or severity of peripheral blood cytopenias, the proportion of bone marrow blasts, and specific karyotype abnormalities. Other factors including age, performance status, co-morbidities, transfusion dependence, and molecular biomarkers can further refine the prediction of prognosis in some models. Novel, disease specific biomarkers with prognostic value in myelodysplastic syndromes including cell surface markers, gene expression profiles, and high resolution copy number analyses have been proposed but not yet adopted clinically. Somatic abnormalities in recurrently mutated genes are the most informative prognostic biomarkers not currently considered by clinical risk models. Mutations in specific genes have independent prognostic significance and, unlike cytogenetic abnormalities, are present in the majority of myelodysplastic syndrome cases. However, mutational information can be complex and there are challenges to its clinical implementation. Despite these limitations, DNA sequencing can refine the prediction of prognosis for myelodysplastic syndrome patients and has become increasingly available in the clinic where it will help improve the care of patients with myelodysplastic syndromes.
Collapse
Affiliation(s)
- Rafael Bejar
- Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
25
|
EVI1 promotes tumor growth via transcriptional repression of MS4A3. J Hematol Oncol 2015; 8:28. [PMID: 25886616 PMCID: PMC4389965 DOI: 10.1186/s13045-015-0124-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 02/26/2015] [Indexed: 01/07/2023] Open
Abstract
Background The transcription factor Ecotropic Virus Integration site 1 (EVI1) regulates cellular proliferation, differentiation, and apoptosis, and its overexpression contributes to an aggressive course of disease in myeloid leukemias and other malignancies. Notwithstanding, knowledge about the target genes mediating its biological and pathological functions remains limited. We therefore aimed to identify and characterize novel EVI1 target genes in human myeloid cells. Methods U937T_EVI1, a human myeloid cell line expressing EVI1 in a tetracycline regulable manner, was subjected to gene expression profiling. qRT-PCR was used to confirm the regulation of membrane-spanning-4-domains subfamily-A member-3 (MS4A3) by EVI1. Reporter constructs containing various parts of the MS4A3 upstream region were employed in luciferase assays, and binding of EVI1 to the MS4A3 promoter was investigated by chromatin immunoprecipitation. U937 derivative cell lines experimentally expressing EVI1 and/or MS4A3 were generated by retroviral transduction, and tested for their tumorigenicity by subcutaneous injection into severe combined immunodeficient mice. Results Gene expression microarray analysis identified 27 unique genes that were up-regulated, and 29 unique genes that were down-regulated, in response to EVI1 induction in the human myeloid cell line U937T. The most strongly repressed gene was MS4A3, and its down-regulation by EVI1 was confirmed by qRT-PCR in additional, independent experimental model systems. MS4A3 mRNA levels were also negatively correlated with those of EVI1 in several published AML data sets. Reporter gene assays and chromatin immunoprecipitation showed that EVI1 regulated MS4A3 via direct binding to a promoter proximal region. Experimental re-expression of MS4A3 in an EVI1 overexpressing cell line counteracted the tumor promoting effect of EVI1 in a murine xenograft model by increasing the rate of apoptosis. Conclusions Our data reveal MS4A3 as a novel direct target of EVI1 in human myeloid cells, and show that its repression plays a role in EVI1 mediated tumor aggressiveness. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0124-6) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Thol F, Scherr M, Kirchner A, Shahswar R, Battmer K, Kade S, Chaturvedi A, Koenecke C, Stadler M, Platzbecker U, Thiede C, Schroeder T, Kobbe G, Bug G, Ottmann O, Hofmann WK, Kröger N, Fiedler W, Schlenk R, Döhner K, Döhner H, Krauter J, Eder M, Ganser A, Heuser M. Clinical and functional implications of microRNA mutations in a cohort of 935 patients with myelodysplastic syndromes and acute myeloid leukemia. Haematologica 2014; 100:e122-4. [PMID: 25552704 DOI: 10.3324/haematol.2014.120345] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Felicitas Thol
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Michaela Scherr
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Aylin Kirchner
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Rabia Shahswar
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Karin Battmer
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Sofia Kade
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Anuhar Chaturvedi
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Michael Stadler
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Uwe Platzbecker
- Medizinische Klinik und Poliklinik I, Universtitätsklinikum Carl Gustav Carus, Dresden
| | - Christian Thiede
- Medizinische Klinik und Poliklinik I, Universtitätsklinikum Carl Gustav Carus, Dresden
| | - Thomas Schroeder
- Klinik für Hämatologie, Onkologie und Klinische Immunologie, Heinreich Heine Universit¨at, Düsseldorf
| | - Guido Kobbe
- Klinik für Hämatologie, Onkologie und Klinische Immunologie, Heinreich Heine Universit¨at, Düsseldorf
| | - Gesine Bug
- Department of Medicine, Hematology/Oncology, University Hospital, Frankfurt
| | - Oliver Ottmann
- Department of Medicine, Hematology/Oncology, University Hospital, Frankfurt
| | | | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Walter Fiedler
- Department of Medicine II, Oncological Center, Hubertus Wald University Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg
| | | | | | | | - Jürgen Krauter
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover Klinikum Braunschweig, Germany
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and HSCT, Hannover Medical School, Hannover
| |
Collapse
|
27
|
Jonas BA, Greenberg PL. MDS prognostic scoring systems – past, present, and future. Best Pract Res Clin Haematol 2014; 28:3-13. [PMID: 25659725 DOI: 10.1016/j.beha.2014.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 11/04/2014] [Indexed: 11/30/2022]
Abstract
The myelodysplastic syndromes (MDS) are a heterogeneous group of clonal myeloid haemopathies characterized by defective differentiation of haematopoietic cells and expansion of the abnormal clone. This leads to bone marrow failure with the resulting peripheral blood cytopenias and evolution to or toward acute myeloid leukaemia that characterize MDS clinically. The clinical heterogeneity of MDS has led several groups to analyze patient and clinical characteristics to develop prognostic scoring systems yielding estimates of overall and leukaemia-free survival to guide clinical decision-making. These models have evolved over time as our understanding of the pathogenesis, natural history, and treatment of MDS has improved. Rapid advances in flow cytometric analysis, adjuncts to standard metaphase cytogenetics, and gene mutation analysis are revolutionizing our understanding of MDS pathogenesis and prognosis. Despite the existence of multiple well-validated prognostic scoring systems, further refinements of current models with these new sources of prognostic data are needed and are described herein.
Collapse
Affiliation(s)
- Brian A Jonas
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis School of Medicine, UC Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA 95817, United States.
| | - Peter L Greenberg
- Department of Internal Medicine, Division of Hematology, Stanford University School of Medicine, Stanford Comprehensive Cancer Center, 875 Blake Wilbur Drive, Stanford, CA 94305, United States.
| |
Collapse
|
28
|
Volkert S, Schnittger S, Zenger M, Kern W, Haferlach T, Haferlach C. Amplification of EVI1 on cytogenetically cryptic double minutes as new mechanism for increased expression of EVI1. Cancer Genet 2014; 207:103-8. [PMID: 24726268 DOI: 10.1016/j.cancergen.2014.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 10/25/2022]
Abstract
In acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS), increased expression of EVI1 (ecotropic virus integration site 1) was found to be associated with adverse prognosis. Although increased expression of the EVI1 gene is mainly caused by chromosomal rearrangements involving chromosome band 3q26, where EVI1 is located, it can also be identified in cases without these rearrangements. The mechanisms that cause increased EVI1 expression in the absence of 3q26 rearrangements remain unclear. Here, we present four cases with increased EVI1 expression due to EVI1 amplification on cytogenetically cryptic double minutes (dmin). The dmin are small acentric chromosome structures and were observed in about 1% of AML and MDS. We investigated the four cases by conventional cytogenetics, fluorescence in situ hybridization, and array comparative genomic hybridization. Furthermore, EVI1 expression was measured by quantitative reverse transcriptase-PCR. By conventional chromosome analysis, the EVI1 dmin cannot be detected, due to the small size of the amplicons of 0.49-0.78 Mbp. Median % EVI1/ABL expression was 88.9% and therefore comparable to the median % EVI1/ABL expression of patients with EVI1 rearrangements. In conclusion, EVI1 amplification on cytogenetically cryptic dmin causes increased expression of EVI1 and is a new mechanism that causes increased EVI1 expression without a 3q26 rearrangement.
Collapse
|
29
|
Liu Y, Ying W, Ren Z, Gu W, Zhang Y, Yan G, Yang P, Liu Y, Yin X, Chang C, Jiang J, Fan F, Zhang C, Xu P, Wang Q, Wen B, Lin L, Wang T, Du C, Zhong J, Wang T, He QY, Qian X, Lou X, Zhang G, Zhong F. Chromosome-8-coded proteome of Chinese Chromosome Proteome Data set (CCPD) 2.0 with partial immunohistochemical verifications. J Proteome Res 2013; 13:126-36. [PMID: 24328083 DOI: 10.1021/pr400902u] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We upgraded the preliminary CCPD 1.0 to CCPD 2.0 using the latest deep-profiling proteome (CCPD 2013) of three hepatocellular carcinoma (HCC) cell lines, namely, Hep3B, MHCC97H, and HCCLM3 (ProteomeXchange identifiers: PXD000529, PXD000533, and PXD000535). CCPD 2.0 totally covered 63.6% (438/689) of Chr. 8-coded proteins and 62.6% (439/701) of Chr. 8-coded protein-coding genes. Interestingly, we found that the missing proteins exhibited a tendency to form a cluster region in chromosomes, such as two β-defensins clusters in Chr. 8, caused perhaps by their inflammation-related features. For the 41 Chr. 8-coded proteins being weakly or barely identified previously, we have performed an immunohistochemical (IHC) verification in 30 pairs of carcinoma/para-carcinoma HCC and 20 noncancerous liver tissues and confirmed their expressional evidence and occurrence proportions in tissue samples. We also verified 13 Chr. 8-coded HCC tumorigenesis-associated depleting or deficient proteins reported in CCPD 1.0 using IHC and screened 16 positive and 24 negative HCC metastatic potential-correlated proteins from large-scale label-free proteome quantitation data of CCPD 2013. Our results suggest that the selection of proper samples and the methodology to look for targeted missing proteins should be carefully considered in further verifications for the remaining Chr. 8-coded proteins.
Collapse
Affiliation(s)
- Yang Liu
- Institutes of Biomedical Sciences, Fudan University , Mingdao Bldg. 815, 138 Yixueyuan Road, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Abstract
Establishing the prognosis for patients with myelodysplastic syndromes (MDS) is a key element of their care. It helps patients understand the severity of their disease and set expectations for their future. For physicians, an accurate estimate of prognosis drives decisions about the timing and choice of therapeutic options to consider. The International Prognostic Scoring System (IPSS) has been the standard tool for MDS risk stratification since it was released in 1997. It has been used to describe patients in pivotal clinical trials and is a key element of practice guidelines. Subsequent changes to the classification scheme for MDS and an underestimation of risk in some patients from the low and intermediate-1 categories have led to the development of several newer prognostic models. The most recent is the revised IPSS (IPSS-R), which addresses several of the perceived deficiencies of its predecessor. Despite their utility, none of the available prognostic systems incorporates disease-related molecular abnormalities such as somatic mutations. These lesions are present in the nearly all cases and many have been shown to improve upon existing prognostic models. However, the interpretation of somatic mutations can be challenging and it is not yet clear how best to combine them with clinical predictors of outcome. Here I review several prognostic scoring systems developed after the IPSS and describe the emerging use of molecular markers to refine risk stratification in the MDS patient population.
Collapse
|
31
|
Immunophenotyping in myelodysplastic syndromes can add prognostic information to well-established and new clinical scores. PLoS One 2013; 8:e81048. [PMID: 24324660 PMCID: PMC3855682 DOI: 10.1371/journal.pone.0081048] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 10/17/2013] [Indexed: 12/30/2022] Open
Abstract
Background myelodysplastic syndromes (MDS) are a heterogeneous group of hematopoietic clonal disorders. So, prognostic variables are important to separate patients with a similar biology and clinical outcome. We compared the importance of risk stratification in primary MDS of IPSS and WPSS with the just described revision of IPSS (IPSS-R), and examined if variables obtained by bone marrow immunophenotyping could add prognostic information to any of the scores. Methods In this prospective study of 101 cases of primary MDS we compared the relation of patients’ overall survival with WHO types, IPSS, IPSS-R, WPSS and phenotypic abnormalities of hematopoietic precursors. We examined aberrancies in myelomonocytic precursors and CD34+ cells. Patients were censored when receiving chemotherapy or BM transplantation. Survival analysis was made by Cox regressions and stability of the models was examined by bootstrap resampling. Results median age: 64 years (15–93). WHO types: 2 cases of 5q- syndrome, 7 of RA, 64 of RCDM and 28 of RAEB. In the univariate Cox analysis, increasing risk category of all scores, degree of anemia, higher percentage of BM blasts, higher number of CD34+ cells and their myeloid fractions besides increasing number of phenotypic abnormalities detected were significantly associated with a shorter survival. In the multivariate analysis comparing the three scores, IPSS-R was the only independent risk factor. Comparing WPSS with phenotypic variables (CD34+/CD13+ cells, CD34+/CD13− cells and “total alterations”) the score and “CD34+/CD13+ cells” remained in the model. When IPSS was tested together with these phenotypic variables, only “CD34+/CD13+ cells”, and “total alterations” remained in the model. Testing IPSS-R with the phenotypic variables studied, only the score and “CD34+/CD13+ cells” entered the model. Conclusions Immunophenotypic analysis of myelomonocytic progenitors provides additional prognostic information to all clinical scores studied. IPSS-R improved risk stratification in MDS compared to the former scores.
Collapse
|