1
|
Jia ZY, Abulimiti M, Wu Y, Ma LN, Li XY, Wang J. A novel perspective on survival prediction for AML patients: Integration of machine learning in SEER database applications. Heliyon 2025; 11:e42030. [PMID: 39911442 PMCID: PMC11795080 DOI: 10.1016/j.heliyon.2025.e42030] [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: 03/13/2024] [Revised: 07/30/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Objective The purpose of this study is to explore the epidemiological characteristics of acute myeloid leukemia (AML) and establish a more accurate model for predicting the prognosis of AML patients based on machine learning. Methods We obtained clinical data of a total of 87,090 AML patients between 1975 and 2019 from the SEER database. First, we used Kaplan-Meier analysis to examine the prognosis of patients in different strata. Then, we discussed the independent factors that influenced the overall survival (OS) of AML patients, using univariate and multivariate Cox regression analysis. Finally, we used 11 machine learning algorithms to predict the survival rate of AML patients at 1, 2, and 3 years, respectively. We also used five-fold cross-validation with 20 cycles to obtain the optimal parameters for each model, in order to improve the accuracy of predictions. Results The Kaplan-Meier analysis showed that the survival rate of patients diagnosed after 2010 was significantly higher than that of those diagnosed before. In addition, older age, male gender, and non-black race were associated with poor prognosis. Among the FAB subtypes, M3 AML had a better prognosis than other subtypes, and among the WHO subtypes, AML associated with Down syndrome had the best prognosis, followed by AML with eosinophilic abnormalities. The Cox regression analysis demonstrated that gender, age, race, and family income were significantly related to the survival of AML patients. Among the 11 machine learning models, the random forest classifier performed best on multiple evaluation metrics in predicting survival at 1, 2, and 3 years. In addition, both the XGBoost classifier and the neural network classifier showed high accuracy and reliability at each prediction stage. Conclusion Through in-depth analysis, this study provides a deeper understanding of the epidemiological characteristics of AML and successfully establishes a prediction model based on machine learning, which demonstrates good accuracy and reliability in predicting the prognosis of AML patients.
Collapse
Affiliation(s)
- Zheng-yi Jia
- School of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | | | - Yun Wu
- Department of General Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Li-na Ma
- School of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Xiao-yu Li
- School of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Jie Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| |
Collapse
|
2
|
Zhao Z, Wang D, Sheng X, Li S, Liu T, Chang M, Feng L, Zhang D, Ji C, Lu F, Ye J. Identification and validation of BATF as a prognostic biomarker and regulator of immune cell infiltration in acute myeloid leukemia. Front Immunol 2025; 15:1429855. [PMID: 39872530 PMCID: PMC11769954 DOI: 10.3389/fimmu.2024.1429855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Background Basic leucine zipper ATF-like transcription factor (BATF) is a nuclear basic leucine zipper protein affiliated with the AP-1/ATF superfamily. Previous research has confirmed that BATF expression plays a significant role in the tumour microenvironment. However, the associations between BATF expression and prognoses in acute myeloid leukaemia (AML) patients and their immunological effects remain unclear. Methods Genomic and clinical AML data were got from the TCGA (TCGA-LAML) and GEO (GSE37642) databases for the subsequent analysis. The expression levels of BATF in AML patients were assessed using GEPIA, and the results were verified by qRT-PCR and Western blotting. In the meantime, the prognostic value of BATF was evaluated using univariate and multivariate analyses, receiver operating characteristic (ROC) curve (AUC) analysis, and Kaplan-Meier (KM) survival analysis. EdU, colony formation, and CCK-8 assays were employed to evaluate the proliferation of cells. Moreover, we detected the association of BATF expression with drug sensitivity through database analysis and in vivo experiments. To further investigate the mechanism of action of BATF in AML, RNA sequencing (RNA-seq) analysis was performed, followed by pathway enrichment analysis using Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Meanwhile, we detected the connection between BATF expression and the proportions of immune cells via flow cytometry in C1498 mouse model of AML. Finally, we investigated the association between BATF expression and cell-cell communication within the AML cell population using single-cell sequencing. Results In this study, we thoroughly investigated the role of BATF in AML. First, we observed a significant elevation in the expression of BATF in patients and cells in AML. Further analysis revealed an association between high BATF expression and poor prognosis in AML. Additionally, BATF expression was found to promote the proliferative capacity of AML cells. Moreover, the results showed that the expression level of BATF dramatically affected the effect of chemotherapy in AML patients. We also discovered that BATF expression could activate multiple immune-related pathways, altering the proportions of CD8+T cells and NK cells, suggesting that BATF may be a regulator of immune cell infiltration. Finally, there were differences in receptor ligand pairs between AML cells with high and low expression of BATF and immune cells. Conclusion Bioinformatics analysis and experimental verification revealed that BATF expression could alter the proportions of CD8+T cells and NK cells in AML and affect drug sensitivity, making it a potential treatment target for AML.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- Humans
- Animals
- Prognosis
- Mice
- Biomarkers, Tumor/metabolism
- Tumor Microenvironment/immunology
- Female
- Male
- Cell Line, Tumor
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Gene Expression Regulation, Leukemic
- Cell Proliferation
- Middle Aged
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Dongmei Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xue Sheng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Shuying Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Tingting Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Mengyuan Chang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Feng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Di Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fei Lu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Jingjing Ye
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
3
|
Ajonu CI, Grundy RI, Ball GR, Zafeiris D. Application of a high-throughput swarm-based deep neural network Algorithm reveals SPAG5 downregulation as a potential therapeutic target in adult AML. Funct Integr Genomics 2025; 25:8. [PMID: 39762615 PMCID: PMC11703901 DOI: 10.1007/s10142-024-01514-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/22/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025]
Abstract
Gene‒gene interactions play pivotal roles in disease pathogenesis and are fundamental in the development of targeted therapeutics, particularly through the elucidation of oncogenic gene drivers in cancer. The systematic analysis of pathways and gene interactions is critical in the drug discovery process for various cancer subtypes. SPAG5, known for its role in spindle formation during cell division, has been identified as an oncogene in several cancers, although its specific impact on AML remains underexplored. This study leverages a high-throughput swarm-based deep neural network (SDNN) and transcriptomic data-an approach that enhances predictive accuracy and robustness through collective intelligence-to augment, model, and enhance the understanding of the TP53 pathway in AML cohorts. Our integrative systems biology approach identified SPAG5 as a uniquely downregulated driver in adult AML, underscoring its potential as a novel therapeutic target. The interaction of SPAG5 with key hub genes such as MDM2 and CDK1 not only reinforces its role in tumour suppression through negative regulation but also highlights its potential in moderating the phenotypic and genomic alterations associated with AML progression. This study of the role and interaction dynamics of SPAG5 sets the stage for future research aimed at developing targeted and personalized treatment approaches for AML, utilizing the capabilities of genetic interventions.
Collapse
Affiliation(s)
- Chinyere I Ajonu
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.
- Intelligent OMICS Limited, Nottingham, United Kingdom.
| | | | - Graham R Ball
- Intelligent OMICS Limited, Nottingham, United Kingdom
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | | |
Collapse
|
4
|
Fang J, Zhang J, Zhu L, Xin X, Hu H. The epigenetic role of EZH2 in acute myeloid leukemia. PeerJ 2024; 12:e18656. [PMID: 39655332 PMCID: PMC11627098 DOI: 10.7717/peerj.18656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/17/2024] [Indexed: 12/12/2024] Open
Abstract
Acute myeloid leukemia (AML), a malignant disease of the bone marrow, is characterized by the clonal expansion of myeloid progenitor cells and a block in differentiation. The high heterogeneity of AML significantly impedes the development of effective treatment strategies. Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the polycomb repressive complex 2 (PRC2), regulates the expression of downstream target genes through the trimethylation of lysine 27 on histone 3 (H3K27me3). Increasing evidence suggests that the dysregulation of EZH2 expression in various cancers is closely associated with tumorigenesis. In the review, we examine the role of EZH2 in AML, highlighting its crucial involvement in regulating stemness, proliferation, differentiation, immune response, drug resistance and recurrence. Furthermore, we summarize the application of EZH2 inhibitors in AML treatment and discuss their potential in combination with other therapeutic modalities. Therefore, targeting EZH2 may represent a novel and promising strategy for the treatment of AML.
Collapse
MESH Headings
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Epigenesis, Genetic
- Drug Resistance, Neoplasm/genetics
- Cell Proliferation/genetics
- Cell Proliferation/drug effects
- Cell Differentiation/genetics
Collapse
Affiliation(s)
- Jinyong Fang
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jingcheng Zhang
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Xiaoru Xin
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Huixian Hu
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| |
Collapse
|
5
|
Zhang H, Zhong L, Wang M, Wan P, Chu X, Chen S, Zhou Z, Shao X, Liu B. p110CUX1 promotes acute myeloid leukemia progression via regulating pyridoxal phosphatase expression and activating PI3K/AKT/mTOR signaling pathway. Mol Carcinog 2024; 63:2063-2077. [PMID: 38994801 DOI: 10.1002/mc.23793] [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: 03/05/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
As an evolutionarily conserved transcription factor, Cut-like homeobox 1 (CUX1) plays crucial roles in embryonic and nervous system development, cell differentiation, and DNA damage repair. One of its major isoforms, p110CUX1, exhibits stable DNA binding capabilities and contributes to the regulation of cell cycle progression, proliferation, migration, and invasion. While p110CUX1 has been implicated in the progression of various malignant tumors, its involvement in acute myeloid leukemia (AML) remains uncertain. This study aims to elucidate the role of p110CUX1 in AML. Our findings reveal heightened expression levels of both p110CUX1 and pyridoxal phosphatase (PDXP) in AML cell lines. Overexpression of p110CUX1 promotes AML cell proliferation while inhibiting apoptosis and differentiation, whereas knockdown of PDXP yields contrasting effects. Mechanistically, p110CUX1 appears to facilitate AML development by upregulating PDXP expression and activating the PI3K/AKT/mTOR signaling pathway. Animal experimental corroborate the pro-AML effect of p110CUX1. These results provide experimental evidence supporting the involvement of the p110CUX1-PDXP-PI3K/AKT/mTOR axis in AML progression. Hence, targeting p110CUX1 may hold promise as a therapeutic strategy for AML.
Collapse
Affiliation(s)
- Hongyan Zhang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Meng Wang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Peng Wan
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xuan Chu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Shuyu Chen
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Ziwei Zhou
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Shao
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Zuo Y, Li H, Wang X, Liang Y, Huang C, Nai G, Ruan J, Dong W, Lu X. Inhibition of JAK/STAT3 Expression by Acute Myeloid Leukemia-Targeted Nanoliposome for Chemotherapy Enhancement. ACS OMEGA 2024; 9:37901-37909. [PMID: 39281932 PMCID: PMC11391460 DOI: 10.1021/acsomega.4c00710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024]
Abstract
Acute myeloid leukemia (AML) is a relatively common malignant hematological disease whose development is mostly associated with abnormal activation of the JAK/STAT3 signaling pathway. Our previous study revealed that SAR317461, a novel JAK2/STAT3 inhibitor, can effectively inhibit the activation of the JAK2/STAT3 signaling pathway and has significant damaging and pro-apoptotic effects on AML cell lines. This project aims to build upon our prior research to enhance the application of SAR317461 in AML. The surface modification of liposomes with the CD34 antibody, along with the inclusion of the SAR317461 and cytarabine (a common AML chemotherapeutic agent), is observed. Due to the high expression of CD34 on the surface of AML cells, the nanoliposome could target AML cells specifically, further achieving an effective treatment for AML through the synergistic effect of JAK2/STAT3 inhibitors and chemotherapeutic agents. The implementation of this project will provide more theoretical support and ideas for the clinical application of JAK/STAT3 inhibitors in malignant tumors and for overcoming chemotherapy resistance.
Collapse
Affiliation(s)
- Yao Zuo
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Hongwen Li
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Xiaochao Wang
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Yejin Liang
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Caihong Huang
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Guanye Nai
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Jingrong Ruan
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Wenzheng Dong
- Department of Hematology & Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi 533000, P. R. China
| | - Xiang Lu
- Department of Hematology, The First People's Hospital of Nanning, Guangxi 530022, P. R. China
- The Fifth Affiliated Hospital of Guangxi Medical University, Guangxi 537406, P. R. China
| |
Collapse
|
7
|
Zhao P, Xia L, Chen D, Xu W, Guo H, Xu Y, Yan B, Wu X, Li Y, Zhang Y, Zhang X. METTL1 mediated tRNA m 7G modification promotes leukaemogenesis of AML via tRNA regulated translational control. Exp Hematol Oncol 2024; 13:8. [PMID: 38268051 PMCID: PMC10807064 DOI: 10.1186/s40164-024-00477-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND RNA modifications have been proven to play fundamental roles in regulating cellular biology process. Recently, maladjusted N7-methylguanosine (m7G) modification and its modifiers METTL1/WDR4 have been confirmed an oncogene role in multiple cancers. However, the functions and molecular mechanisms of METTL1/WDR4 in acute myeloid leukemia (AML) remain to be determined. METHODS METTL1/WDR4 expression levels were quantified using qRT-PCR, western blot analysis on AML clinical samples, and bioinformatics analysis on publicly available AML datasets. CCK-8 assays and cell count assays were performed to determine cell proliferation. Flow cytometry assays were conducted to assess cell cycle and apoptosis rates. Multiple techniques were used for mechanism studies in vitro assays, such as northern blotting, liquid chromatography-coupled mass spectrometry (LC-MS/MS), tRNA stability analysis, transcriptome sequencing, small non-coding RNA sequencing, quantitative proteomics, and protein synthesis measurements. RESULTS METTL1/WDR4 are significantly elevated in AML patients and associated with poor prognosis. METTL1 knockdown resulted in reduced cell proliferation and increased apoptosis in AML cells. Mechanically, METTL1 knockdown leads to significant decrease of m7G modification abundance on tRNA, which further destabilizes tRNAs and facilitates the biogenesis of tsRNAs in AML cells. In addition, profiling of nascent proteins revealed that METTL1 knockdown and transfection of total tRNAs that were isolated from METTL1 knockdown AML cells decreased global translation efficiency in AML cells. CONCLUSIONS Taken together, our study demonstrates the important role of METTL1/WDR4 in AML leukaemogenesis, which provides a promising target candidate for AML therapy.
Collapse
Affiliation(s)
- Pan Zhao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Lin Xia
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Dan Chen
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Wei Xu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Huanping Guo
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Yinying Xu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Bingbing Yan
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Xiao Wu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Yuxia Li
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Yunfang Zhang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400037, China.
| |
Collapse
|
8
|
Peng CJ, Fan Z, Luo JS, Wang LN, Li Y, Liang C, Zhang XL, Luo XQ, Huang LB, Tang YL. The Potential Transcriptomic and Metabolomic Mechanisms of ATO and ATRA in Treatment of FLT3-ITD Acute Myeloid Leukemia. Technol Cancer Res Treat 2024; 23:15330338231223080. [PMID: 38179723 PMCID: PMC10771057 DOI: 10.1177/15330338231223080] [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] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) with Fms-like tyrosine kinase 3 gene internal tandem duplication (FLT3-ITD) mutations has a poor prognosis. The combination of arsenic trioxide (ATO) and all-trans retinoic acid (ATRA) has a synergistic killing effect on leukemia cells with FLT3-ITD mutation. However, the mechanism, especially the changes of gene expression and metabolic activity remain unclear. Here we explore the transcriptome and metabolomics changes of FLT3-ITD AML cells treated with ATO/ATRA. METHODS RNA-seq was used to identify differential expressed genes (DEGs), and ultra-high performance liquid chromatography-quadrupole electrostatic field orbital trap mass spectrometry (UHPLC-QE-MS) nontargeted metabolomics method was used to screen out the differential metabolites in FLT3-ITD mutant cell lines treated with ATRA and ATO. KEGG pathway database was utilized for pathway exploration and Seahorse XF24 was used to detect extracellular acidification rate (ECAR). Metabolic polymerase chain reaction (PCR) array and real-time quantitative PCR (RT-qPCR) were used to detect mRNA levels of key metabolic genes of glycolysis and fatty acid after drug treatment. RESULTS A total of 3873 DEGs were identified and enriched in 281 Gene Ontology (GO) terms, among which 210 were related to biological processes, 43 were related to cellular components, and 28 were related to molecular functions. Besides, 1794 and 927 differential metabolites were screened in positive and negative ion mode separately, and 59 different metabolic pathways were involved, including alanine-aspartate-glutamate metabolic pathway, arginine, and proline metabolic pathway, glycerophospholipid metabolic pathways, etc. According to KEGG Pathway analysis of transcriptome combined with metabolome, glycolysis/gluconeogenesis pathway and fatty acid metabolism pathway were significantly founded enriched. ATRA + ATO may inhibit the glycolysis of FLT3-ITD AML cells by inhibiting FLT3 and its downstream AKT/HK2-VDAC1 signaling pathway. CONCLUSIONS The gene transcription profile and metabolites of FLT3-ITD mutant cells changes significantly after treatment, which might be related to the anti-FLT3-ITD AML effect. The screened DEGs, differential metabolites pathway are helpful in studying the mechanism of anti-leukemia effects and drug targets.
Collapse
Affiliation(s)
- Chun-Jin Peng
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Fan
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie-Si Luo
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Na Wang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Li
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cong Liang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Li Zhang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xue-Qun Luo
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Bin Huang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Lai Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Farzaneh M, Najafi S, Sheykhi-Sabzehpoush M, Nezhad Dehbashi F, Anbiyaee O, Nasrolahi A, Azizidoost S. The stem cell-specific long non-coding RNAs in leukemia. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:345-351. [PMID: 36168086 DOI: 10.1007/s12094-022-02952-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/09/2022] [Indexed: 01/27/2023]
Abstract
Leukemia is defined as a heterogeneous group of hematological cancers whose prevalence is on the rise worldwide. Despite the large body of studies, the etiology of leukemia has not been fully elucidated. Leukemia stem cells (LSCs) are a subpopulation of cancer cells that sustain the growth of the leukemic clone and are the main culprit for the maintenance of the neoplasm. In contrast to most leukemia cells, LSCs are resistant to chemo- and radiotherapy. Several recent studies demonstrated the altered expression profile of long non-coding RNAs (lncRNAs) in LSCs and shed light on the role of lncRNAs in the survival, proliferation, and differentiation of LSCs. LncRNAs are transcripts longer than 200 nucleotides that are implicated in several cellular and molecular processes such as gene expression, apoptosis, and carcinogenesis. Likewise, lncRNAs have shown a prognostic marker in leukemia patients and represent novel treatment options. Herein, we review the current knowledge concerning lncRNAs' implication in the pathogenesis of LSCs and discuss their prognostic, diagnostic, and therapeutic potential.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Fereshteh Nezhad Dehbashi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, School of Medicine, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ava Nasrolahi
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
10
|
Zhai J, Gu X, Liu Y, Hu Y, Jiang Y, Zhang Z. Chemotherapeutic and targeted drugs-induced immunogenic cell death in cancer models and antitumor therapy: An update review. Front Pharmacol 2023; 14:1152934. [PMID: 37153795 PMCID: PMC10160433 DOI: 10.3389/fphar.2023.1152934] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/04/2023] [Indexed: 05/10/2023] Open
Abstract
As traditional strategies for cancer treatment, some chemotherapy agents, such as doxorubicin, oxaliplatin, cyclophosphamide, bortezomib, and paclitaxel exert their anti-tumor effects by inducing immunogenic cell death (ICD) of tumor cells. ICD induces anti-tumor immunity through release of, or exposure to, damage-related molecular patterns (DAMPs), including high mobility group box 1 (HMGB1), calreticulin, adenosine triphosphate, and heat shock proteins. This leads to activation of tumor-specific immune responses, which can act in combination with the direct killing functions of chemotherapy drugs on cancer cells to further improve their curative effects. In this review, we highlight the molecular mechanisms underlying ICD, including those of several chemotherapeutic drugs in inducing DAMPs exposed during ICD to activate the immune system, as well as discussing the prospects for application and potential role of ICD in cancer immunotherapy, with the aim of providing valuable inspiration for future development of chemoimmunotherapy.
Collapse
|
11
|
Low JY, Ko M, Hanratty B, Patel RA, Bhamidipati A, Heaphy CM, Sayar E, Lee JK, Li S, De Marzo AM, Nelson WG, Gupta A, Yegnasubramanian S, Ha G, Epstein JI, Haffner MC. Genomic Characterization of Prostatic Basal Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:4-10. [PMID: 36309102 PMCID: PMC9768679 DOI: 10.1016/j.ajpath.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Basal cell carcinoma (BCC) of the prostate is a rare tumor. Compared with the more common acinar adenocarcinoma (AAC) of the prostate, BCCs show features of basal cell differentiation and are thought to be biologically distinct from AAC. The spectrum of molecular alterations of BCC has not been comprehensively described, and genomic studies are lacking. Herein, whole genome sequencing was performed on archival formalin-fixed, paraffin-embedded specimens of two cases with BCC. Prostatic BCCs were characterized by an overall low copy number and mutational burden. Recurrent copy number loss of chromosome 16 was observed. In addition, putative driver gene alterations in KIT, DENND3, PTPRU, MGA, and CYLD were identified. Mechanistically, depletion of the CYLD protein resulted in increased proliferation of prostatic basal cells in vitro. Collectively, these studies show that prostatic BCC displays distinct genomic alterations from AAC and highlight a potential role for loss of chromosome 16 in the pathogenesis of this rare tumor type.
Collapse
Affiliation(s)
- Jin-Yih Low
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Minjeong Ko
- Division of Public Health Science, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Brian Hanratty
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Radhika A Patel
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Akshay Bhamidipati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher M Heaphy
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Erolcan Sayar
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - John K Lee
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington; Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Shan Li
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Angelo M De Marzo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William G Nelson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anuj Gupta
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gavin Ha
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington; Division of Public Health Science, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jonathan I Epstein
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Michael C Haffner
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington.
| |
Collapse
|
12
|
Chen Y, Wang J, Zhang F, Liu P. A perspective of immunotherapy for acute myeloid leukemia: Current advances and challenges. Front Pharmacol 2023; 14:1151032. [PMID: 37153761 PMCID: PMC10154606 DOI: 10.3389/fphar.2023.1151032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 05/10/2023] Open
Abstract
During the last decade, the underlying pathogenic mechanisms of acute myeloid leukemia (AML) have been the subject of extensive study which has considerably increased our understanding of the disease. However, both resistance to chemotherapy and disease relapse remain the principal obstacles to successful treatment. Because of acute and chronic undesirable effects frequently associated with conventional cytotoxic chemotherapy, consolidation chemotherapy is not feasible, especially for elderly patients, which has attracted a growing body of research to attempt to tackle this problem. Immunotherapies for acute myeloid leukemia, including immune checkpoint inhibitors, monoclonal antibodies, dendritic cell (DC) vaccines, together with T-cell therapy based on engineered antigen receptor have been developed recently. Our review presents the recent progress in immunotherapy for the treatment of AML and discusses effective therapies that have the most potential and major challenges.
Collapse
Affiliation(s)
- Ying Chen
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
- *Correspondence: Jishi Wang,
| | - Fengqi Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, Guiyang, China
| |
Collapse
|
13
|
Qi Y, Wang M, Jiang Q. PABPC1--mRNA stability, protein translation and tumorigenesis. Front Oncol 2022; 12:1025291. [PMID: 36531055 PMCID: PMC9753129 DOI: 10.3389/fonc.2022.1025291] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/08/2022] [Indexed: 09/29/2023] Open
Abstract
Mammalian poly A-binding proteins (PABPs) are highly conserved multifunctional RNA-binding proteins primarily involved in the regulation of mRNA translation and stability, of which PABPC1 is considered a central regulator of cytoplasmic mRNA homing and is involved in a wide range of physiological and pathological processes by regulating almost every aspect of RNA metabolism. Alterations in its expression and function disrupt intra-tissue homeostasis and contribute to the development of various tumors. There is increasing evidence that PABPC1 is aberrantly expressed in a variety of tumor tissues and cancers such as lung, gastric, breast, liver, and esophageal cancers, and PABPC1 might be used as a potential biomarker for tumor diagnosis, treatment, and clinical application in the future. In this paper, we review the abnormal expression, functional role, and molecular mechanism of PABPC1 in tumorigenesis and provide directions for further understanding the regulatory role of PABPC1 in tumor cells.
Collapse
Affiliation(s)
- Ya Qi
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University, Shenyang, Liaoning, China
| | - Min Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University, Shenyang, Liaoning, China
| | - Qi Jiang
- Second Department of Clinical Medicine, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
14
|
Aqil B, Gao J, Rahn HR, Sukhanova M, Lu X, Altman J, Jennings L, Chen Q, Chen YH. Non-leukemic presentation of acute promyelocytic leukemia as a testicular mass with associated non-canonical FLT3 mutation. Leuk Lymphoma 2022; 63:3493-3496. [PMID: 36215147 DOI: 10.1080/10428194.2022.2131425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Barina Aqil
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Juehua Gao
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Heidi R Rahn
- Winfield Pathology Consultants, Northwestern Medicine, Winfield, IL, USA
| | - Madina Sukhanova
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Xinyan Lu
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Jessica Altman
- Department of Medicine, Hematology Oncology Division, Feinberg School of Medicine, Chicago, IL, USA
| | - Lawrence Jennings
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Qing Chen
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Yi-Hua Chen
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
15
|
Lee C, Lee S, Park E, Hong J, Shin DY, Byun JM, Yun H, Koh Y, Yoon SS. Transcriptional signatures of the BCL2 family for individualized acute myeloid leukaemia treatment. Genome Med 2022; 14:111. [PMID: 36171613 PMCID: PMC9520894 DOI: 10.1186/s13073-022-01115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although anti-apoptotic proteins of the B-cell lymphoma-2 (BCL2) family have been utilized as therapeutic targets in acute myeloid leukaemia (AML), their complicated regulatory networks make individualized therapy difficult. This study aimed to discover the transcriptional signatures of BCL2 family genes that reflect regulatory dynamics, which can guide individualized therapeutic strategies. METHODS From three AML RNA-seq cohorts (BeatAML, LeuceGene, and TCGA; n = 451, 437, and 179, respectively), we constructed the BCL2 family signatures (BFSigs) by applying an innovative gene-set selection method reflecting biological knowledge followed by non-negative matrix factorization (NMF). To demonstrate the significance of the BFSigs, we conducted modelling to predict response to BCL2 family inhibitors, clustering, and functional enrichment analysis. Cross-platform validity of BFSigs was also confirmed using NanoString technology in a separate cohort of 47 patients. RESULTS We established BFSigs labeled as the BCL2, MCL1/BCL2, and BFL1/MCL1 signatures that identify key anti-apoptotic proteins. Unsupervised clustering based on BFSig information consistently classified AML patients into three robust subtypes across different AML cohorts, implying the existence of biological entities revealed by the BFSig approach. Interestingly, each subtype has distinct enrichment patterns of major cancer pathways, including MAPK and mTORC1, which propose subtype-specific combination treatment with apoptosis modulating drugs. The BFSig-based classifier also predicted response to venetoclax with remarkable performance (area under the ROC curve, AUROC = 0.874), which was well-validated in an independent cohort (AUROC = 0.950). Lastly, we successfully confirmed the validity of BFSigs using NanoString technology. CONCLUSIONS This study proposes BFSigs as a biomarker for the effective selection of apoptosis targeting treatments and cancer pathways to co-target in AML.
Collapse
Affiliation(s)
- Chansub Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sungyoung Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Center for Precision Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Eunchae Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junshik Hong
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Yeop Shin
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ja Min Byun
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Center for Precision Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Youngil Koh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Mao Y, Xu J, Xu X, Qiu J, Hu Z, Jiang F, Zhou G. Comprehensive analysis for cellular senescence-related immunogenic characteristics and immunotherapy prediction of acute myeloid leukemia. Front Pharmacol 2022; 13:987398. [PMID: 36225590 PMCID: PMC9548549 DOI: 10.3389/fphar.2022.987398] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/23/2022] [Indexed: 01/10/2023] Open
Abstract
In malignancies, cellular senescence is critical for carcinogenesis, development, and immunological regulation. Patients with acute myeloid leukemia (AML) have not investigated a reliable cellular senescence-associated profile and its significance in outcomes and therapeutic response. Cellular senescence-related genes were acquired from the CellAge database, while AML data were obtained from the GEO and TCGA databases. The TCGA-AML group served as a training set to construct a prognostic risk score signature, while the GSE71014 set was used as a testing set to validate the accuracy of the signature. Through exploring the expression profiles of cellular senescence-related genes (SRGs) in AML patients, we used Lasso and Cox regression analysis to establish the SRG-based signature (SRGS), which was validated as an independent prognostic predictor for AML patients via clinical correlation. Survival analysis showed that AML patients in the low-risk score group had a longer survival time. Tumor immune infiltration and functional enrichment analysis demonstrated that AML patients with low-risk scores had higher immune infiltration and active immune-related pathways. Meanwhile, drug sensitivity analysis and the TIDE algorithm showed that the low-risk score group was more susceptible to chemotherapy and immunotherapy. Cell line analysis in vitro further confirmed that the SRGs in the proposed signature played roles in the susceptibility to cytarabine and YM155. Our results indicated that SRGS, which regulates the immunological microenvironment, is a reliable predictor of the clinical outcome and immunotherapeutic response in AML.
Collapse
Affiliation(s)
- Yan Mao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinwen Xu
- Department of Pediatric Nephrology, Wuxi Children’s Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Xuejiao Xu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayun Qiu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengyun Hu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Pediatrics, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- *Correspondence: Guoping Zhou, ; Feng Jiang,
| | - Guoping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Guoping Zhou, ; Feng Jiang,
| |
Collapse
|
17
|
Huang W, Sun G, Wang Q, Long Z. The research progress of targeted therapy in acute myeloid leukemia based on bibliometric analysis. Front Oncol 2022; 12:957370. [PMID: 36119476 PMCID: PMC9481238 DOI: 10.3389/fonc.2022.957370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Wanxue Huang
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
| | - Gongrui Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qi Wang
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
| | - Zhiguo Long
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
- *Correspondence: Zhiguo Long,
| |
Collapse
|
18
|
Rong D, Chen X, Xiao J, Liu D, Ni X, Tong X, Wang H. Histone methylation modification patterns and relevant M-RiskScore in acute myeloid leukemia. Heliyon 2022; 8:e10610. [PMID: 36164519 PMCID: PMC9508520 DOI: 10.1016/j.heliyon.2022.e10610] [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: 02/23/2022] [Revised: 07/13/2022] [Accepted: 09/07/2022] [Indexed: 12/05/2022] Open
Abstract
Objective We tried to identify novel molecular subtypes of acute myeloid leukemia (AML) associated with histone methylation and established a relevant scoring system to predict treatment response and prognosis of AML. Methods Gene expression data and clinical characteristics of patients with AML were obtained from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. Molecular subtyping was carried out by consensus clustering analysis, based on the expression of 24 histone methylation modification regulators (HMMRs). The clinical and biological features of each clustered pattern were taken into account. The scoring system was constructed by using differential expression analysis, Cox regression method and lasso regression analysis. Subsequently, the scoring system in the roles of prognostic and chemotherapeutic prediction of AML were explored. Finally, an independent GSE dataset was used for validating the established clustering system. Results Two distinct subtypes of AML were identified based on the expression of the 24 HMMRs, which exhibited remarkable differences in several clinical and biological characteristics, including HMMRs expression, AML-M0 distribution, NPM1 mutation, tumor mutation burden, somatic mutations, pathway activation, immune cell infiltration and patient survival. The scoring system, M-RiskScore, was established. Integrated analysis demonstrated that patients with the low M-RiskScore displayed a prominent survival advantage and a good response to decitabine treatment, while patients with high M-RiskScore have resistance to decitabine, but they could benefit from IA regimen therapy. Conclusion Detection of HMMRs expression would be a potential strategy for AML subtyping. Meanwhile, targeting histone methylation would be a preferred strategy for either AML-M0 or NPM1 mutant patients. M-RiskScore was a useful prognostic biomarker and a guide for the choice of appropriate chemotherapy strategy.
Collapse
Affiliation(s)
- Dade Rong
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Xiaomin Chen
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,GenePlus, Beijing, China
| | - Jing Xiao
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Department of Clinical Laboratory, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, China
| | - Daiyuan Liu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiangna Ni
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiuzhen Tong
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| |
Collapse
|
19
|
Zhou T, Qian K, Li YY, Cai WK, Yin SJ, Wang P, He GH. The pyroptosis-related gene signature predicts prognosis and reveals characterization of the tumor immune microenvironment in acute myeloid leukemia. Front Pharmacol 2022; 13:951480. [PMID: 36034801 PMCID: PMC9399441 DOI: 10.3389/fphar.2022.951480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Pyroptosis is a novel inflammatory form of programmed cell death and a prospective target for cancer therapy. Nevertheless, little is known about the association between pyroptosis-related genes (PRGs) and acute myeloid leukemia (AML) prognosis. Herein, we systematically investigated the specific functions and clinical prognostic value of multiple PRGs in AML. Methods: Univariate and LASSO Cox regression analyses based on TCGA and GTEx databases were used to generate the PRG signature, whose predictive efficacy of survival was evaluated using survival analysis, ROC, univariate and multivariate Cox analyses as well as subgroup analysis. The BeatAML cohort was used for data validation. The association between risk score and immune cell infiltration, HLA, immune checkpoints, cancer stem cell (CSC), tumor mutation burden (TMB), and therapeutic drug sensitivity were also analyzed. Results: Six -PRG signatures, namely, CASP3, ELANE, GSDMA, NOD1, PYCARD, and VDR were generated. The high-risk score represented a poorer prognosis and the PRG risk score was also validated as an independent predictor of prognosis. A nomogram including the cytogenetic risk, age, and risk score was constructed for accurate prediction of 1-, 3-, and 5-year survival probabilities. Meanwhile, this risk score was significantly associated with the tumor immune microenvironment (TIME). A high-risk score is characterized by high immune cell infiltration, HLA, and immune checkpoints, as well as low CSC and TMB. In addition, patients with low-risk scores presented significantly lower IC50 values for ATRA, cytarabine, midostaurin, doxorubicin, and etoposide. Conclusion: Our findings might contribute to further understanding of PRGs in the prognosis and development of AML and provide novel and reliable biomarkers for its precise prevention and treatment.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People’s Liberation Army, Kunming, China
- College of Pharmacy, Dali University, Dali, China
| | - Kai Qian
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People’s Liberation Army, Kunming, China
- College of Pharmacy, Dali University, Dali, China
| | - Yun-Yun Li
- Department of Pharmacy, The Second People’s Hospital of Quzhou Zhejiang, Quzhou, China
| | - Wen-Ke Cai
- Department of Cardiothoracic Surgery, 920th Hospital of Joint Logistics Support Force of People’s Liberation Army, Kunming, China
| | - Sun-Jun Yin
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People’s Liberation Army, Kunming, China
| | - Ping Wang
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People’s Liberation Army, Kunming, China
| | - Gong-Hao He
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People’s Liberation Army, Kunming, China
- Research Center of Clinical Pharmacology, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
- *Correspondence: Gong-Hao He,
| |
Collapse
|
20
|
Tomaz V, Griesi-Oliveira K, Puga RD, Conti BJ, Santos FPS, Hamerschlak N, Campregher PV. Molecular Characterization of a First-in-Human Clinical Response to Nimesulide in Acute Myeloid Leukemia. Front Oncol 2022; 12:874168. [PMID: 35756679 PMCID: PMC9215211 DOI: 10.3389/fonc.2022.874168] [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: 02/11/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy associated with high morbidity and mortality. Here we describe a case of a patient with AML who presented a partial response after utilization of the non-steroidal anti-inflammatory drug nimesulide. The response was characterized by complete clearance of peripheral blood blasts and an 82% decrease of bone marrow blasts associated with myeloblast differentiation. We have then shown that nimesulide induces in vitro cell death and cell cycle arrest in all AML cell lines (HL-60, THP-1, OCI-AML2, and OCI-AML3). Weighted Correlation Network Analysis (WGCNA) of serial whole-transcriptome data of cell lines treated with nimesulide revealed that the sets of genes upregulated after treatment with nimesulide were enriched for genes associated with autophagy and apoptosis, and on the other hand, the sets of downregulated genes were associated with cell cycle and RNA splicing. Serial transcriptome of bone marrow patient sample confirmed the upregulation of genes associated with autophagy after the response to nimesulide. Lastly, we demonstrated that nimesulide potentiates the cytotoxic in vitro effect of several Food and Drug Administration (FDA)-approved chemotherapy drugs used in AML, including cytarabine.
Collapse
Affiliation(s)
- Victória Tomaz
- Experimental Research Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Renato D Puga
- Medicina Personalizada, Grupo Pardini, São Paulo, Brazil
| | - Bruno J Conti
- Experimental Research Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Fabio P S Santos
- Centro de Hematologia e Oncologia Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Nelson Hamerschlak
- Centro de Hematologia e Oncologia Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Paulo V Campregher
- Centro de Hematologia e Oncologia Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
21
|
Song Y, Tian S, Zhang P, Zhang N, Shen Y, Deng J. Construction and Validation of a Novel Ferroptosis-Related Prognostic Model for Acute Myeloid Leukemia. Front Genet 2022; 12:708699. [PMID: 35111195 PMCID: PMC8803125 DOI: 10.3389/fgene.2021.708699] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/28/2021] [Indexed: 12/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal malignant proliferative blood disorder with a poor prognosis. Ferroptosis, a novel form of programmed cell death, holds great promise for oncology treatment, and has been demonstrated to interfere with the development of various diseases. A range of genes are involved in regulating ferroptosis and can serve as markers of it. Nevertheless, the prognostic significance of these genes in AML remains poorly understood. Transcriptomic and clinical data for AML patients were acquired from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). Univariate Cox analysis was performed to identify ferroptosis-related genes with prognostic value, and the least absolute shrinkage and selection operator (LASSO) algorithm and stepwise multivariate Cox regression analysis were utilized to optimize gene selection from the TCGA cohort (132 samples) for model construction. Tumor samples from the GEO database (136 samples and 104 samples) were used as validation groups to estimate the predictive performance of the risk model. Finally, an eight-gene prognostic signature (including CHAC1, CISD1, DPP4, GPX4, AIFM2, SQLE, PGD, and ACSF2) was identified for the prediction of survival probability and was used to stratify AML patients into high- and low-risk groups. Survival analysis illustrated significantly prolonged overall survival and lower mortality in the low-risk group. The area under the receiver operating characteristic curve demonstrated good results for the training set (1-year: 0.846, 2-years: 0.826, and 3-years: 0.837), which verified the accuracy of the model for predicting patient survival. Independent prognostic analysis indicated that the model could be used as a prognostic factor (p ≤ 0.001). Functional enrichment analyses revealed underlying mechanisms and notable differences in the immune status of the two risk groups. In brief, we conducted and validated a novel ferroptosis-related prognostic model for outcome prediction and risk stratification in AML, with great potential to guide individualized treatment strategies in the future.
Collapse
Affiliation(s)
- Ying Song
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shufang Tian
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Zhang
- Hematology Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Zhang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Shen
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianchuan Deng
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Jianchuan Deng,
| |
Collapse
|
22
|
Huang R, Liao X, Li Q. Integrative genomic analysis of a novel small nucleolar RNAs prognostic signature in patients with acute myelocytic leukemia. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:2424-2452. [PMID: 35240791 DOI: 10.3934/mbe.2022112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
This study mainly used The Cancer Genome Atlas (TCGA) RNA sequencing dataset to screen prognostic snoRNAs of acute myeloid leukemia (AML), and used for the construction of prognostic snoRNAs signature for AML. A total of 130 AML patients with RNA sequencing dataset were used for prognostic snoRNAs screenning. SnoRNAs co-expressed genes and differentially expressed genes (DEGs) were used for functional annotation, as well as gene set enrichment analysis (GSEA). Connectivity Map (CMap) also used for potential targeted drugs screening. Through genome-wide screening, we identified 30 snoRNAs that were significantly associated with the prognosis of AML. Then we used the step function to screen a prognostic signature composed of 14 snoRNAs (SNORD72, SNORD38, U3, SNORA73B, SNORD79, SNORA73, SNORD12B, SNORA74, SNORD116-12, SNORA65, SNORA14, snoU13, SNORA75, SNORA31), which can significantly divide AML patients into high- and low-risk groups. Through GSEA, snoRNAs co-expressed genes and DEGs functional enrichment analysis, we screened a large number of potential functional mechanisms of this prognostic signature in AML, such as phosphatidylinositol 3-kinase-Akt, Wnt, epithelial to mesenchymal transition, T cell receptors, NF-kappa B, mTOR and other classic cancer-related signaling pathways. In the subsequent targeted drug screening using CMap, we also identified six drugs that can be used for AML targeted therapy, they were alimemazine, MG-262, fluoxetine, quipazine, naltrexone and oxybenzone. In conclusion, our current study was constructed an AML prognostic signature based on the 14 prognostic snoRNAs, which may serve as a novel prognostic biomarker for AML.
Collapse
Affiliation(s)
- Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Qiaochuan Li
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
23
|
Xiao Y, Chen J, Wang J, Guan W, Wang M, Zhang L, Wang Z, Wang L, Yu L. Acute Myeloid Leukemia Epigenetic Immune Escape From Nature Killer Cells by ICAM-1. Front Oncol 2021; 11:751834. [PMID: 34722306 PMCID: PMC8548470 DOI: 10.3389/fonc.2021.751834] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 01/05/2023] Open
Abstract
Acute myeloid leukemia (AML), a malignant disorder of hemopoietic stem cells. AML can escape immunosurveillance of natural killer (NK) by gene mutation, fusions, and epigenetic modification, while the mechanism is not clearly understood. Here we show that the expression of Intercellular adhesion molecule‐1 (ICAM‐1, CD54) is silenced in AML cells. Decitabine could upregulate ICAM-1 expression, which contributes to the NK-AML cell conjugates and helps NK cells kill AML cells. We also show that ICAM-1 high expression can reverse the AML immune evasion and activate NK cells function in vivo. This study suggests that a combination of the hypomethylating agent and NK cell infusion could be a new strategy to cure AML.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Jinghong Chen
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Jia Wang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Wei Guan
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Mengzhen Wang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Linlin Zhang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Zhiding Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lixin Wang
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Li Yu
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China.,Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
24
|
Peng M, Ren J, Jing Y, Jiang X, Xiao Q, Huang J, Tao Y, Lei L, Wang X, Yang Z, Yang Z, Zhan Q, Lin C, Jin G, Zhang X, Zhang L. Tumour-derived small extracellular vesicles suppress CD8+ T cell immune function by inhibiting SLC6A8-mediated creatine import in NPM1-mutated acute myeloid leukaemia. J Extracell Vesicles 2021; 10:e12168. [PMID: 34807526 PMCID: PMC8607980 DOI: 10.1002/jev2.12168] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/11/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukaemia (AML) carrying nucleophosmin (NPM1) mutations has been defined as a distinct entity of acute leukaemia. Despite remarkable improvements in diagnosis and treatment, the long-term outcomes for this entity remain unsatisfactory. Emerging evidence suggests that leukaemia, similar to other malignant diseases, employs various mechanisms to evade killing by immune cells. However, the mechanism of immune escape in NPM1-mutated AML remains unknown. In this study, both serum and leukemic cells from patients with NPM1-mutated AML impaired the immune function of CD8+ T cells in a co-culture system. Mechanistically, leukemic cells secreted miR-19a-3p into the tumour microenvironment (TME) via small extracellular vesicles (sEVs), which was controlled by the NPM1-mutated protein/CCCTC-binding factor (CTCF)/poly (A)-binding protein cytoplasmic 1 (PABPC1) signalling axis. sEV-related miR-19a-3p was internalized by CD8+ T cells and directly repressed the expression of solute-carrier family 6 member 8 (SLC6A8; a creatine-specific transporter) to inhibit creatine import. Decreased creatine levels can reduce ATP production and impair CD8+ T cell immune function, leading to immune escape by leukemic cells. In summary, leukemic cell-derived sEV-related miR-19a-3p confers immunosuppression to CD8+ T cells by targeting SLC6A8-mediated creatine import, indicating that sEV-related miR-19a-3p might be a promising therapeutic target for NPM1-mutated AML.
Collapse
Affiliation(s)
- Meixi Peng
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Jun Ren
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Yipei Jing
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Xueke Jiang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Qiaoling Xiao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Junpeng Huang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Yonghong Tao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Li Lei
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Xin Wang
- Department of HematologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zailin Yang
- Department of Clinical Laboratory The Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing University Cancer HospitalChongqingChina
| | - Zesong Yang
- Department of HematologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Qian Zhan
- The Center for Clinical Molecular Medical detectionThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Can Lin
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Guoxiang Jin
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Xian Zhang
- Immunology ProgramMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Ling Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| |
Collapse
|
25
|
Functional Proteomic Profiling of Triple-Negative Breast Cancer. Cells 2021; 10:cells10102768. [PMID: 34685748 PMCID: PMC8535076 DOI: 10.3390/cells10102768] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/26/2021] [Indexed: 01/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that comprises various disease entities, all of which share a set of common features: a lack of expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2, respectively. Because of their receptor status, conventional chemotherapy remains the main therapeutic option for TNBC patients. We employed a reverse phase protein array approach (RPPA), complemented by immunohistochemistry, to quantitatively profile the activation state of 84 actionable key signaling intermediates and phosphoproteins in a set of 44 TNBC samples. We performed supervised and unsupervised approaches to proteomic data analysis to identify groups of samples sharing common characteristics that could be amenable to existing therapies. We found the heterogenous activation of multiple pathways, with PI3 K/AKT/mTOR signaling being the most common event. Some specific individualized therapeutic possibilities include the expression of oncogenic KIT in association with cytokeratin 15 and Erk1/2 positive tumors, both of which may have clinical value.
Collapse
|
26
|
Hultmark S, Baudet A, Schmiderer L, Prabhala P, Palma-Tortosa S, Sandén C, Fioretos T, Sasidharan R, Larsson C, Lehmann S, Juliusson G, Ek F, Magnusson M. Combinatorial molecule screening identified a novel diterpene and the BET inhibitor CPI-203 as differentiation inducers of primary acute myeloid leukemia cells. Haematologica 2021; 106:2566-2577. [PMID: 32855276 PMCID: PMC8485661 DOI: 10.3324/haematol.2020.249177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Combination treatment has proven effective for patients with acute promyelocytic leukemia, exemplifying the importance of therapy targeting multiple components of oncogenic regulation for a successful outcome. However, recent studies have shown that the mutational complexity of acute myeloid leukemia (AML) precludes the translation of molecular targeting into clinical success. Here, as a complement to genetic profiling, we used unbiased, combinatorial in vitro drug screening to identify pathways that drive AML and to develop personalized combinatorial treatments. First, we screened 513 natural compounds on primary AML cells and identified a novel diterpene (H4) that preferentially induced differentiation of FLT3 wild-type AML, while FLT3-ITD/mutations conferred resistance. The samples responding to H4, displayed increased expression of myeloid markers, a clear decrease in the nuclear-cytoplasmic ratio and the potential of re-activation of the monocytic transcriptional program reducing leukemia propagation in vivo. By combinatorial screening using H4 and molecules with defined targets, we demonstrated that H4 induces differentiation by the activation of the protein kinase C (PKC) signaling pathway, and in line with this, activates PKC phosphorylation and translocation of PKC to the cell membrane. Furthermore, the combinatorial screening identified a bromo- and extra-terminal domain (BET) inhibitor that could further improve H4-dependent leukemic differentiation in FLT3 wild-type monocytic AML. These findings illustrate the value of an unbiased, multiplex screening platform for developing combinatorial therapeutic approaches for AML.
Collapse
Affiliation(s)
- Simon Hultmark
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Aurélie Baudet
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Ludwig Schmiderer
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Pavan Prabhala
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sara Palma-Tortosa
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Carl Sandén
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thoas Fioretos
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Christer Larsson
- Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Sören Lehmann
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Gunnar Juliusson
- Department of Hematology, Skane University Hospital, Lund, Sweden
| | - Fredrik Ek
- Chemical Biology and Therapeutics, Lund University, Lund, Sweden
| | - Mattias Magnusson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| |
Collapse
|
27
|
Park J, Park H, Byun JM, Hong J, Shin DY, Koh Y, Yoon SS. Pan-RAF inhibitor LY3009120 is highly synergistic with low-dose cytarabine, but not azacitidine, in acute myeloid leukemia with RAS mutations. Oncol Lett 2021; 22:745. [PMID: 34539849 DOI: 10.3892/ol.2021.13006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/26/2021] [Indexed: 11/06/2022] Open
Abstract
Alterations in RAS oncogenes have been implicated in various types of cancer, including acute myeloid leukemia (AML). Considering that currently, there are no targeted therapies for patients with RAS-mutated AML despite the poor outcomes, RAF may be a potential target for AML. In this study, we first analyzed the efficacy of different MAPK inhibitors in AML cell lines. We found that LY3009120, a pan-RAF inhibitor, significantly decreased cell survival in RAS-mutated AML cell lines. We then investigated the synergistic effects of LY3009120 with either cytarabine or azacitidine. We found that the combination of low-dose cytarabine and LY3009120 showed a synergistic effect in NRAS-mutated HL-60 cells and KRAS-mutated NB4 cells. This effect was caused by a decrease in proliferation, induction of apoptosis, and cell growth arrest through a decrease in phosphorylated MEK and ERK along with a cytotoxic response occurring specifically for the RAS mutation of the pan-RAF inhibitor LY3009120. In addition, we confirmed that combination treatment with low-dose cytarabine and LY3009120 led to an increase in apoptosis in primary AML cells. Our findings indicate that combination therapy with pan-RAF inhibitor LY3009120 and low-dose cytarabine may be a promising treatment strategy for RAS-mutated AML.
Collapse
Affiliation(s)
- Jihyun Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Hematology Oncology Department, Center for Medical Innovation, Seoul National University Hospital, Seoul 03082, Republic of Korea
| | - Hyejoo Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Hematology Oncology Department, Center for Medical Innovation, Seoul National University Hospital, Seoul 03082, Republic of Korea
| | - Ja Min Byun
- Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Junshik Hong
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Hematology Oncology Department, Center for Medical Innovation, Seoul National University Hospital, Seoul 03082, Republic of Korea.,Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Dong-Yeop Shin
- Hematology Oncology Department, Center for Medical Innovation, Seoul National University Hospital, Seoul 03082, Republic of Korea.,Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Youngil Koh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Hematology Oncology Department, Center for Medical Innovation, Seoul National University Hospital, Seoul 03082, Republic of Korea.,Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Hematology Oncology Department, Center for Medical Innovation, Seoul National University Hospital, Seoul 03082, Republic of Korea.,Department of Internal Medicine, Division of Hematology and Medical Oncology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| |
Collapse
|
28
|
Lu J, Chen S, Tan H, Huang Z, Li B, Liu L, Chen Y, Zeng X, Zou Y, Xu L. Eukaryotic initiation factor-2, gamma subunit, suppresses proliferation and regulates the cell cycle via the MAPK/ERK signaling pathway in acute myeloid leukemia. J Cancer Res Clin Oncol 2021; 147:3157-3168. [PMID: 34232382 DOI: 10.1007/s00432-021-03712-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/23/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE The expression of eukaryotic translation initiation factor-2 subunit 3 (EIF2S3) in patients with non-small cell lung and colorectal cancer is lower than that in healthy individuals. However, the functions of EIF2S3 remain unclear, and its study in leukemia has not been reported. The article aims to explore the role of EIF2S3 in AML (acute myeloid leukemia) and its underlying mechanism. METHODS Reverse transcription-quantitative PCR was performed to evaluate the expression levels of EIF2S3, and its association with patient prognosis was determined. Inducible HEL-EIF2S3 and HL-60-EIF2S3 cell lines were established by retrovirus infection. Cellular proliferation and the cell cycle were analyzed using Cell Counting Kit-8 and flow cytometric analyses. Tumorigenic ability was evaluated using xenograft nude mouse model. Gene expression profiles were analyzed in HL-60-EIF2S3 cells by next-generation sequencing, and WB analysis was performed to detect the expression of related proteins. RESULTS The expression of EIF2S3 in patients with AML was lower than that experiencing CR (P = 0.02). Furthermore, EIF2S3 overexpression inhibited cellular proliferation, halted G0/1 to S phase cell cycle progression, and inhibited tumorigenicity (P = 0.015). 479 differentially expressed genes were identified between HL60-EIF2S3 DOX (-) and HL60-EIF2S3 DOX ( +) cells via NGS and several of them involved in MAPK/ERK signaling pathway. The phosphorylation levels of ERK decreased when EIF2S3 was overexpressed (P < 0.050). CONCLUSION EIF2S3 overexpression may result in a decrease in cellular proliferation, cell cycle arrest, and tumorigenic inhibition via the MAPK/ERK signaling pathway in AML cells.
Collapse
Affiliation(s)
- Jielun Lu
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China.,Department of Urology and Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Urology, Guangzhou, 510230, Guangdong, People's Republic of China
| | - Shuyi Chen
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China.,Department of Urology and Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Urology, Guangzhou, 510230, Guangdong, People's Republic of China
| | - Huo Tan
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Zhenqian Huang
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Bo Li
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Ling Liu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 51260, People's Republic of China
| | - Yimin Chen
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Xiaozhen Zeng
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China
| | - Yawei Zou
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China.
| | - Lihua Xu
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Yuexiu, Guangzhou, 510000, Guangdong, People's Republic of China. .,Department of Urology and Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Urology, Guangzhou, 510230, Guangdong, People's Republic of China.
| |
Collapse
|
29
|
Vadakekolathu J, Minden MD, Hood T, Church SE, Reeder S, Altmann H, Sullivan AH, Viboch EJ, Patel T, Ibrahimova N, Warren SE, Arruda A, Liang Y, Smith TH, Foulds GA, Bailey MD, Gowen-MacDonald J, Muth J, Schmitz M, Cesano A, Pockley AG, Valk PJM, Löwenberg B, Bornhäuser M, Tasian SK, Rettig MP, Davidson-Moncada JK, DiPersio JF, Rutella S. Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia. Sci Transl Med 2021; 12:12/546/eaaz0463. [PMID: 32493790 DOI: 10.1126/scitranslmed.aaz0463] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/20/2020] [Accepted: 04/21/2020] [Indexed: 01/13/2023]
Abstract
Acute myeloid leukemia (AML) is a molecularly and clinically heterogeneous hematological malignancy. Although immunotherapy may be an attractive modality to exploit in patients with AML, the ability to predict the groups of patients and the types of cancer that will respond to immune targeting remains limited. This study dissected the complexity of the immune architecture of AML at high resolution and assessed its influence on therapeutic response. Using 442 primary bone marrow samples from three independent cohorts of children and adults with AML, we defined immune-infiltrated and immune-depleted disease classes and revealed critical differences in immune gene expression across age groups and molecular disease subtypes. Interferon (IFN)-γ-related mRNA profiles were predictive for both chemotherapy resistance and response of primary refractory/relapsed AML to flotetuzumab immunotherapy. Our compendium of microenvironmental gene and protein profiles provides insights into the immuno-biology of AML and could inform the delivery of personalized immunotherapies to IFN-γ-dominant AML subtypes.
Collapse
Affiliation(s)
| | - Mark D Minden
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | - Tressa Hood
- NanoString Technologies Inc., Seattle, WA 98109, USA
| | | | - Stephen Reeder
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Heidi Altmann
- Department of Medicine, Universitätsklinikum Carl Gustav Carus, 01307 Dresden, Germany
| | | | | | - Tasleema Patel
- Department of Pediatrics, Division of Oncology and Centre for Childhood Cancer Research, Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, PA 19104, USA
| | - Narmin Ibrahimova
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | | | - Andrea Arruda
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | - Yan Liang
- NanoString Technologies Inc., Seattle, WA 98109, USA
| | | | - Gemma A Foulds
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK
| | | | | | - John Muth
- MacroGenics Inc., Rockville, MD 20850, USA
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | | | - A Graham Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK.,Centre for Health, Ageing and Understanding Disease (CHAUD), Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Peter J M Valk
- Department of Hematology, Erasmus University Medical Centre, 3000CA Rotterdam, Netherlands
| | - Bob Löwenberg
- Department of Hematology, Erasmus University Medical Centre, 3000CA Rotterdam, Netherlands
| | - Martin Bornhäuser
- Department of Medicine, Universitätsklinikum Carl Gustav Carus, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sarah K Tasian
- Department of Pediatrics, Division of Oncology and Centre for Childhood Cancer Research, Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, PA 19104, USA
| | - Michael P Rettig
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - John F DiPersio
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK. .,Centre for Health, Ageing and Understanding Disease (CHAUD), Nottingham Trent University, Nottingham NG11 8NS, UK
| |
Collapse
|
30
|
Yang L, Zhang H, Yang X, Lu T, Ma S, Cheng H, Yen K, Cheng T. Prognostic Prediction of Cytogenetically Normal Acute Myeloid Leukemia Based on a Gene Expression Model. Front Oncol 2021; 11:659201. [PMID: 34123815 PMCID: PMC8190396 DOI: 10.3389/fonc.2021.659201] [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: 01/27/2021] [Accepted: 03/26/2021] [Indexed: 11/22/2022] Open
Abstract
Acute myeloid leukemia (AML) refers to a heterogeneous group of hematopoietic malignancies. The well-known European Leukemia Network (ELN) stratifies AML patients into three risk groups, based primarily on the detection of cytogenetic abnormalities. However, the prognosis of cytogenetically normal AML (CN-AML), which is the largest AML subset, can be hard to define. Moreover, the clinical outcomes associated with this subgroup are diverse. In this study, using transcriptome profiles collected from CN-AML patients in the BeatAML cohort, we constructed a robust prognostic Cox model named NEST (Nine-gEne SignaTure). The validity of NEST was confirmed in four external independent cohorts. Moreover, the risk score predicted by the NEST model remained an independent prognostic factor in multivariate analyses. Further analysis revealed that the NEST model was suitable for bone marrow mononuclear cell (BMMC) samples but not peripheral blood mononuclear cell (PBMC) samples, which indirectly indicated subtle differences between BMMCs and PBMCs. Our data demonstrated the robustness and accuracy of the NEST model and implied the importance of the immune dysfunction in the leukemogenesis that occurs in CN-AML, which shed new light on the further exploration of molecular mechanisms and treatment guidance for CN-AML.
Collapse
Affiliation(s)
- Liu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Houyu Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xue Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Ting Lu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Shihui Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Kuangyu Yen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
31
|
Li R, Ding Z, Jin P, Wu S, Jiang G, Xiang R, Wang W, Jin Z, Li X, Xue K, Wu X, Li J. Development and Validation of a Novel Prognostic Model for Acute Myeloid Leukemia Based on Immune-Related Genes. Front Immunol 2021; 12:639634. [PMID: 34025649 PMCID: PMC8131848 DOI: 10.3389/fimmu.2021.639634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/19/2021] [Indexed: 01/02/2023] Open
Abstract
The prognosis of acute myeloid leukemia (AML) is closely related to immune response changes. Further exploration of the pathobiology of AML focusing on immune-related genes would contribute to the development of more advanced evaluation and treatment strategies. In this study, we established a novel immune-17 signature based on transcriptome data from The Cancer Genome Atlas (TCGA) and The Genotype-Tissue Expression (GTEx) databases. We found that immune biology processes and transcriptional dysregulations are critical factors in the development of AML through enrichment analyses. We also formulated a prognostic model to predict the overall survival of AML patients by using LASSO (Least Absolute Shrinkage and Selection Operator) regression analysis. Furthermore, we incorporated the immune-17 signature to improve the prognostic accuracy of the ELN2017 risk stratification system. We concluded that the immune-17 signature represents a novel useful model for evaluating AML survival outcomes and may be implemented to optimize treatment selection in the next future.
Collapse
Affiliation(s)
- Ran Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zuoyou Ding
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Peng Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shishuang Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ge Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rufang Xiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenfang Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyang Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Xue
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolu Wu
- Department of Children Health Care, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Junmin Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Huang R, Liao X, Wang X, Li Q. Comprehensive investigation of the clinical significance of long non-coding RNA HOXA-AS2 in acute myeloid leukemia using genome-wide RNA sequencing dataset. J Cancer 2021; 12:2151-2164. [PMID: 33754013 PMCID: PMC7974522 DOI: 10.7150/jca.48045] [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: 05/11/2020] [Accepted: 10/09/2020] [Indexed: 12/20/2022] Open
Abstract
Objective: The present study aimed to determine the prognostic value of HOXA cluster antisense RNA2 (HOXA-AS2) in acute myeloid leukemia (AML), and to explore its potential molecular mechanisms. We also screening of potential drugs targeting HOXA-AS2 in AML. Methods: The level 3 raw genome-wide RNA sequencing dataset of AML was download from The Cancer Genome Atlas (TCGA) Data Portal, and the potential molecular mechanisms and drugs prediction of HOXA-AS2 in AML were explored using multiple bioinformatics analysis approaches. Results: TCGA AML cohort dataset indicated that HOXA-AS2 was significantly up-regulated in AML bone marrow tissues, and high HOXA-AS2 expression was related to poor overall survival (log-rank P=0.0284, hazard ratio 1.640, 95% confidence interval 1.046-2.573). Functional enrichment of differentially expressed genes (DEGs) suggested that the difference in prognosis between AML patients with high- and low-HOXA-AS2 expression may be due to differences in biological processes and pathways, including cell adhesion, angiogenesis, mitogen-activated protein kinase, cell differentiation, and other biological processes, and phosphatidylinositol 3 kinase-protein kinase B and Wnt signaling pathways. We also screened out three potential HOXA-AS2-targeted therapeutic drugs for AML, megestrol, carmustine, and cefoxitin, based on these DEGs. Functional enrichment analysis of HOXA-AS2-co-expressed genes revealed that HOXA-AS2 may act a part in AML by regulating nuclear factor-κB transcription factor activity, DNA methylation, angiogenesis, apoptosis, cell migration, Toll-like receptor 4, and Wnt signaling pathways. Conclusion: Our findings suggest that HOXA-AS2 is up-regulated in the bone marrow in patients with AML, and may serve as a novel prognostic biomarker for AML.
Collapse
Affiliation(s)
- Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qiaochuan Li
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|
33
|
Alsufyani A, Alanazi R, Woolley JF, Dahal LN. Old Dog, New Trick: Type I IFN-Based Treatment for Acute Myeloid Leukemia. Mol Cancer Res 2021; 19:753-756. [PMID: 33500358 DOI: 10.1158/1541-7786.mcr-20-0871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/20/2020] [Accepted: 01/21/2021] [Indexed: 11/16/2022]
Abstract
Despite strong biological rationale for the use of type-I IFNs for the treatment of acute myeloid leukemia (AML), their usage is limited to few hematologic malignancies. Here, we propose that innate immune sensing machinery, particularly the stimulator of IFN genes pathway, may be exploited to deliver antileukemic effects in AML.
Collapse
Affiliation(s)
- Abdullah Alsufyani
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, England, United Kingdom
| | - Rehab Alanazi
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, England, United Kingdom
| | - John F Woolley
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, England, United Kingdom
| | - Lekh N Dahal
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, England, United Kingdom. .,MRC Centre for Drug Safety Science, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, England, United Kingdom
| |
Collapse
|
34
|
Zhang C, Liu T, Luo P, Gao L, Liao X, Ma L, Jiang Z, Liu D, Yang Z, Jiang Q, Wang Y, Tan X, Luo S, Wang Y, Shi C. Near-infrared oxidative phosphorylation inhibitor integrates acute myeloid leukemia-targeted imaging and therapy. SCIENCE ADVANCES 2021; 7:7/1/eabb6104. [PMID: 33523835 PMCID: PMC7775779 DOI: 10.1126/sciadv.abb6104] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 11/04/2020] [Indexed: 05/11/2023]
Abstract
Acute myeloid leukemia (AML) is a deadly hematological malignancy with frequent disease relapse. The biggest challenge for AML therapy is the lack of methods to target and kill the heterogeneous leukemia cells, which lead to disease relapse. Here, we describe a near-infrared (NIR) fluorescent dye, IR-26, which preferentially accumulates in the mitochondria of AML cells, depending on the hyperactive glycolysis of malignant cell, and simultaneously impairs oxidative phosphorylation (OXPHOS) to exert targeted therapeutic effects for AML cells. In particular, IR-26 also exhibits potential for real-time monitoring of AML cells with an in vivo flow cytometry (IVFC) system. Therefore, IR-26 represents a novel all-in-one agent for the integration of AML targeting, detection, and therapy, which may help to monitor disease progression and treatment responses, prevent unnecessary delays in administering upfront therapy, and improve therapeutic efficiency to the residual AML cells, which are responsible for disease relapse.
Collapse
Affiliation(s)
- Chi Zhang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peng Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Li Gao
- Department of Hematology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xingyun Liao
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Le Ma
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhongyong Jiang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Dengqun Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zeyu Yang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qingzhi Jiang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xu Tan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shenglin Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Yang Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
35
|
Madan V, Koeffler HP. Differentiation therapy of myeloid leukemia: four decades of development. Haematologica 2021; 106:26-38. [PMID: 33054125 PMCID: PMC7776344 DOI: 10.3324/haematol.2020.262121] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia is characterized by arrested differentiation, and agents that overcome this block are therapeutically useful, as shown by the efficacy of all-trans retinoic acid in acute promyelocytic leukemia. However, the early promise of differentiation therapy did not translate into clinical benefit for other subtypes of acute myeloid leukemia, in which cytotoxic chemotherapeutic regimens remained the standard of care. Recent advances, including insights from sequencing of acute myeloid leukemia genomes, have led to the development of targeted therapies, comprising agents that induce differentiation of leukemic cells in preclinical models and clinical trials, thus rejuvenating interest in differentiation therapy. These agents act on various cellular processes including dysregulated metabolic programs, signaling pathways, epigenetic machinery and the cell cycle. In particular, inhibitors of mutant IDH1/2 and FLT3 have shown clinical benefit, leading to approval by regulatory bodies of their use. Besides the focus on recently approved differentiation therapies, this review also provides an overview of differentiation- inducing agents being tested in clinical trials or investigated in preclinical research. Combinatorial strategies are currently being tested for several agents (inhibitors of KDM1A, DOT1L, BET proteins, histone deacetylases), which were not effective in clinical studies as single agents, despite encouraging anti-leukemic activity observed in preclinical models. Overall, recently approved drugs and new investigational agents being developed highlight the merits of differentiation therapy; and ongoing studies promise further advances in the treatment of acute myeloid leukemia in the near future.
Collapse
Affiliation(s)
- Vikas Madan
- Cancer Science Institute of Singapore, National University of Singapore.
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore; Cedars-Sinai Medical Center, Division of Hematology/Oncology, UCLA School of Medicine, Los Angeles, CA, USA; Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), National University Hospital.
| |
Collapse
|
36
|
Altenburg B, Frings M, Schöbel JH, Goßen J, Pannen K, Vanderliek K, Rossetti G, Koschmieder S, Chatain N, Bolm C. Chiral Analogues of PFI-1 as BET Inhibitors and Their Functional Role in Myeloid Malignancies. ACS Med Chem Lett 2020; 11:1928-1934. [PMID: 33062175 DOI: 10.1021/acsmedchemlett.9b00625] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Structural analogues of PFI-1 varying at the sulfur core were prepared, and their activities as BET inhibitors in myeloid cell lines and primary cells from patients with acute myeloid leukemia were studied. Docking calculations followed by molecular dynamics simulations revealed the binding mode of the newly prepared inhibitors, suggesting explanations for the observed high enantiospecificity of the inhibitory activity.
Collapse
Affiliation(s)
- Bianca Altenburg
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Marcus Frings
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074 Aachen, Germany
| | - Jan-Hendrik Schöbel
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074 Aachen, Germany
| | - Jonas Goßen
- Institute of Neuroscience and Medicine (INM-9)/Institute for Advanced Simulation (IAS-5), Forschungszentrum Jülich, Wilhelm-Johnen-Straße, 52425 Jülich, Germany
- Faculty of Mathematics, Computer Science and Natural Sciences, RWTH Aachen University, 52074 Aachen, Germany
| | - Kristina Pannen
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Kim Vanderliek
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Giulia Rossetti
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
- Institute of Neuroscience and Medicine (INM-9)/Institute for Advanced Simulation (IAS-5), Forschungszentrum Jülich, Wilhelm-Johnen-Straße, 52425 Jülich, Germany
- Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, Wilhelm-Johnen-Straße, 52425 Jülich, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Carsten Bolm
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074 Aachen, Germany
| |
Collapse
|
37
|
Yuasa M, Yamamoto H, Mitsuki T, Kageyama K, Kaji D, Taya Y, Nishida A, Ishiwata K, Takagi S, Yamamoto G, Asano-Mori Y, Wake A, Koike Y, Makino S, Uchida N, Taniguchi S. Prognostic Impact of Cytogenetic Evolution on the Outcome of Allogeneic Stem Cell Transplantation in Patients with Acute Myeloid Leukemia in Nonremission: A Single-Institute Analysis of 212 Recipients. Biol Blood Marrow Transplant 2020; 26:2262-2270. [PMID: 32871257 DOI: 10.1016/j.bbmt.2020.08.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/16/2020] [Accepted: 08/22/2020] [Indexed: 12/16/2022]
Abstract
Recent progress in genetic analysis technology has helped researchers understand the pathogenesis of acute myeloid leukemia (AML). Considering this progress, AML karyotype is still one of the most significant prognostic factors that provides risk-adapted treatment approaches. Karyotype changes during treatment have been observed at times, but their prognostic impact is sparse, especially on allogeneic stem cell transplantation (allo-SCT). Here, we retrospectively investigated the effect of chromosomal changes between diagnosis and pretransplantation on the prognosis of allo-SCT by analyzing the outcomes of 212 consecutive patients who underwent allo-SCT for the first time at Toranomon Hospital, Tokyo, Japan, between 2008 and 2018. Cytogenetic abnormalities at diagnosis and pretransplantation were categorized based on the 2017 European Leukemia Net risk stratification. Genetic abnormalities such as FLT3-ITD and NPM1 were not considered in this study due to lack of genetic information in most patients. We defined cytogenetic evolution as chromosomal changes classified from lower category to higher category. Seventeen patients (8%) had cytogenetic evolution between diagnosis and pretransplantation, and they showed a significantly worse relapse rate than those who were categorized in the intermediate group based on the karyotype at diagnosis (3-year confidence interval [CI] of relapse, 57.4% versus 24.9%; P < .01). In multivariate analysis, cytogenetic evolution before allo-SCT had a significant impact on the CI of relapse (hazard ratio [HR], 3.89; CI, 1.75 to 8.67; P < .01), as well as the high score of the hematopoietic cell transplantation-specific comorbidity index (HR, 0.54; CI, 0.31 to 0.94; P = .03), but had no significant impact on overall survival or nonrelapse mortality. These results indicate that cytogenetic evolution has a significant impact after allo-SCT and should be considered during AML treatment.
Collapse
Affiliation(s)
| | | | - Takashi Mitsuki
- Department of Hematology, Toranomon Hospital Kajigaya, Kanagawa, Japan
| | - Kosei Kageyama
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Daisuke Kaji
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Yuki Taya
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Aya Nishida
- Department of Hematology, Toranomon Hospital Kajigaya, Kanagawa, Japan
| | - Kazuya Ishiwata
- Department of Hematology, Toranomon Hospital Kajigaya, Kanagawa, Japan
| | | | - Go Yamamoto
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | | | - Atsushi Wake
- Department of Hematology, Toranomon Hospital Kajigaya, Kanagawa, Japan
| | - Yukako Koike
- Department of Clinical Laboratory, Toranomon Hospital, Tokyo, Japan
| | - Shigeyoshi Makino
- Department of Transfusion Medicine, Toranomon Hospital, Tokyo, Japan
| | - Naoyuki Uchida
- Department of Hematology, Toranomon Hospital, Tokyo, Japan.
| | - Shuichi Taniguchi
- Department of Hematology, Toranomon Hospital, Tokyo, Japan; Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| |
Collapse
|
38
|
Dong Y, Han Y, Huang Y, Jiang S, Huang Z, Chen R, Yu Z, Yu K, Zhang S. PD-L1 Is Expressed and Promotes the Expansion of Regulatory T Cells in Acute Myeloid Leukemia. Front Immunol 2020; 11:1710. [PMID: 32849603 PMCID: PMC7412746 DOI: 10.3389/fimmu.2020.01710] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/26/2020] [Indexed: 12/13/2022] Open
Abstract
Intratumoral accumulation of CD4+CD25+Foxp3+ regulatory T (Treg) cells occurs in acute myeloid leukemia (AML), but little is known about the role of tumor cells themselves in this process. Here, we showed that an immune checkpoint PD-L1 expressed by AML cells promoted the conversion and expansion of Treg cells sustaining high expression of Foxp3 and PD-1 as well as a suppressive function. Furthermore, an AML cell line HEL overexpressed PD-L1 promoted the conversion and expansion of Treg cells and CD4+PD-1+Foxp3+ T (PD-1+Treg) cells from the conventional CD4+ T cells. CD4+CD25highPD-1+ T cells secreted more IL-10 production than CD4+CD25highPD-1− T cells. IL-35, another cytokine secreted by Treg cells, promoted the proliferation of HL-60 cells and enhanced chemoresistance to cytarabine. Blockade of PD-1 signaling using anti-PD-L1 antibody dramatically impaired the generation of Treg cells and sharply retarded the progression of a murine AML model injected with C1498 cells. The frequency of intratumoral PD-1+ Treg cells was capable of predicting patient survival in patients with AML. In conclusion, our data suggest that PD-L1 expression by AML cells may directly drive Treg cell expansion as a mechanism of immune evasion and the frequency of PD-1+ Treg cells is a potential prognostic predictor in patients with AML.
Collapse
Affiliation(s)
- Yuqing Dong
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yixiang Han
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yisha Huang
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Songfu Jiang
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ziyang Huang
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongrong Chen
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhijie Yu
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kang Yu
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenghui Zhang
- Wenzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Clinical Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
39
|
Villatoro A, Konieczny J, Cuminetti V, Arranz L. Leukemia Stem Cell Release From the Stem Cell Niche to Treat Acute Myeloid Leukemia. Front Cell Dev Biol 2020; 8:607. [PMID: 32754595 PMCID: PMC7367216 DOI: 10.3389/fcell.2020.00607] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/19/2020] [Indexed: 01/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous, complex, and deadly disease, whose treatment has hardly evolved for decades and grounds on the use of intensive chemotherapy regimens. Chemotherapy helps reduce AML bulk, but promotes relapse in the long-run by selection of chemoresistant leukemia stem cells (LSC). These may diversify and result in progression to more aggressive forms of AML. In vivo models suggest that the bone marrow stem cell niche helps LSC stay dormant and protected from chemotherapy. Here, we summarize relevant changes in stem cell niche homing and adhesion of AML LSC vs. healthy hematopoietic stem cells, and provide an overview of clinical trials aiming at targeting these processes for AML treatment and future directions within this field. Promising results with various non-mutation-targeted novel therapies directed to LSC eradication via interference with their anchoring to the stem cell niche have encouraged on-going or future advanced phase III clinical trials. In the coming years, we may see a shift in the focus of AML treatment to LSC-directed therapies if the prospect of improved cure rates holds true. In the future, AML treatment should lean toward personalized therapies using combinations of these compounds plus mutation-targeted agents and/or targeted delivery of chemotherapy, aiming at LSC eradication with reduced side effects.
Collapse
Affiliation(s)
- Alicia Villatoro
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Joanna Konieczny
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Vincent Cuminetti
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Lorena Arranz
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Norwegian Center for Molecular Medicine (NCMM), University of Oslo, Oslo, Norway
| |
Collapse
|
40
|
Yamashita M, Dellorusso PV, Olson OC, Passegué E. Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis. Nat Rev Cancer 2020; 20:365-382. [PMID: 32415283 PMCID: PMC7658795 DOI: 10.1038/s41568-020-0260-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2020] [Indexed: 12/17/2022]
Abstract
Haematopoiesis is governed by haematopoietic stem cells (HSCs) that produce all lineages of blood and immune cells. The maintenance of blood homeostasis requires a dynamic response of HSCs to stress, and dysregulation of these adaptive-response mechanisms underlies the development of myeloid leukaemia. Leukaemogenesis often occurs in a stepwise manner, with genetic and epigenetic changes accumulating in pre-leukaemic HSCs prior to the emergence of leukaemic stem cells (LSCs) and the development of acute myeloid leukaemia. Clinical data have revealed the existence of age-related clonal haematopoiesis, or the asymptomatic clonal expansion of mutated blood cells in the elderly, and this phenomenon is connected to susceptibility to leukaemic transformation. Here we describe how selection for specific mutations that increase HSC competitive fitness, in conjunction with additional endogenous and environmental changes, drives leukaemic transformation. We review the ways in which LSCs take advantage of normal HSC properties to promote survival and expansion, thus underlying disease recurrence and resistance to conventional therapies, and we detail our current understanding of leukaemic 'stemness' regulation. Overall, we link the cellular and molecular mechanisms regulating HSC behaviour with the functional dysregulation of these mechanisms in myeloid leukaemia and discuss opportunities for targeting LSC-specific mechanisms for the prevention or cure of malignant diseases.
Collapse
Affiliation(s)
- Masayuki Yamashita
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Paul V Dellorusso
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Oakley C Olson
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
41
|
Liang C, Peng CJ, Wang LN, Li Y, Zheng LM, Fan Z, Huang DP, Tang WY, Zhang XL, Huang LB, Tang YL, Luo XQ. Arsenic trioxide and all-trans retinoic acid suppress the expression of FLT3-ITD. Leuk Lymphoma 2020; 61:2692-2699. [PMID: 32536235 DOI: 10.1080/10428194.2020.1775212] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Cong Liang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chun-Jin Peng
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Na Wang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Li
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Min Zheng
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Fan
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan-Ping Huang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen-Yan Tang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Li Zhang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Bin Huang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Lai Tang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xue-Qun Luo
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
42
|
A parsimonious 3-gene signature predicts clinical outcomes in an acute myeloid leukemia multicohort study. Blood Adv 2020; 3:1330-1346. [PMID: 31015209 DOI: 10.1182/bloodadvances.2018030726] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous hematological malignancy with variable responses to chemotherapy. Although recurring cytogenetic abnormalities and gene mutations are important predictors of outcome, 50% to 70% of AMLs harbor normal or risk-indeterminate karyotypes. Therefore, identifying more effective biomarkers predictive of treatment success and failure is essential for informing tailored therapeutic decisions. We applied an artificial neural network (ANN)-based machine learning approach to a publicly available data set for a discovery cohort of 593 adults with nonpromyelocytic AML. ANN analysis identified a parsimonious 3-gene expression signature comprising CALCRL, CD109, and LSP1, which was predictive of event-free survival (EFS) and overall survival (OS). We computed a prognostic index (PI) using normalized gene-expression levels and β-values from subsequently created Cox proportional hazards models, coupled with clinically established prognosticators. Our 3-gene PI separated the adult patients in each European LeukemiaNet cytogenetic risk category into subgroups with different survival probabilities and identified patients with very high-risk features, such as those with a high PI and either FLT3 internal tandem duplication or nonmutated nucleophosmin 1. The PI remained significantly associated with poor EFS and OS after adjusting for established prognosticators, and its ability to stratify survival was validated in 3 independent adult cohorts (n = 905 subjects) and 1 cohort of childhood AML (n = 145 subjects). Further in silico analyses established that AML was the only tumor type among 39 distinct malignancies for which the concomitant upregulation of CALCRL, CD109, and LSP1 predicted survival. Therefore, our ANN-derived 3-gene signature refines the accuracy of patient stratification and the potential to significantly improve outcome prediction.
Collapse
|
43
|
The role of small molecule Flt3 receptor protein-tyrosine kinase inhibitors in the treatment of Flt3-positive acute myelogenous leukemias. Pharmacol Res 2020; 155:104725. [DOI: 10.1016/j.phrs.2020.104725] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 02/08/2023]
|
44
|
Salt-inducible kinase inhibition suppresses acute myeloid leukemia progression in vivo. Blood 2020; 135:56-70. [PMID: 31697837 DOI: 10.1182/blood.2019001576] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
Lineage-defining transcription factors (TFs) are compelling targets for leukemia therapy, yet they are among the most challenging proteins to modulate directly with small molecules. We previously used CRISPR screening to identify a salt-inducible kinase 3 (SIK3) requirement for the growth of acute myeloid leukemia (AML) cell lines that overexpress the lineage TF myocyte enhancer factor (MEF2C). In this context, SIK3 maintains MEF2C function by directly phosphorylating histone deacetylase 4 (HDAC4), a repressive cofactor of MEF2C. In this study, we evaluated whether inhibition of SIK3 with the tool compound YKL-05-099 can suppress MEF2C function and attenuate disease progression in animal models of AML. Genetic targeting of SIK3 or MEF2C selectively suppressed the growth of transformed hematopoietic cells under in vitro and in vivo conditions. Similar phenotypes were obtained when cells were exposed to YKL-05-099, which caused cell-cycle arrest and apoptosis in MEF2C-expressing AML cell lines. An epigenomic analysis revealed that YKL-05-099 rapidly suppressed MEF2C function by altering the phosphorylation state and nuclear localization of HDAC4. Using a gatekeeper allele of SIK3, we found that the antiproliferative effects of YKL-05-099 occurred through on-target inhibition of SIK3 kinase activity. Based on these findings, we treated 2 different mouse models of MLL-AF9 AML with YKL-05-099, which attenuated disease progression in vivo and extended animal survival at well-tolerated doses. These findings validate SIK3 as a therapeutic target in MEF2C-addicted AML and provide a rationale for developing druglike inhibitors of SIK3 for definitive preclinical investigation and for studies in human patients.
Collapse
|
45
|
Zhang Y, Xiao L. Identification and validation of a prognostic 8-gene signature for acute myeloid leukemia. Leuk Lymphoma 2020; 61:1981-1988. [PMID: 32268820 DOI: 10.1080/10428194.2020.1742898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In the present study, we aimed to identify some genes closely related to AML prognosis and investigate their potential roles. RNA-seq data of AML samples were accessed from the TCGA database and then analyzed in the Wilcox test. AML survival-related genes were selected and an 8-gene signature-based risk score model was in turn constructed (including TET3, S100A4, BATF, CLEC11A, PTP4A3, SPATS2L, SDHA, and ATOX1 8 feature genes) using the multivariate Cox regression analysis. Kaplan-Meier analysis was performed on the 8 genes in the training set (p = 2.826e - 11) and the test set (p = 2.213e - 2), and there was a remarkable difference in survival between the high and low-risk samples. Meanwhile, ROC analysis was conducted and revealed the relative higher accuracy of the risk score model applied in both the training set (1-year AUC = 0.864; 3-year AUC = 0.85) and test set (1-year AUC = 0.685; 3-year AUC = 0.678). Our study helps to extend our knowledge of the potential methods for AML prognosis.HighlightsA prognostic 8-gene (including TET3, CLEC11A, ATOX1, S100A4, BATF, PTP4A3, SPATS2L and SDHA 8) signature for acute myeloid leukemia (AML) was identified and validated.The influence of the expression of single gene in the model on the survival risk of AML patients was confirmed and the risk rate of 8 single-gene was compared.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Longyan Xiao
- Department of Hematology, Linyi People's Hospital, Linyi, Shandong, China
| |
Collapse
|
46
|
Design and synthesis of Imidazo[1,2-b]pyridazine IRAK4 inhibitors for the treatment of mutant MYD88 L265P diffuse large B-cell lymphoma. Eur J Med Chem 2020; 190:112092. [DOI: 10.1016/j.ejmech.2020.112092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/13/2022]
|
47
|
MicroRNA-181a-3p as a Diagnostic and Prognostic Biomarker for Acute Myeloid Leukemia. Mediterr J Hematol Infect Dis 2020; 12:e2020012. [PMID: 32180907 PMCID: PMC7059742 DOI: 10.4084/mjhid.2020.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/03/2020] [Indexed: 12/23/2022] Open
Abstract
Background Micro (mi) RNAs play an important role in the pathogenesis and development of acute myeloid leukemia (AML), and their abnormal expression may be sufficient to predict the prognosis and outcomes in AML patients. We evaluated the clinical diagnostic value of miRNA-181a-3p in predicting prognosis and outcomes in patients with AML. Methods A total of 119 newly diagnosed adult patients with AML and 60 healthy controls were recruited. Blood specimens were obtained from all AML patients at diagnosis, and 10 blood specimens were obtained on day 28 after induction chemotherapy. The controls also provided blood samples. Relative gene expression was quantified by PCR and determined using the comparative Ct method. Publicly available clinical data and gene expressions for 188 patients with AML were downloaded from TCGA data portal. Results Compared with healthy controls, the expression of miRNA-181a-3p was significantly increased in patients with AML. MiR-181a-3p expression could be used to discriminate AML patients from controls, with up-regulated expression correlating with favorable prognosis. Moreover, miRNA-181a-3p expression was significantly decreased in patients who achieved a complete response after induction chemotherapy. The multivariate Cox analysis highlighted the prognostic value of miR-181a-3p for patients with AML. Finally, we found that miR-181a-3p expression was negatively correlated with the expression of the NF-κB essential modulator (NEMO/IKBKG). Conclusions MiR-181a-3p may be clinically useful as a disease marker for AML, and enhanced the prediction of patient outcomes to chemotherapy.
Collapse
|
48
|
Abadir E, Gasiorowski RE, Silveira PA, Larsen S, Clark GJ. Is Hematopoietic Stem Cell Transplantation Required to Unleash the Full Potential of Immunotherapy in Acute Myeloid Leukemia? J Clin Med 2020; 9:E554. [PMID: 32085578 PMCID: PMC7073661 DOI: 10.3390/jcm9020554] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
From monoclonal antibodies (mAbs) to Chimeric Antigen Receptor (CAR) T cells, immunotherapies have enhanced the efficacy of treatments against B cell malignancies. The same has not been true for Acute Myeloid Leukemia (AML). Hematologic toxicity has limited the potential of modern immunotherapies for AML at preclinical and clinical levels. Gemtuzumab Ozogamicin has demonstrated hematologic toxicity, but the challenge of preserving normal hematopoiesis has become more apparent with the development of increasingly potent immunotherapies. To date, no single surface molecule has been identified that is able to differentiate AML from Hematopoietic Stem and Progenitor Cells (HSPC). Attempts have been made to spare hematopoiesis by targeting molecules expressed only on later myeloid progenitors as well as AML or using toxins that selectively kill AML over HSPC. Other strategies include targeting aberrantly expressed lymphoid molecules or only targeting monocyte-associated proteins in AML with monocytic differentiation. Recently, some groups have accepted that stem cell transplantation is required to access potent AML immunotherapy and envision it as a rescue to avoid severe hematologic toxicity. Whether it will ever be possible to differentiate AML from HSPC using surface molecules is unclear. Unless true specific AML surface targets are discovered, stem cell transplantation could be required to harness the true potential of immunotherapy in AML.
Collapse
Affiliation(s)
- Edward Abadir
- Dendritic Cell Research, ANZAC Research Institute, Concord 2139, NSW, Australia;
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown 2050, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| | - Robin E. Gasiorowski
- The University of Sydney, Camperdown 2039, NSW, Australia;
- Department of Haematology, Concord Repatriation and General Hospital, Concord 2039, NSW, Australia
| | - Pablo A. Silveira
- Dendritic Cell Research, ANZAC Research Institute, Concord 2139, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| | - Stephen Larsen
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown 2050, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| | - Georgina J. Clark
- Dendritic Cell Research, ANZAC Research Institute, Concord 2139, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| |
Collapse
|
49
|
Zhang X, Xu Y, Wang J, Zhao S, Li J, Huang X, Xu H, Zhang X, Suo S, Lv Y, Zhang Y, Yu W. miR-221-3p Delivered by BMMSC-Derived Microvesicles Promotes the Development of Acute Myelocytic Leukemia. Front Bioeng Biotechnol 2020; 8:81. [PMID: 32117949 PMCID: PMC7033425 DOI: 10.3389/fbioe.2020.00081] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/29/2020] [Indexed: 12/20/2022] Open
Abstract
Objective: The study aims to investigate the effects of miR-221-3p in bone marrow mesenchymal stem cell (BMMSC)-derived microvesicles (MVs) on cell cycle, proliferation and invasion of acute myelocytic leukemia (AML). Methods: Bioinformatics was used to predict differentially expressed miRNAs (DEmiRNAs) in AML. The morphology of BMMSC-derived MVs was observed under an electron microscope, and the positional relation of MVs and OCI-AML2 cells was observed by a fluorescence microscope. MTT, Transwell, and flow cytometry assays were used to analyze the effects of MVs on OCI-AML2 cells. The targeted relationship between miR-221-3p and CDKN1C was detected by dual luciferase assay. Results: It was verified that miR-221-3p promoted the proliferation, invasion and migration of OCI-AML2 cells, and induced the cell cycle arrest in G1/S phase as well as inhibited cell apoptosis. Further studies showed that MVs promoted the proliferation, migration and invasion of AML, and induced the cell cycle arrest in G1/S phase through miR-221-3p. It was confirmed that miR-221-3p can directly target CDKN1C to regulate cell cycle, proliferation and invasion of AML. Conclusion: miR-221-3p in BMMSC-derived MVs regulated AML cell cycle, cell proliferation and invasion through targeting CDKN1C. miR-221-3p and CDKN1C were considered to be potential targets and biomarkers for the treatment of AML in clinic.
Collapse
Affiliation(s)
- Xuewu Zhang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yu Xu
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jinghan Wang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Shuqi Zhao
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jianhu Li
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Xin Huang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Huan Xu
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Xiang Zhang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Shanshan Suo
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yunfei Lv
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yi Zhang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Wenjuan Yu
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| |
Collapse
|
50
|
Acute myeloid leukemia immune escape by epigenetic CD48 silencing. Clin Sci (Lond) 2020; 134:261-271. [PMID: 31922199 DOI: 10.1042/cs20191170] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/22/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is a malignant disorder of hemopoietic stem cells. AML can escape immunosurveillance of natural killer (NK) by gene mutation, fusions and epigenetic modification. The mechanism of AML immune evasion is not clearly understood. Here we show that CD48 high expression is a favorable prognosis factor that is down-regulated in AML patients, which can help AML evade from NK cell recognition and killing. Furthermore, we demonstrate that CD48 expression is regulated by methylation and that a hypomethylating agent can increase the CD48 expression, which increases the NK cells killing in vitro. Finally, we show that CD48 high expression can reverse the AML immune evasion and activate NK cells function in vivo. The present study suggests that a combination the hypomethylating agent and NK cell infusion could be a new strategy to cure AML.
Collapse
|