1
|
Zhu C, Feng X, Tong L, Mu P, Wang F, Quan W, Dong Y, Zhu X. Prediction of acute myeloid leukemia prognosis based on autophagy features and characterization of its immune microenvironment. Front Immunol 2024; 15:1489171. [PMID: 39650664 PMCID: PMC11621098 DOI: 10.3389/fimmu.2024.1489171] [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: 08/31/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Background Autophagy promotes the survival of acute myeloid leukemia (AML) cells by removing damaged organelles and proteins and protecting them from stress-induced apoptosis. Although many studies have identified candidate autophagy genes associated with AML prognosis, there are still great challenges in predicting the survival prognosis of AML patients. Therefore, it is necessary to identify more novel autophagy gene markers to improve the prognosis of AML by utilizing information at the molecular level. Methods In this study, the Random Forest, SVM and XGBoost algorithms were utilized to identify autophagy genes linked to prognosis, respectively. Subsequently, six autophagy genes (TSC2, CALCOCO2, BAG3, UBQLN4, ULK1 and DAPK1) that were significantly associated with patients' overall survival (OS) were identified using Lasso-Cox regression analysis. A prediction model incorporating these autophagy genes was then developed. In addition, the immunological microenvironment analysis of autophagy genes was performed in this study. Results The experimental results showed that the predictive model had good predictive ability. After adjusting for clinicopathologic parameters, this feature proved an independent prognostic predictor and was validated in an external AML sample set. Analysis of differentially expressed genes in patients in the high-risk and low-risk groups showed that these genes were enriched in immune-related pathways such as humoral immune response, T cell differentiation in thymus and lymphocyte differentiation. Then immune infiltration analysis of autophagy genes in patients showed that the cellular abundance of T cells CD4+ memory activated, NK cells activated and T cells CD4+ in the high-risk group was significantly lower than that in the low-risk group. Conclusion This study systematically analyzed autophagy-related genes (ARGs) and developed prognostic predictors related to OS for patients with AML, thus more accurately assessing the prognosis of AML patients. This not only helps to improve the prognostic assessment and therapeutic outcome of patients, but may also provide new help for future research and clinical applications.
Collapse
Affiliation(s)
- Chaoqun Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Xiangyan Feng
- Department of Hematology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Lanxin Tong
- Guangzhou Dublin International College of Life Sciences and Technology, South China Agricultural University, Guangzhou, Guangdong, China
| | - Peizheng Mu
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Fei Wang
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Wei Quan
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, China
| | - Xiao Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| |
Collapse
|
2
|
Travaglini S, Gurnari C, Ottone T, Voso MT. Advances in the pathogenesis of FLT3 -mutated acute myeloid leukemia and targeted treatments. Curr Opin Oncol 2024; 36:569-576. [PMID: 39246183 PMCID: PMC11460763 DOI: 10.1097/cco.0000000000001094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
PURPOSE OF REVIEW FLT3 mutations are among the most common myeloid drivers identified in adult acute myeloid leukemia (AML). Their identification is crucial for the precise risk assessment because of the strong prognostic significance of the most recurrent type of FLT3 alterations, namely internal tandem duplications (ITDs). Recent advances in the pathogenesis and biology of FLT3 -mutated AML have opened an opportunity for development and application of selective inhibition of FLT3 pathway. RECENT FINDINGS In the last decade, at least three targeted treatments have been approved by regulatory agencies and several others are currently under investigations. Here, we review the latest advance in the role of FLT3 mutations in AML, providing an outline of the available therapeutic strategies, their mechanisms of actions and of resistance, as well as routes for potential improvement. SUMMARY The availability of FLT3 inhibitors has improved outcomes in AML harboring such mutations, currently also reflected in disease stratification and recommendations. Newer inhibitors are under investigations, and combinations with chemotherapy or other targeted treatments are being explored to further improve disease outcomes.
Collapse
Affiliation(s)
- Serena Travaglini
- Department of Biomedicine and Prevention, University of Tor Vergata
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmelo Gurnari
- Department of Biomedicine and Prevention, University of Tor Vergata
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Tor Vergata
| | | |
Collapse
|
3
|
Zeng X, Zhang H, Guo J, Yang D, Zhu Y, Liu N, Tang J, Liu T, Zhao X. A novel bispecific T-cell engager using the ligand-target csGRP78 against acute myeloid leukemia. Cell Mol Life Sci 2024; 81:371. [PMID: 39196413 PMCID: PMC11358366 DOI: 10.1007/s00018-024-05410-0] [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: 01/03/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
Current medical therapies for treating acute myeloid leukemia (AML) remain unmet, and AML patients may benefit from targeted immunotherapy approaches that focus on specific tumor antigens. GRP78, which is upregulated in various malignant tumors such as AML, is partially expressed as cell surface GRP78 (csGRP78) on the cell membrane, making it an ideal target for redirecting T cells, including T-cell engagers. However, considering the conventional approach of using two scFv segments to construct a bispecific T-cell engager (BiTE), we have undertaken the development of a novel BiTE that utilizes a cyclic peptide ligand to specifically target csGRP78, which we refer to as GRP78-CD3/BiTE. We studied the effects of GRP78-CD3/BiTE on treatments for AML in vitro and in vivo and assessed the pharmacokinetics of this engager. Our findings demonstrated that GRP78-CD3/BiTE could not only effectively mediate the cytotoxicity of T cells against csGRP78-expressing AML cells but also specifically eliminate primary AML tumor cells in vitro. Furthermore, GRP78-CD3/BiTE exhibited a longer half-life despite having a lower molecular weight than CD19-CD3/BiTE. In a xenograft mouse model of AML, treatment with GRP78-CD3/BiTE prolonged the survival time of the mice. Our findings demonstrate that GRP78-CD3/BiTE is effective and selective for eliminating csGRP78-expressing AML cells and suggest that this approach to targeted immunotherapy could lead to effective new treatments for AML.
Collapse
MESH Headings
- Endoplasmic Reticulum Chaperone BiP
- Humans
- Animals
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Mice
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- CD3 Complex/immunology
- Heat-Shock Proteins/immunology
- Heat-Shock Proteins/metabolism
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Ligands
- Female
- Mice, SCID
- Immunotherapy/methods
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Xiaozhu Zeng
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hang Zhang
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Guo
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dong Yang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongjie Zhu
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Nan Liu
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jie Tang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ting Liu
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xudong Zhao
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
4
|
Chen Z, Zheng S, Han J, Fu L, Fu J, Zhang Z, Hong P, Feng W. Molecular mechanisms of ferroptosis and its roles in leukemia. Front Oncol 2023; 13:1308869. [PMID: 38125948 PMCID: PMC10731040 DOI: 10.3389/fonc.2023.1308869] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Cell death is a complex process required to maintain homeostasis and occurs when cells are damage or reach end of life. As research progresses, it is apparent that necrosis and apoptosis do not fully explain the whole phenomenon of cell death. Therefore, new death modalities such as autophagic cell death, and ferroptosis have been proposed. In recent years, ferroptosis, a new type of non-apoptotic cell death characterized by iron-dependent lipid peroxidation and reactive oxygen species (ROS) accumulation, has been receiving increasing attention. Ferroptosis can be involved in the pathological processes of many disorders, such as ischemia-reperfusion injury, nervous system diseases, and blood diseases. However, the specific mechanisms by which ferroptosis participates in the occurrence and development of leukemia still need to be more fully and deeply studied. In this review, we present the research progress on the mechanism of ferroptosis and its role in leukemia, to provide new theoretical basis and strategies for the diagnosis and treatment of clinical hematological diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weiying Feng
- Department of Hematology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| |
Collapse
|
5
|
Yao Q, Zhang L, Liu Z, Yu L, Wang Y, Liu J, Wang Y. LncRNA MAFG-AS1-induced acute myeloid leukemia development via modulating miR-147b/HOXA9. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:19250-19258. [PMID: 36229729 DOI: 10.1007/s11356-022-23537-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 02/22/2021] [Indexed: 06/16/2023]
Abstract
Recent references discovered that lncRNAs acted roles in malignant cancer development. However, the role of MAFG-AS1 in acute myeloid leukemia (AML) development remains unknown. MAFG-AS1 and miR-147b were determined in AML cells and specimens using qRT-PCR assay. Cell proliferation was detected by CCK-8 analysis and flow cytometry was carried out to measure cell cycle. Luciferase reporter analysis was done to determine the mechanism of MAFG-AS1 and miR-147b. We noted that MAFG-AS1 was overexpressed in AML cells and in serum and bone narrow from AML compared with normal controls specimen. Elevated expression of MAFG-AS1 increased cell growth, cycle and EMT in AML cell HL-60 cell. MAFG-AS1 sponged miR-147b expression in HL-60 cell. Moreover, miR-147b was downregulated in AML cells and in serum and bone narrow from AML compared with normal control specimen. miR-147b was negatively correlated with MAFG-AS1 in the serum and bone narrow of AML cases. We illustrated that HOXA9 was one target of miR-147b and ectopic expression of MAFG-AS1 enhanced HOXA9 expression HL-60 cell. Forced expression of MAFG-AS1 induced cell growth, cycle, and EMT via promoting HOXA9. These data illustrated that MAFG-AS1 acted as one oncogenic gene and accelerated AML progression via modulating miR-147b/HOXA9 axis.
Collapse
Affiliation(s)
- Qiying Yao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Li Zhang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Zhengjuan Liu
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Lei Yu
- Department of Infectious Disease, The Fourth Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yuchuan Wang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Junli Liu
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China
| | - Yingjie Wang
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning, China.
| |
Collapse
|
6
|
A Focus on Intermediate-Risk Acute Myeloid Leukemia: Sub-Classification Updates and Therapeutic Challenges. Cancers (Basel) 2022; 14:cancers14174166. [PMID: 36077703 PMCID: PMC9454629 DOI: 10.3390/cancers14174166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/16/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) represents a heterogeneous group of hematopoietic neoplasms deriving from the abnormal proliferation of myeloid progenitors in the bone marrow. Patients with AML may have highly variable outcomes, which are generally dictated by individual clinical and genomic characteristics. As such, the European LeukemiaNet 2017 and 2022 guidelines categorize newly diagnosed AML into favorable-, intermediate-, and adverse-risk groups, based on their molecular and cytogenetic profiles. Nevertheless, the intermediate-risk category remains poorly defined, as many patients fall into this group as a result of their exclusion from the other two. Moreover, further genomic data with potential prognostic and therapeutic influences continue to emerge, though they are yet to be integrated into the diagnostic and prognostic models of AML. This review highlights the latest therapeutic advances and challenges that warrant refining the prognostic classification of intermediate-risk AML.
Collapse
|
7
|
Huang Y, Hong M, Qu Z, Zheng W, Hu H, Li L, Lu T, Xie Y, Ying S, Zhu Y, Liu L, Huang W, Fu S, Chen J, Wu K, Liu M, Luo Q, Wu Y, He F, Zhang J, Zhang J, Chen Y, Zhao M, Cai Z, Huang H, Sun J. Non-Ablative Chemotherapy Followed by HLA-Mismatched Allogeneic CD3 + T-Cells Infusion Causes An Augment of T-Cells With Mild CRS: A Multi-Centers Single-Arm Prospective Study on Elderly Acute Myeloid Leukemia and int-2/High Risk Myelodysplastic Syndrome Patients. Front Oncol 2021; 11:741341. [PMID: 34722293 PMCID: PMC8548743 DOI: 10.3389/fonc.2021.741341] [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: 07/14/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To evaluate the efficacy and safety of standard or low-dose chemotherapy followed by HLA-mismatched allogeneic T-cell infusion (allo-TLI) for the treatment of elderly patients with acute myeloid leukemia (AML) and patients with intermediate-2 to high-risk myelodysplastic syndrome (MDS). Methods We carried out a prospective, multicenter, single-arm clinical trial. Totally of 25 patients were enrolled, including 17 AML patients and 8 MDS patients. Each patient received four courses of non-ablative chemotherapy, with HLA-mismatched donor CD3+ allo-TLI 24 h after each course. AML patients received chemotherapy with decitabine, idarubicin, and cytarabine, and MDS patients received decitabine, cytarabine, aclarubicin, and granulocyte colony-stimulating factor. Results A total of 79 procedures were performed. The overall response rates of the AML and MDS patients were 94% and 75% and the 1-year overall survival rates were 88% (61-97%) and 60% (13-88%), respectively. The overall 60-day treatment-related mortality was 8%. Compared with a historical control cohort that received idarubicin plus cytarabine (3 + 7), the study group showed significantly better overall response (94% vs. 50%, P=0.002) and overall survival rates (the 1-year OS rate was 88% vs. 27%, P=0.014). Post-TLI cytokine-release syndrome (CRS) occurred after 79% of allo-TLI operations, and 96% of CRS reactions were grade 1. Conclusion Elderly AML patients and intermediate-2 to high-risk MDS patients are usually insensitive to or cannot tolerate regular chemotherapies, and may not have the opportunity to undergo allogeneic stem cell transplantation. Our study showed that non-ablative chemotherapy followed by HLA-mismatched allo-TLI is safe and effective, and may thus be used as a first-line treatment for these patients. Clinical Trial Registration https://www.chictr.org.cn/showproj.aspx?proj=20112.
Collapse
Affiliation(s)
- Yan Huang
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minghua Hong
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhigang Qu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Weiyan Zheng
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huixian Hu
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Linjie Li
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Ting Lu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Xie
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuangwei Ying
- Department of Hematology, Taizhou Hospital of Zhejiang Province, Taizhou, China
| | - Yuanyuan Zhu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lizhen Liu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weijia Huang
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Fu
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Chen
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Kangli Wu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Mingsuo Liu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Qiulian Luo
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Yajun Wu
- Department of Hematology, Yiwu Central Hospital, Yiwu, China
| | - Fang He
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jingcheng Zhang
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Junyu Zhang
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Yu Chen
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Minlei Zhao
- Department of Hematology, The Central Hospital of Lishui City, Lishui, China
| | - Zhen Cai
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - He Huang
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Sun
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy; Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Institute of Hematology, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Zhao Y, Huang Z, Peng H. Molecular Mechanisms of Ferroptosis and Its Roles in Hematologic Malignancies. Front Oncol 2021; 11:743006. [PMID: 34778060 PMCID: PMC8582018 DOI: 10.3389/fonc.2021.743006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
Cell death is essential for the normal metabolism of human organisms. Ferroptosis is a unique regulated cell death (RCD) mode characterized by excess accumulation of iron-dependent lipid peroxide and reactive oxygen species (ROS) compared with other well-known programmed cell death modes. It has been currently recognized that ferroptosis plays a rather important role in the occurrence, development, and treatment of traumatic brain injury, stroke, acute kidney injury, liver damage, ischemia-reperfusion injury, tumor, etc. Of note, ferroptosis may be explained by the expression of various molecules and signaling components, among which iron, lipid, and amino acid metabolism are the key regulatory mechanisms of ferroptosis. Meanwhile, tumor cells of hematological malignancies, such as leukemia, lymphoma, and multiple myeloma (MM), are identified to be sensitive to ferroptosis. Targeting potential regulatory factors in the ferroptosis pathway may promote or inhibit the disease progression of these malignancies. In this review, a systematic summary was conducted on the key molecular mechanisms of ferroptosis and the current potential relationships of ferroptosis with leukemia, lymphoma, and MM. It is expected to provide novel potential therapeutic approaches and targets for hematological malignancies.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, China
| |
Collapse
|
9
|
Gurnari C, Pagliuca S, Visconte V. The Interactome between Metabolism and Gene Mutations in Myeloid Malignancies. Int J Mol Sci 2021; 22:ijms22063135. [PMID: 33808599 PMCID: PMC8003366 DOI: 10.3390/ijms22063135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 12/19/2022] Open
Abstract
The study of metabolic deregulation in myeloid malignancies has led to the investigation of metabolic-targeted therapies considering that cells undergoing leukemic transformation have excessive energy demands for growth and proliferation. However, the most difficult challenge in agents targeting metabolism is to determine a window of therapeutic opportunities between normal and neoplastic cells, considering that all or most of the metabolic pathways important for cancer ontogeny may also regulate physiological cell functions. Targeted therapies have used the properties of leukemic cells to produce altered metabolic products when mutated. This is the case of IDH1/2 mutations generating the abnormal conversion of α-ketoglutarate (KG) to 2-hydroxyglutarate, an oncometabolite inhibiting KG-dependent enzymes, such as the TET family of genes (pivotal in characterizing leukemia cells either by mutations, e.g., TET2, or by altered expression, e.g., TET1/2/3). Additional observations derive from the high sensitivity of leukemic cells to oxidative phosphorylation and its amelioration using BCL-2 inhibitors (Venetoclax) or by disrupting the mitochondrial respiration. More recently, nicotinamide metabolism has been described to mediate resistance to Venetoclax in patients with acute myeloid leukemia. Herein, we will provide an overview of the latest research on the link between metabolic pathways interactome and leukemogenesis with a comprehensive analysis of the metabolic consequences of driver genetic lesions and exemplificative druggable pathways.
Collapse
Affiliation(s)
- Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (S.P.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
- Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Simona Pagliuca
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (S.P.)
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (S.P.)
- Correspondence:
| |
Collapse
|
10
|
Koebley SR, Mikheikin A, Leslie K, Guest D, McConnell-Wells W, Lehman JH, Al Juhaishi T, Zhang X, Roberts CH, Picco L, Toor A, Chesney A, Reed J. Digital Polymerase Chain Reaction Paired with High-Speed Atomic Force Microscopy for Quantitation and Length Analysis of DNA Length Polymorphisms. ACS NANO 2020; 14:15385-15393. [PMID: 33169971 DOI: 10.1021/acsnano.0c05897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
DNA length polymorphisms are found in many serious diseases, and assessment of their length and abundance is often critical for accurate diagnosis. However, measuring their length and frequency in a mostly wild-type background, as occurs in many situations, remains challenging due to their variable and repetitive nature. To overcome these hurdles, we combined two powerful techniques, digital polymerase chain reaction (dPCR) and high-speed atomic force microscopy (HSAFM), to create a simple, rapid, and flexible method for quantifying both the size and proportion of DNA length polymorphisms. In our approach, individual amplicons from each dPCR partition are imaged and sized directly. We focused on internal tandem duplications (ITDs) located within the FLT3 gene, which are associated with acute myeloid leukemia and often indicative of a poor prognosis. In an analysis of over 1.5 million HSAFM-imaged amplicons from cell line and clinical samples containing FLT3-ITDs, dPCR-HSAFM returned the expected variant length and variant allele frequency, down to 5% variant samples. As a high-throughput method with single-molecule resolution, dPCR-HSAFM thus represents an advance in HSAFM analysis and a powerful tool for the diagnosis of length polymorphisms.
Collapse
Affiliation(s)
- Sean R Koebley
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Andrey Mikheikin
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Kevin Leslie
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Daniel Guest
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Wendy McConnell-Wells
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Joshua H Lehman
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Taha Al Juhaishi
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Xiaojie Zhang
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Catherine H Roberts
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Loren Picco
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Amir Toor
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Alden Chesney
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Jason Reed
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
11
|
Mancuso RI, Foglio MA, Olalla Saad ST. Artemisinin-type drugs for the treatment of hematological malignancies. Cancer Chemother Pharmacol 2020; 87:1-22. [PMID: 33141328 DOI: 10.1007/s00280-020-04170-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
Qinghaosu, known as artemisinin (ARS), has been for over two millennia, one of the most common herbs prescribed in traditional Chinese medicine (TCM). ARS was developed as an antimalarial drug and currently belongs to the established standard treatments of malaria as a combination therapy worldwide. In addition to the antimalarial bioactivity of ARS, anticancer activities have been shown both in vitro and in vivo. Like other natural products, ARS acts in a multi-specific manner also against hematological malignancies. The chemical structure of ARS is a sesquiterpene lactone, which contains an endoperoxide bridge essential for activity. The main mechanism of action of ARS and its derivatives (artesunate, dihydroartemisinin, artemether) toward leukemia, multiple myeloma, and lymphoma cells comprises oxidative stress response, inhibition of proliferation, induction of various types of cell death as apoptosis, autophagy, ferroptosis, inhibition of angiogenesis, and signal transducers, as NF-κB, MYC, amongst others. Therefore, new pharmaceutically active compounds, dimers, trimers, and hybrid molecules, could enhance the existing therapeutic alternatives in combating hematologic malignancies. Owing to the high potency and good tolerance without side effects of ARS-type drugs, combination therapies with standard chemotherapies could be applied in the future after further clinical trials in hematological malignancies.
Collapse
Affiliation(s)
- R I Mancuso
- Hematology and Hemotherapy Center, University of Campinas, HEMOCENTRO UNICAMP, Campinas, São Paulo, Brazil
| | - M A Foglio
- Faculty of Pharmaceutical Science, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - S T Olalla Saad
- Hematology and Hemotherapy Center, University of Campinas, HEMOCENTRO UNICAMP, Campinas, São Paulo, Brazil.
| |
Collapse
|
12
|
Zuo W, Zhou K, Deng M, Lin Q, Yin Q, Zhang C, Zhou J, Song Y. LINC00963 facilitates acute myeloid leukemia development by modulating miR-608/MMP-15. Aging (Albany NY) 2020; 12:18970-18981. [PMID: 33012724 PMCID: PMC7732318 DOI: 10.18632/aging.103252] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/20/2020] [Indexed: 01/24/2023]
Abstract
Despite continuous improvements of AML therapy, the prognosis of AML patients remains unsatisfactory. Recently, lncRNAs have been reported to participate in the development of AML. Our data demonstrated that MMP15 and LINC00963 were upregulated and miR-608 was decreased in AML cells (THP-1, HL-60, HEL and MOLM-13) compared to HS-5 cells. RT-qPCR results showed that LINC00963 levels were higher in the serum and bone marrow of AML cases than in controls. Moreover, overexpression of LINC00963 promoted AML cell growth and EMT progression in both THP-1 and HL-60 cells. Furthermore, miR-608 levels were downregulated in the serum and bone marrow of AML cases compared with controls, and Pearson's correlation analysis indicated that LINC00963 was negatively correlated with miR-608 in the serum and bone marrow of AML samples. In addition, we demonstrated that LINC00963 sponged miR-608 expression and that MMP-15 was a target of miR-608 in AML cells. Finally, rescue experiments indicated that ectopic expression of LINC00963 accelerated cell growth and EMT development by modulating MMP-15. These data demonstrated that LINC00963 acted as an oncogene and may be a potential target for AML treatment.
Collapse
Affiliation(s)
- Wenli Zuo
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| | - Keshu Zhou
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| | - Mei Deng
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| | - Quande Lin
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| | - Qingsong Yin
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| | - Chunlei Zhang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| | - Jian Zhou
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, China
| |
Collapse
|