1
|
Xie Z, Xia Y, Li Z, Zhang M, Tan Y, Han Y, Wang M, Zhang P, Li J. Integrated bioinformatics and functional studies identify CDK9 as a potential prognostic biomarker and therapeutic target in AML. Discov Oncol 2025; 16:1038. [PMID: 40490668 DOI: 10.1007/s12672-025-02841-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 05/28/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a highly heterogeneous disease characterized by complex genetic and molecular features that contribute to poor prognosis and low cure rates. Therefore, identifying novel therapeutic targets is crucial for improving treatment efficacy and patient survival. This study investigated the potential role of cyclin-dependent kinase 9 (CDK9), a known regulator of gene expression, in AML pathogenesis and prognosis. METHODS This study employed multiple bioinformatics approaches, including analysis of CDK9 expression across various cancers using the Tumor Immune Estimation Resource (TIMER2.0) database and further investigation of its expression and prognostic significance in AML using data from the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. Survival analysis and Cox regression analysis were used to assess the association between CDK9 expression and patient prognosis. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were performed to elucidate the pathways and biological processes influenced by CDK9. Furthermore, the relationship between CDK9 expression and tumor immune infiltration was evaluated, and a protein-protein interaction (PPI) network was constructed. In vitro experiments, including Western blotting, CCK-8 assays, and flow cytometry, were conducted to validate the bioinformatics findings. RESULTS Bioinformatics analysis revealed significantly elevated CDK9 expression in AML samples, which correlated with poor patient prognosis. Functional enrichment analysis indicated that CDK9 is involved in key pathways related to cell proliferation, differentiation, and the tumor microenvironment. Moreover, the study observed a strong correlation between CDK9 expression and altered immune cell infiltration, suggesting a potential role in immune evasion. In vitro experiments confirmed that CDK9 overexpression promoted AML cell proliferation and inhibited apoptosis. Additionally, CDK9 showed a strong correlation with epithelial-mesenchymal transition (EMT)-related proteins, suggesting a potential role in AML progression and the EMT process. CONCLUSIONS This study demonstrates that CDK9 is a potential prognostic biomarker and therapeutic target in AML. Its involvement in multiple key pathways during AML development and its influence on the tumor immune microenvironment support further exploration of CDK9-targeted therapies to improve AML treatment outcomes.
Collapse
Affiliation(s)
- Zhibin Xie
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Yang Xia
- Department of Finance, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhongyu Li
- Department of Hematology, The Third People's Hospital of Bengbu, Bengbu, Anhui, China
| | - Mengmeng Zhang
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Yuanyuan Tan
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Yuqing Han
- Bengbu Medical University, Bengbu, Anhui, China
| | - Meng Wang
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Pingping Zhang
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Jiajia Li
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China.
| |
Collapse
|
2
|
Xu P, Sun X, Pan L, Zhu J, Qian S. Disulfidptosis-related LncRNAs forecast the prognosis of acute myeloid leukemia. Sci Rep 2025; 15:13635. [PMID: 40254646 PMCID: PMC12009979 DOI: 10.1038/s41598-025-95607-5] [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: 07/24/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematologic malignancy with a poor prognosis for patients. Disulfidptosis response-related long non-coding RNAs (DRLs) have been demonstrated to be closely associated with cancer development. Therefore, this study aims to construct a prognostic DRL signature and investigate the immune microenvironment for AML. RNA-seq and clinical data for AML patients were obtained from The Cancer Genome Atlas (TCGA) database. A total of 344 disulfidptosis-associated lncRNAs were identified, and a prognostic model consisting of seven lncRNAs was constructed and validated. Two risk groups, high-risk and low-risk, were identified. The model demonstrated a robust capacity to predict prognosis, with a worse overall survival for patients in the high-risk group. Additionally, differential expression of the seven lncRNAs were relatively higher in AML samples than in control samples via quantitative polymerase chain reaction(qPCR). The Kyoto Encyclopedia of Genes and Genomes (KEGG) and immune infiltration analysis revealed a substantial infiltration of immune cells and enrichment of immune pathways in the high-risk group. The sensitivity of AML patients to drugs varied according to their risk grade. This study identified a DRL signature, which can effectively predict the prognosis of AML and better understand the mechanism of disulfidptosis in AML. This provides a basis for personalized immunotherapy in AML patients.
Collapse
Affiliation(s)
- Pei Xu
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Xiaolin Sun
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Lingxiao Pan
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Jianfeng Zhu
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China.
| | - Sixuan Qian
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China.
| |
Collapse
|
3
|
Liu S, Liu C, He Y, Li J. Benign non-immune cells in tumor microenvironment. Front Immunol 2025; 16:1561577. [PMID: 40248695 PMCID: PMC12003390 DOI: 10.3389/fimmu.2025.1561577] [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: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
The tumor microenvironment (TME) is a highly complex and continuous evolving ecosystem, consisting of a diverse array of cellular and non-cellular components. Among these, benign non-immune cells, including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells (ECs), pericytes (PCs), Schwann cells (SCs) and others, are crucial factors for tumor development. Benign non-immune cells within the TME interact with both tumor cells and immune cells. These interactions contribute to tumor progression through both direct contact and indirect communication. Numerous studies have highlighted the role that benign non-immune cells exert on tumor progression and potential tumor-promoting mechanisms via multiple signaling pathways and factors. However, these benign non-immune cells may play different roles across cancer types. Therefore, it is important to understand the potential roles of benign non-immune cells within the TME based on tumor heterogeneity. A deep understanding allows us to develop novel cancer therapies by targeting these cells. In this review, we will introduce several types of benign non-immune cells that exert on different cancer types according to tumor heterogeneity and their roles in the TME.
Collapse
Affiliation(s)
- Shaowen Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunhui Liu
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
4
|
Mohammed MH, Al-Aouadi RFA, Uthirapathy S, Sanghvi G, M M R, Abdulamer RS, Prasad GVS, Kundlas M, Khamraev F, Ullah MI. Therapeutic innovations: targeting ROS production in AML with natural and synthetic compounds. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04054-6. [PMID: 40163149 DOI: 10.1007/s00210-025-04054-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025]
Abstract
Reactive oxygen species (ROS) play a dual role in the pathophysiology of acute myeloid leukemia (AML), functioning as both signaling molecules and agents of cellular damage. This review offers an in-depth analysis of ROS production in AML, highlighting their impact on essential cellular pathways that govern cell survival, proliferation, and apoptosis. It explores both natural and synthetic pharmacological agents that modulate ROS generation and enhance oxidative stress, assessing their therapeutic potential and the challenges they present in clinical practice. Additionally, the review identifies ROS-associated prognostic biomarkers that could enhance patient stratification and improve treatment outcomes in AML. Despite the promising potential of ROS-targeted therapies, significant challenges remain, such as the complexity of ROS dynamics, resistance mechanisms, and the influence of the tumor microenvironment. This review aims to shed light on current advancements and emphasize the need for further research to refine therapeutic strategies that leverage the ROS pathway in AML.
Collapse
Affiliation(s)
- Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University, Anbar, Iraq.
| | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Resan Shakir Abdulamer
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | | | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, 72388, Sakaka, Aljouf, Saudi Arabia
| |
Collapse
|
5
|
Zhu B, Yang C, Hua S, Li K, Shang P, Li Z, Qian W, Xue S, Zhi Q, Hua Z. Decoding the Implications of Zinc in the Development and Therapy of Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412225. [PMID: 39887881 PMCID: PMC11884550 DOI: 10.1002/advs.202412225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/04/2025] [Indexed: 02/01/2025]
Abstract
Zinc plays a central role in the hematological development. Therapeutic interventions with zinc are shown to improve the health status of patients with malignancies by stimulating the immune system and reducing side effects. Despite the abnormal zinc homeostasis in leukemia, the role and mechanisms of zinc signaling in leukemia development remain poorly understood. Recently, some important breakthroughs are made in laboratory and clinical studies of zinc in leukemia, such as the role of zinc in regulating ferroptosis and the effects of zinc in immunotherapy. Zinc-based strategies are urgently needed to refine the current zinc intervention regimen for side-effect free therapy in chemotherapy-intolerant patients. This review provides a comprehensive overview of the role of zinc homeostasis in leukemia patients and focuses on the therapeutic potential of zinc signaling modulation in leukemia.
Collapse
Affiliation(s)
- Bo Zhu
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
| | - Chunhao Yang
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
| | - Siqi Hua
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
- Changzhou High‐tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc.Changzhou213164China
| | - Kaiqiang Li
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
- Changzhou High‐tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc.Changzhou213164China
| | - Pengyou Shang
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
- Changzhou High‐tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc.Changzhou213164China
| | - Zhonghua Li
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
| | - Wei Qian
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
| | - Shunkang Xue
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
| | - Qi Zhi
- Department of RadiologyAffiliated Hospital of Nanjing University of Chinese MedicineNanjing210029China
| | - Zichun Hua
- School of BiopharmacyChina Pharmaceutical UniversityNanjing211198China
- Changzhou High‐tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc.Changzhou213164China
- The State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjing210023China
- Faculty of Pharmaceutical SciencesXinxiang Medical UniversityXinxiang453003China
| |
Collapse
|
6
|
Luo C, Abudukeremu Y, Rao M, Zhou D, Fang J, Li Y, Xu L. A Novel Prognostic Risk-Scoring Model Based on RAS Gene-Associated Cluster in Pediatric Acute Myeloid Leukemia. Cancer Med 2025; 14:e70716. [PMID: 40062727 PMCID: PMC11891924 DOI: 10.1002/cam4.70716] [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: 09/23/2024] [Revised: 11/19/2024] [Accepted: 01/29/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND With the rapid development of diagnostic techniques and treatment strategies, there are notable improvements in pediatric acute myeloid leukemia (AML) prognosis. Nevertheless, the pathogenesis of AML remains largely unknown. This study aims to investigate the RAS pathway-associated genes based on bioinformatics analysis, and investigate their underlying mechanisms in the initiation and progression of AML. MATERIALS AND METHODS The UCSC Xena database was the source of the training set data, while the GSE192638 dataset was the source of the validation set data. Children in the training set were split up into two groups according to RAS pathway-associated genes, and then differentially expressed genes (DEGs) of them were screened. To discover prognosis-related genes and develop a prognostic risk-scoring model, we employed One-way Cox and LASSO regression analysis. The performance of the model was assessed by an independent validation dataset. Survival analysis was performed using the Kaplan-Meier (K-M) curve. Furthermore, we investigated the association between the prognostic risk-scoring model and the correlation between immune infiltration and drug sensitivity. The expression levels of genes associated with reverse transcription-polymerase chain reaction were quantified. RESULTS We built a prognostic risk-scoring model comprising 26 DEGs. Depending on the risk score, AML patients were split up into two groups: high- and low-risk groups. Notably, compared with the survival time of patients in the high- risk group, that in the low-risk group was substantially prolonged. Univariate (uniCox) as well as multivariate Cox (multiCox) regression analyses were carried out, demonstrating that the risk score emerged as a separate risk factor for prognosis. A nomogram that incorporates clinical factors and prognostic risk scores was proposed to increase the accuracy of survival rates estimation. Subsequent analyses revealed significant connections of the risk score with the immune infiltration and drug sensitivity. The experimental results demonstrated significantly elevated expression levels of GCSAML, MED12L, and TCF4 in AML samples compared to normal samples. CONCLUSION The developed prognostic risk-scoring model, along with the identified key risk genes, holds promise as candidate prognostic biomarkers and treatment targets for pediatric AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Prognosis
- Child
- Male
- Female
- Biomarkers, Tumor/genetics
- Gene Expression Profiling
- Child, Preschool
- Risk Assessment
- Genes, ras
- Computational Biology/methods
- Gene Expression Regulation, Leukemic
- Kaplan-Meier Estimate
- Adolescent
Collapse
Affiliation(s)
- Cai‐Ju Luo
- Department of Pediatrics, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education InstitutesSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yilimuguli Abudukeremu
- Department of PediatricsThe First People's Hospital of Kashi PrefectureKashiXinjiangChina
| | - Ming‐Liang Rao
- Department of Pediatrics, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education InstitutesSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Dun‐Hua Zhou
- Department of Pediatrics, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education InstitutesSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Jian‐Pei Fang
- Department of Pediatrics, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education InstitutesSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yang Li
- Department of Pediatrics, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education InstitutesSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Lu‐Hong Xu
- Department of Pediatrics, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education InstitutesSun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
7
|
Tsushima T, Kimeda C, Yoda N, Matsuo K, Tanaka K, Hatanaka Y, Matsumoto R, Shimoji S, Utsu Y, Masuda SI, Aotsuka N. Clinical Outcomes of Early WT1 mRNA Reduction After Remission Induction in Newly Diagnosed Acute Myeloid Leukemia Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2025; 31:168.e1-168.e12. [PMID: 39694194 DOI: 10.1016/j.jtct.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Wilms' Tumor 1 (WT1) mRNA is a non-specific marker of measurable residual disease in acute myeloid leukemia (AML). Few studies have focused on the prognostic value of WT1 mRNA after initial remission induction of patients with AML who have received transplant treatments. Thus, we retrospectively analyzed the clinical features and prognostic impact of WT1 mRNA reduction in patients with AML after initial remission induction at our hospital. We classified the reduction in WT1 mRNA levels using logarithmic stratification, with particular focus on the prognostic impact of a 3-log reduction after initial remission induction. This single-center, retrospective, observational study included 71 consecutive patients with AML who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) between April 2013 and June 2023 and had WT1 mRNA quantified. Patients were grouped based on whether a 3-log reduction was observed during follow-up (N=30) or not (N=41). Among patients who did not achieve a 3-log reduction, European Leukemia Net (ELN) 2022 adverse risk was more common, and fewer patients showed complete hematological responses at transplantation. Patients who reached a 3-log reduction in WT1 mRNA after the initial remission induction had significantly longer overall survival (OS) and progression-free survival (PFS) and a lower relapse rate than patients who had not reached a 3-log reduction (2-year OS: 79.7% vs. 27.5%, 2-year PFS: 83.1% vs. 11.7% and 2-year cumulative relapse rate: 5.9% vs. 81.2%). In multivariate analysis, a 3-log reduction in WT1 mRNA after initial remission induction and ELN 2022 adverse risk by genetics were significantly associated with OS and PFS. We identified that patients with AML undergoing HSCT with an early and deep 3-log reduction in WT1 mRNA after initial remission induction were associated with low relapse rates and better long-term prognosis. Our data highlight the importance of WT1 mRNA reduction after initial remission induction.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/diagnosis
- Hematopoietic Stem Cell Transplantation/methods
- Male
- Female
- WT1 Proteins/genetics
- WT1 Proteins/metabolism
- Adult
- Middle Aged
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Retrospective Studies
- Remission Induction
- Transplantation, Homologous
- Young Adult
- Adolescent
- Prognosis
- Treatment Outcome
- Aged
Collapse
Affiliation(s)
- Takafumi Tsushima
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan.
| | - Chiharu Kimeda
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Natsumi Yoda
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Kosuke Matsuo
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Kazusuke Tanaka
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Yasuhito Hatanaka
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Rena Matsumoto
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Sonoko Shimoji
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Yoshikazu Utsu
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Shin-Ichi Masuda
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Nobuyuki Aotsuka
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| |
Collapse
|
8
|
Reis R, Müller GS, Santos MM, Santos AS, Santos H, Santos LS, Lopes BA, Trindade SC, Meyer RJ, Freire SM. Description of lymphocyte and cytokine profiles in individuals with acute myeloid leukemia associated with FLT3-ITD and NPM1 mutation status. Eur J Cancer Prev 2025; 34:115-123. [PMID: 38904445 DOI: 10.1097/cej.0000000000000905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The pathogenesis of acute myeloid leukemia (AML) involves mutations in genes such as FLT3 and NPM1 , which are also associated with the prognosis of the disease. The immune system influences disease progression, but the mechanisms underlying the interaction between the immune system and AML are not clear. In this study, the profiles of lymphocytes and cytokines were described in individuals with AML stratified by molecular changes associated with prognosis. The participants included in this study were newly diagnosed AML patients ( n = 43) who were about to undergo chemotherapy. Subtypes of lymphocytes in peripheral blood, including B cells, T cells, and natural killer cells, and serum concentrations of cytokines, including Th1, Th2, and Th17, were studied by flow cytometry assays (BD FACSCanto II). The correlations between lymphocyte subsets, cytokines, and genetic/prognostic risk stratification (based on the FLT3 and NPM1 genes) were analyzed. The differences in B lymphocytes (%), T lymphocytes (%), plasmablasts (%), leukocytes (cells/µl), and tumor necrosis factor (pg/ml) were determined between groups with FLT3-ITD+ and FLT3-ITD- mutations. The presence of mutations in NPM1 and FLT3-ITD and age suggested changes in the lymphocyte and cytokine profile in individuals with AML.
Collapse
Affiliation(s)
- Rogério Reis
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
- Postgraduate Program in Immunology, Federal University of Bahia
| | - Gabriel S Müller
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
- Postgraduate Program in Immunology, Federal University of Bahia
| | - Mariane M Santos
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
- Postgraduate Program in Immunology, Federal University of Bahia
| | - Allan S Santos
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
- Postgraduate Program in Immunology, Federal University of Bahia
| | - Herbert Santos
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
- Professor Edgard Santos University Hospital, Salvador, BA
| | - Lorene S Santos
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
| | | | - Soraya C Trindade
- Departament of Health, State University of Feira de Santana, Feira de Santana
| | - Roberto J Meyer
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
- Postgraduate Program in Immunology, Federal University of Bahia
- Departament of Health, State University of Feira de Santana, Feira de Santana
| | - Songelí M Freire
- Immunology and Molecular Biology Laboratory, Federal University of Bahia
- Postgraduate Program in Immunology, Federal University of Bahia
- Department of Biointeraction, Federal University of Bahia, Salvador, BA, Brazil
| |
Collapse
|
9
|
Sokei J, Kanefsky J, Sykes SM. Reprogramming of Fatty Acid Metabolism in Acute Leukemia. J Cell Physiol 2025; 240:e70000. [PMID: 39835485 DOI: 10.1002/jcp.70000] [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: 05/08/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Fatty acids are essential biomolecules that support several cellular processes, such as membrane structures, energy storage and production, as well as signal transduction. Accordingly, changes in fatty acid metabolism can have a significant impact on cell behavior, such as growth, survival, proliferation, differentiation, and motility. Therefore, it is not surprising that many aspects of fatty acid metabolism are frequently dysregulated in human cancer, including in highly aggressive blood cancers such as acute leukemia. The aims of this review are to summarize the aspects of fatty acid metabolism that are specifically coopted in acute leukemia as well as current preclinical strategies for targeting fatty acid metabolism in these cancers.
Collapse
Affiliation(s)
- Judith Sokei
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Joice Kanefsky
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University Health System, Philadelphia, Pennsylvania, USA
| | - Stephen M Sykes
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University in Saint Louis, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Kuhestani-Dehaghi B, Amirpour M, Nabigol M, Keshavarz A, Vazifeh Shiran N, Rahmani-Seraji H, Dehghani-Ghorbi M, Allahbakhshian Farsani M. Evaluating the effect of acute myeloblastic leukemia-derived exosomes on the human bone marrow mesenchymal stromal cell proliferation. Mol Biol Rep 2024; 52:62. [PMID: 39692942 DOI: 10.1007/s11033-024-10155-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND The progression of leukemia is substantially associated with the interactions of leukemic cells with surrounding cells within the bone marrow microenvironment (BMM), and these interactions were facilitated using exosomes as vital mediators. The current study aimed to examine the proliferative effects of exosomes derived from the HL-60 cell line, a representative of acute myeloblastic leukemia (AML), on the cell cycle progression of human bone marrow mesenchymal stromal cells (hBM-MSCs), a key element of the BMM. METHODS AND RESULTS hBM-MSCs were treated with different concentrations of AML-derived exosomes from the HL-60 cell line. The results were obtained from MTT, cell proliferation, cell cycle, and RT-qPCR evaluations. In the current study, we found that the proliferation effects of AML-derived exosomes relied on the dose and the time, and the optimal effects of exosomes were seen in 50 μg/ml, 48 h treatment. Flow cytometry analysis revealed a significant increase in the G1 phase, showing a 1.6-fold change compared to the control group (p value < 0.0001). RT-qPCR results demonstrated a significant upregulation of CCND1 (3.3-fold, p value < 0.0001), CDK4 (3.7-fold, p value < 0.0001), CDK6 (3.3-fold, p value < 0.0001), RAS (3.2-fold, p value < 0.0001), and Erk (3.4-fold, p value < 0.0001) expression levels, along with increased Ki-67 (2.6-fold, p value < 0.0001) levels. Moreover, treatment with 50 μg/ml, 48 h of AML-derived exosomes resulted in a notable reduction in BM-MSC apoptosis both in early (p value < 0.0001) and late (p value < 0.0001) apoptosis rate compared to control group. CONCLUSIONS The findings will be of interest to AML-derived exosomes, which were able to potentiate the activation of the signaling pathways involved in the survival and proliferation of hBM-MSCs. Our findings suggest their specific targeting as a potential therapeutic strategy against cancer progression and metastasis.
Collapse
Affiliation(s)
- Bentolhoda Kuhestani-Dehaghi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 15468-15514, Tehran, Iran
| | - Mozhgan Amirpour
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 15468-15514, Tehran, Iran
| | - Maryam Nabigol
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 15468-15514, Tehran, Iran
| | - Ali Keshavarz
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 15468-15514, Tehran, Iran
| | - Nader Vazifeh Shiran
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 15468-15514, Tehran, Iran
| | - Hamideh Rahmani-Seraji
- Department of Hematology and Oncology, Taleghani Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mahmoud Dehghani-Ghorbi
- Department of Hematology and Oncology, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Allahbakhshian Farsani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 15468-15514, Tehran, Iran.
| |
Collapse
|
11
|
Chen T, Zhang Y, Zhang D, Zhou H. Immune-based subgroups uncover diverse tumor immunogenicity and implications for prognosis and precision therapy in acute myeloid leukemia. Front Immunol 2024; 15:1451486. [PMID: 39582863 PMCID: PMC11581856 DOI: 10.3389/fimmu.2024.1451486] [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: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/26/2024] Open
Abstract
Background Although a considerable proportion of acute myeloid leukemia (AML) patients achieve remission through chemotherapy, relapse remains a recurring and significant event leading to treatment failure. This study aims to investigate the immune landscape in AML and its potential implications for prognosis and chemo-/immune-therapy. Methods Integrated analyses based on multiple sequencing datasets of AML were performed. Various algorithms estimated immune infiltration in AML samples. A subgroup prediction model was developed, and comprehensive bioinformatics and machine learning algorithms were applied to compare immune-based subgroups in relation to clinical features, mutational landscapes, immune characterizations, drug sensitivities, and cellular hierarchies at the single-cell level. Results Two immune-based AML subgroups, G1 and G2, were identified. G1 demonstrated higher immune infiltration, a more monocytic phenotype, increased proportions of monocytes/macrophages, and higher FLT3, DNMT3A, and NPM1 mutation frequencies. It was associated with a poorer prognosis, lower proportions of various immune cell types and a lower T cell infiltration score (TIS). AML T-cell-based immunotherapy target antigens, including CLEC12A, Folate receptor β, IL1RAP and TIM3, showed higher expression levels in G1, while CD117, CD244, CD96, WT and TERT exhibited higher expression levels in G2. G1 samples demonstrated higher sensitivity to elesclomol and panobinostat but increased resistance to venetoclax compared to G2 samples. Moreover, we observed a positive correlation between sample immune infiltration and sample resistance to elesclomol and panobinostat, whereas a negative correlation was found with venetoclax resistance. Conclusion Our study enriches the current AML risk stratification and provides guidance for precision medicine in AML.
Collapse
Affiliation(s)
| | | | | | - Hebing Zhou
- Department of Hematology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Mu S, Zhao K, Zhong S, Wang Y. The Role of m6A Methylation in Tumor Immunity and Immune-Associated Disorder. Biomolecules 2024; 14:1042. [PMID: 39199429 PMCID: PMC11353047 DOI: 10.3390/biom14081042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
N6-methyladenosine (m6A) represents the most prevalent and significant internal modification in mRNA, with its critical role in gene expression regulation and cell fate determination increasingly recognized in recent research. The immune system, essential for defense against infections and maintaining internal stability through interactions with other bodily systems, is significantly influenced by m6A modification. This modification acts as a key post-transcriptional regulator of immune responses, though its effects on different immune cells vary across diseases. This review delineates the impact of m6A modification across major system-related cancers-including those of the respiratory, digestive, endocrine, nervous, urinary reproductive, musculoskeletal system malignancies, as well as acute myeloid leukemia and autoimmune diseases. We explore the pathogenic roles of m6A RNA modifications within the tumor immune microenvironment and the broader immune system, highlighting how RNA modification regulators interact with immune pathways during disease progression. Furthermore, we discuss how the expression patterns of these regulators can influence disease susceptibility to immunotherapy, facilitating the development of diagnostic and prognostic models and pioneering new therapeutic approaches. Overall, this review emphasizes the challenges and prospective directions of m6A-related immune regulation in various systemic diseases throughout the body.
Collapse
Affiliation(s)
- Siyu Mu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110000, China; (S.M.); (S.Z.)
| | - Kaiyue Zhao
- Department of Hepatology, Beijing Tsinghua Changgeng Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China;
| | - Shanshan Zhong
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110000, China; (S.M.); (S.Z.)
| | - Yanli Wang
- Department of Infectious Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110000, China
| |
Collapse
|
13
|
Tong Z, Yin Z. Distribution, contribution and regulation of nestin + cells. J Adv Res 2024; 61:47-63. [PMID: 37648021 PMCID: PMC11258671 DOI: 10.1016/j.jare.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Nestin is an intermediate filament first reported in neuroepithelial stem cells. Nestin expression could be found in a variety of tissues throughout all systems of the body, especially during tissue development and tissue regeneration processes. AIM OF REVIEW This review aimed to summarize and discuss current studies on the distribution, contribution and regulation of nestin+ cells in different systems of the body, to discuss the feasibility ofusing nestin as a marker of multilineage stem/progenitor cells, and better understand the potential roles of nestin+ cells in tissue development, regeneration and pathological processes. KEY SCIENTIFIC CONCEPTS OF REVIEW This review highlights the potential of nestin as a marker of multilineage stem/progenitor cells, and as a key factor in tissue development and tissue regeneration. The article discussed the current findings, limitations, and potential clinical implications or applications of nestin+ cells. Additionally, it included the relationship of nestin+ cells to other cell populations. We propose potential future research directions to encourage further investigation in the field.
Collapse
Affiliation(s)
- Ziyang Tong
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zi Yin
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
14
|
Travaglini S, Marinoni M, Visconte V, Guarnera L. Therapy-Related Myeloid Neoplasm: Biology and Mechanistic Aspects of Malignant Progression. Biomedicines 2024; 12:1054. [PMID: 38791019 PMCID: PMC11118122 DOI: 10.3390/biomedicines12051054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Therapy-related myeloid neoplasms (t-MN) arise after a documented history of chemo/radiotherapy as treatment for an unrelated condition and account for 10-20% of myelodysplastic syndromes and acute myeloid leukemia. T-MN are characterized by a specific genetic signature, aggressive features and dismal prognosis. The nomenclature and the subsets of these conditions have changed frequently over time, and despite the fact that, in the last classification, they lost their autonomous entity status and became disease qualifiers, the recognition of this feature remains of major importance. Furthermore, in recent years, extensive studies focusing on clonal hematopoiesis and germline variants shed light on the mechanisms of positive pressure underpinning the rise of driver gene mutations in t-MN. In this manuscript, we aim to review the evolution of defining criteria and characteristics of t-MN from a clinical and biological perspective, the advances in mechanistic aspects of malignant progression and the challenges in prevention and management.
Collapse
Affiliation(s)
- Serena Travaglini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Massimiliano Marinoni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Luca Guarnera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
15
|
Guijarro-Albaladejo B, Marrero-Cepeda C, Rodríguez-Arbolí E, Sierro-Martínez B, Pérez-Simón JA, García-Guerrero E. Chimeric antigen receptor (CAR) modified T Cells in acute myeloid leukemia: limitations and expectations. Front Cell Dev Biol 2024; 12:1376554. [PMID: 38694825 PMCID: PMC11061469 DOI: 10.3389/fcell.2024.1376554] [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: 01/25/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with a poor prognosis despite the advent of novel therapies. Consequently, a major need exists for new therapeutic options, particularly for patients with relapsed/refractory (R/R) AML. In recent years, it has been possible to individualize the treatment of a subgroup of patients, particularly with the emergence of multiple targeted therapies. Nonetheless, a considerable number of patients remain without therapeutic options, and overall prognosis remains poor because of a high rate of disease relapse. In this sense, cellular therapies, especially chimeric antigen receptor (CAR)-T cell therapy, have dramatically shifted the therapeutic options for other hematologic malignancies, such as diffuse large B cell lymphoma and acute lymphoblastic leukemia. In contrast, effectively treating AML with CAR-based immunotherapy poses major biological and clinical challenges, most of them derived from the unmet need to identify target antigens with expression restricted to the AML blast without compromising the viability of the normal hematopoietic stem cell counterpart. Although those limitations have hampered CAR-T cell therapy translation to the clinic, there are several clinical trials where target antigens, such as CD123, CLL-1 or CD33 are being used to treat AML patients showing promising results. Moreover, there are continuing efforts to enhance the specificity and efficacy of CAR-T cell therapy in AML. These endeavors encompass the exploration of novel avenues, including the development of dual CAR-T cells and next-generation CAR-T cells, as well as the utilization of gene editing tools to mitigate off-tumor toxicities. In this review, we will summarize the ongoing clinical studies and the early clinical results reported with CAR-T cells in AML, as well as highlight CAR-T cell limitations and the most recent approaches to overcome these barriers. We will also discuss how and when CAR-T cells should be used in the context of AML.
Collapse
Affiliation(s)
- Beatriz Guijarro-Albaladejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Cristina Marrero-Cepeda
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Eduardo Rodríguez-Arbolí
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Belén Sierro-Martínez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - José Antonio Pérez-Simón
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Estefanía García-Guerrero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| |
Collapse
|
16
|
Fiorcari S, Strati P, Dondi E. Editorial: Tumor microenvironment and hematological malignancies: new evidences and new questions. Front Immunol 2024; 15:1407981. [PMID: 38690284 PMCID: PMC11059779 DOI: 10.3389/fimmu.2024.1407981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Affiliation(s)
- Stefania Fiorcari
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Paolo Strati
- Department of Lymphoma and Myeloma & Department of Translational Molecular Pathology The University of Texas MD (UT MD) Anderson Cancer Center, Houston, TX, United States
| | - Elisabetta Dondi
- U978 Institut National de la Santé et de la Recherche Médicale/Université Sorbonne Paris Nord, Labex INFLAMEX, Bobigny, France
| |
Collapse
|
17
|
Azizidoost S, Nasrolahi A, Sheykhi-Sabzehpoush M, Anbiyaiee A, Khoshnam SE, Farzaneh M, Uddin S. Signaling pathways governing the behaviors of leukemia stem cells. Genes Dis 2024; 11:830-846. [PMID: 37692500 PMCID: PMC10491880 DOI: 10.1016/j.gendis.2023.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/02/2023] [Indexed: 08/28/2023] Open
Abstract
Leukemia is a malignancy in the blood that develops from the lymphatic system and bone marrow. Although various treatment options have been used for different types of leukemia, understanding the molecular pathways involved in the development and progression of leukemia is necessary. Recent studies showed that leukemia stem cells (LSCs) play essential roles in the pathogenesis of leukemia by targeting several signaling pathways, including Notch, Wnt, Hedgehog, and STAT3. LSCs are highly proliferative cells that stimulate tumor initiation, migration, EMT, and drug resistance. This review summarizes cellular pathways that stimulate and prevent LSCs' self-renewal, metastasis, and tumorigenesis.
Collapse
Affiliation(s)
- Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Mohadeseh Sheykhi-Sabzehpoush
- Department of Laboratory, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran 2193672411, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
18
|
Wu Y, Li Y, Gao Y, Zhang P, Jing Q, Zhang Y, Jin W, Wang Y, Du J, Wu G. Immunotherapies of acute myeloid leukemia: Rationale, clinical evidence and perspective. Biomed Pharmacother 2024; 171:116132. [PMID: 38198961 DOI: 10.1016/j.biopha.2024.116132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is a prevalent hematological malignancy that exhibits a wide array of molecular abnormalities. Although traditional treatment modalities such as chemotherapy and allogeneic stem cell transplantation (HSCT) have become standard therapeutic approaches, a considerable number of patients continue to face relapse and encounter a bleak prognosis. The emergence of immune escape, immunosuppression, minimal residual disease (MRD), and other contributing factors collectively contribute to this challenge. Recent research has increasingly highlighted the notable distinctions between AML tumor microenvironments and those of healthy individuals. In order to investigate the potential therapeutic mechanisms, this study examines the intricate transformations occurring between leukemic cells and their surrounding cells within the tumor microenvironment (TME) of AML. This review classifies immunotherapies into four distinct categories: cancer vaccines, immune checkpoint inhibitors (ICIs), antibody-based immunotherapies, and adoptive T-cell therapies. The results of numerous clinical trials strongly indicate that the identification of optimal combinations of novel agents, either in conjunction with each other or with chemotherapy, represents a crucial advancement in this field. In this review, we aim to explore the current and emerging immunotherapeutic methodologies applicable to AML patients, identify promising targets, and emphasize the crucial requirement to augment patient outcomes. The application of these strategies presents substantial therapeutic prospects within the realm of precision medicine for AML, encompassing the potential to ameliorate patient outcomes.
Collapse
Affiliation(s)
- Yunyi Wu
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Yan Gao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiangan Jing
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yinhao Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weidong Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Gongqiang Wu
- Department of Hematology, Dongyang Hospitai Affiliated to Wenzhou Medical University, Dongyang People's Hospital, Dongyang, Zhejiang, China.
| |
Collapse
|
19
|
Guarnera L, Santinelli E, Galossi E, Cristiano A, Fabiani E, Falconi G, Voso MT. Microenvironment in acute myeloid leukemia: focus on senescence mechanisms, therapeutic interactions, and future directions. Exp Hematol 2024; 129:104118. [PMID: 37741607 DOI: 10.1016/j.exphem.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023]
Abstract
Acute myeloid leukemia (AML) is a disease with a dismal prognosis, mainly affecting the elderly. In recent years, new drugs have improved life expectancy and quality of life, and a better understanding of the genetic-molecular nature of the disease has shed light on previously unknown aspects of leukemogenesis. In parallel, increasing attention has been attracted to the complex interactions between cells and soluble factors in the bone marrow (BM) environment, collectively known as the microenvironment. In this review, we discuss the central role of the microenvironment in physiologic and pathologic hematopoiesis and the mechanisms of senescence, considered a fundamental protective mechanism against the proliferation of damaged and pretumoral cells. The microenvironment also represents a fertile ground for the development of myeloid malignancies, and the leukemic niche significantly interacts with drugs commonly used in AML treatment. Finally, we focus on the role of the microenvironment in the engraftment and complications of allogeneic hematopoietic stem cell transplantation, the only curative option in a conspicuous proportion of patients.
Collapse
Affiliation(s)
- Luca Guarnera
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Enrico Santinelli
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Elisa Galossi
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Antonio Cristiano
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Emiliano Fabiani
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy; Saint Camillus International, University of Health Sciences, Rome, Italy
| | - Giulia Falconi
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy; Neuro-Oncohematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
20
|
Chen XY, Wen XM, Zhao W, Chu MQ, Gu Y, Huang HH, Yuan Q, Xu ZJ, Qian J, Lin J. ALOX5AP is a new prognostic indicator in acute myeloid leukemia. Discov Oncol 2023; 14:210. [PMID: 37994961 PMCID: PMC10667204 DOI: 10.1007/s12672-023-00826-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND The overexpression of ALOX5AP has been observed in many types of cancer and has been identified as an oncogene. However, its role in acute myeloid leukemia (AML) has not been extensively studied. This study aimed to identify the expression and methylation patterns of ALOX5AP in bone marrow (BM) samples of AML patients, and further explore its clinical significance. METHODS Eighty-two de novo AML patients and 20 healthy donors were included in the study. Meanwhile, seven public datasets from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) were included to confirm the alteration of ALOX5AP. Receiver operating characteristic (ROC) curve analysis was applied to determine the discriminative capacity of ALOX5AP expression to discriminate AML. The prognostic value of ALOX5AP was identified by the Kaplan-Meier method and log-rank test. It was further validated in four independent cohorts (n = 1186). Significantly different genes associated with ALOX5AP expression were subsequently compared by LinkedOmics, and Metascape database. RESULTS The level of ALOX5AP expression was significantly increased in bone marrow cells of AML patients compared with healthy donors (P < 0.05). ROC curve analysis suggested that ALOX5AP expression might be a potential biomarker to discriminate AML from controls. ALOX5AP overexpression was associated with decreased overall survival (OS) in AML according to the TCGA data (P = 0.006), which was validated by other four independent cohorts. DNA methylation levels of ALOX5AP were significantly lower in AML patients compared to normal samples (P < 0.05), as confirmed in the Diseasemeth database and the independent cohort GSE63409. ALOX5AP level was positively associated with genes with proleukemic effects such as PAX2, HOX family, SOX11, H19, and microRNAs that act as oncogenes in leukemia, such as miR125b, miR-93, miR-494, miR-193b, while anti-leukemia-related genes and tumor suppressor microRNAs such as miR-582, miR-9 family and miR-205 were negatively correlated. CONCLUSION ALOX5AP overexpression, associated with its hypomethylation, predicts poorer prognosis in AML.
Collapse
Affiliation(s)
- Xin-Yi Chen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China
| | - Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China
| | - Wei Zhao
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ming-Qiang Chu
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Gu
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hai-Hui Huang
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China
| | - Qian Yuan
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China.
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China.
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, China.
| |
Collapse
|
21
|
Pendse S, Chavan S, Kale V, Vaidya A. A comprehensive analysis of cell-autonomous and non-cell-autonomous regulation of myeloid leukemic cells: The prospect of developing novel niche-targeting therapies. Cell Biol Int 2023; 47:1667-1683. [PMID: 37554060 DOI: 10.1002/cbin.12078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/12/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023]
Abstract
Leukemic cells (LCs) arise from the hematopoietic stem/and progenitor cells (HSCs/HSPCs) and utilize cues from the bone marrow microenvironment (BMM) for their regulation in the same way as their normal HSC counterparts. Mesenchymal stromal cells (MSCs), a vital component of the BMM promote leukemogenesis by creating a protective and immune-tolerant microenvironment that can support the survival of LCs, helping them escape chemotherapy, thereby resulting in the relapse of leukemia. Conversely, MSCs also induce apoptosis in the LCs and inhibit their proliferation by interfering with their self-renewal potential. This review discusses the work done so far on cell-autonomous (intrinsic) and MSCs-mediated non-cell-autonomous (extrinsic) regulation of myeloid leukemia with a special focus on the need to investigate the extrinsic regulation of myeloid leukemia to understand the contrasting role of MSCs in leukemogenesis. These mechanisms could be exploited to formulate novel therapeutic strategies that specifically target the leukemic microenvironment.
Collapse
Affiliation(s)
- Shalmali Pendse
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Sayali Chavan
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Vaijayanti Kale
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| | - Anuradha Vaidya
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra, India
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, Maharashtra, India
| |
Collapse
|
22
|
Pan C, Hu T, Liu P, Ma D, Cao S, Shang Q, Zhang L, Chen Q, Fang Q, Wang J. BM-MSCs display altered gene expression profiles in B-cell acute lymphoblastic leukemia niches and exert pro-proliferative effects via overexpression of IFI6. J Transl Med 2023; 21:593. [PMID: 37670388 PMCID: PMC10478283 DOI: 10.1186/s12967-023-04464-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND The tumor microenvironment (TME) is a supportive environment responsible for promoting the growth and proliferation of tumor cells. Current studies have revealed that the bone marrow mesenchymal stem cells (BM-MSCs), a type of crucial stromal cells in the TME, can promote the malignant progression of tumors. However, in the adult B-cell acute lymphoblastic leukemia (B-ALL) microenvironment, it is still uncertain what changes in BM-MSCs are induced by leukemia cells. METHODS In this study, we mimicked the leukemia microenvironment by constructing a BM-MSC-leukemia cell co-culture system. In vitro cell experiments, in vivo mouse model experiments, lentiviral transfection and transcriptome sequencing analysis were used to investigate the possible change of BM-MSCs in the leukemia niche and the potential factors in BM-MSCs that promote the progression of leukemia. RESULTS In the leukemia niche, the leukemia cells reduced the MSCs' capacity to differentiate towards adipogenic and osteogenic subtypes, which also promoted the senescence and cell cycle arrest of the MSCs. Meanwhile, compared to the mono-cultured MSCs, the gene expression profiles of MSCs in the leukemia niche changed significantly. These differential genes were enriched for cell cycle, cell differentiation, DNA replication, as well as some tumor-promoting biofunctions including protein phosphorylation, cell migration and angiogenesis. Further, interferon alpha-inducible protein 6 (IFI6), as a gene activated by interferon, was highly expressed in leukemia niche MSCs. The leukemia cell multiplication was facilitated evidently by IFI6 both in vitro and in vivo. Mechanistically, IFI6 might promote leukemia cell proliferation by stimulating SDF-1/CXCR4 axis, which leads to the initiation of downstream ERK signaling pathway. As suggested by further RNA sequencing analysis, the high IFI6 level in MSCs somewhat influenced the gene expression profile and biological functions of leukemia cells. CONCLUSIONS BM-MSCs in the leukemia niche have varying degrees of changes in biological characteristics and gene expression profiles. Overexpression of IFI6 in BM-MSCs could be a key factor in promoting the proliferation of B-ALL cells, and this effect might be exerted through the SDF-1/CXCR4/ERK signal stimulation. Targeting IFI6 or related signaling pathways might be an important measure to reduce the leukemia cell proliferation.
Collapse
Affiliation(s)
- Chengyun Pan
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
- School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China
- Hematological Institute of Guizhou Province, Guizhou, China
| | - Tianzhen Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
- Hematological Institute of Guizhou Province, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
- Hematological Institute of Guizhou Province, Guizhou, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
- Hematological Institute of Guizhou Province, Guizhou, China
- Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Shuyun Cao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
- Hematological Institute of Guizhou Province, Guizhou, China
| | - Qin Shang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
| | - Luxin Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
- Hematological Institute of Guizhou Province, Guizhou, China
| | - Qingzhen Chen
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
- Hematological Institute of Guizhou Province, Guizhou, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, 28 Guiyi St., Yunyan District, Guiyang, 550004, Guizhou, China.
- School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China.
- Hematological Institute of Guizhou Province, Guizhou, China.
- Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China.
| |
Collapse
|
23
|
Wang N. Analysis of prognostic biomarker models and immune microenvironment in acute myeloid leukemia by integrative bioinformatics. J Cancer Res Clin Oncol 2023; 149:9609-9619. [PMID: 37222809 DOI: 10.1007/s00432-023-04871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological cancer driven on by aberrant myeloid precursor cell proliferation and differentiation. A prognostic model was created in this study to direct therapeutic care. METHODS Differentially expressed genes (DEGs) were investigated using the RNA-seq data from the TCGA-LAML and GTEx. Weighted Gene Coexpression Network Analysis (WGCNA) examines the genes involved in cancer. Find the intersection genes and construct the PPI network to discover hub genes and remove prognosis-related genes. A nomogram was produced for predicting the prognosis of AML patients using the risk prognosis model that was constructed using COX and Lasso regression analysis. GO, KEGG, and ssGSEA analysis were used to look into its biological function. TIDE score predicts immunotherapy response. RESULTS Differentially expressed gene analysis revealed 1004 genes, WGCNA analysis revealed 19,575 tumor-related genes, and 941 intersection genes in total. Twelve prognostic genes were found using the PPI network and prognostic analysis. To build a risk rating model, RPS3A and PSMA2 were examined using COX and Lasso regression analysis. The risk score was used to divide the patients into two groups, and Kaplan-Meier analysis indicated that the two groups had different overall survival rates. Univariate and multivariate COX studies demonstrated that risk score is an independent prognostic factor. According to the TIDE study, the immunotherapy response was better in the low-risk group than in the high-risk group. CONCLUSIONS We eventually selected out two molecules to construct prediction models that might be used as biomarkers for predicting AML immunotherapy and prognosis.
Collapse
Affiliation(s)
- Naihong Wang
- Lanzhou University Second Hospital, Gansu, 730000, China.
| |
Collapse
|
24
|
Bahrami E, Schmid JP, Jurinovic V, Becker M, Wirth AK, Ludwig R, Kreissig S, Duque Angel TV, Amend D, Hunt K, Öllinger R, Rad R, Frenz JM, Solovey M, Ziemann F, Mann M, Vick B, Wichmann C, Herold T, Jayavelu AK, Jeremias I. Combined proteomics and CRISPR‒Cas9 screens in PDX identify ADAM10 as essential for leukemia in vivo. Mol Cancer 2023; 22:107. [PMID: 37422628 PMCID: PMC10329331 DOI: 10.1186/s12943-023-01803-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 06/08/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Acute leukemias represent deadly malignancies that require better treatment. As a challenge, treatment is counteracted by a microenvironment protecting dormant leukemia stem cells. METHODS To identify responsible surface proteins, we performed deep proteome profiling on minute numbers of dormant patient-derived xenograft (PDX) leukemia stem cells isolated from mice. Candidates were functionally screened by establishing a comprehensive CRISPR‒Cas9 pipeline in PDX models in vivo. RESULTS A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) was identified as an essential vulnerability required for the survival and growth of different types of acute leukemias in vivo, and reconstitution assays in PDX models confirmed the relevance of its sheddase activity. Of translational importance, molecular or pharmacological targeting of ADAM10 reduced PDX leukemia burden, cell homing to the murine bone marrow and stem cell frequency, and increased leukemia response to conventional chemotherapy in vivo. CONCLUSIONS These findings identify ADAM10 as an attractive therapeutic target for the future treatment of acute leukemias.
Collapse
Affiliation(s)
- Ehsan Bahrami
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
| | - Jan Philipp Schmid
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Vindi Jurinovic
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Martin Becker
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
| | - Anna-Katharina Wirth
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
| | - Romina Ludwig
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Sophie Kreissig
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Tania Vanessa Duque Angel
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
| | - Diana Amend
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
| | - Katharina Hunt
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
| | - Rupert Öllinger
- Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine, and Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany
| | - Roland Rad
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine, and Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany
| | - Joris Maximilian Frenz
- Proteomics and Cancer Cell Signaling Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, and Immunology, University of Heidelberg and Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
| | - Maria Solovey
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- Chair of Physiological Chemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Frank Ziemann
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Munich, Germany
| | - Binje Vick
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Tobias Herold
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Ashok Kumar Jayavelu
- Proteomics and Cancer Cell Signaling Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, and Immunology, University of Heidelberg and Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Munich, Germany
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, Feodor-Lynen-Str. 21, Munich, 81377 Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Department of Pediatrics, Dr. Von Hauner Children’s Hospital, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
25
|
Shimizu K, Ueda S, Kawamura M, Aoshima H, Satoh M, Nakabayashi J, Fujii SI. Combination of cancer vaccine with CD122-biased IL-2/anti-IL-2 Ab complex shapes the stem-like effector NK and CD8 + T cells against tumor. J Immunother Cancer 2023; 11:e006409. [PMID: 37400134 PMCID: PMC10577731 DOI: 10.1136/jitc-2022-006409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND A key to success of cancer immunotherapy is the amplification and sustenance of various effector cells. The hallmark of prominent antitumor T cells is their long-term effector function. Although interleukin (IL)-2 is an attractive cytokine, several attempts have been made towards developing IL-2 modalities with improved effectiveness and safety that enhance natural killer (NK) cells or T cells in cancer models. However, whether such IL-2 modalities can simultaneously support long-term innate and adaptive immunity, particularly stem-like memory, has not been shown. To resolve this issue, we compared the antitumor cellular mechanism with two IL-2/anti-IL-2 complexes (IL-2Cxs) administered in combination with a therapeutic cancer vaccine, which we had previously established as an in vivo dendritic cell-targeting therapy. METHODS Two types of IL-2Cxs, CD25-biased IL-2Cx and CD122-biased IL-2Cx, together with a Wilms' tumor 1-expressing vaccine, were evaluated in a leukemic model. The immunological response and synergistic antitumor efficacy of these IL-2Cxs were then evaluated. RESULTS When CD25-biased or CD122-biased IL-2Cxs in combination with the vaccine were assessed in an advanced-leukemia model, the CD122-biased IL-2Cx combination showed 100% survival, but the CD25-biased IL-2Cx did not. We first showed that invariant natural killer T (NKT) 1 cells are predominantly activated by CD122-biased IL-2Cx. In addition, in-depth analysis of immune responses by CD122-biased IL-2Cx in lymphoid tissues and the tumor microenvironment revealed a dramatic increase in the distinct subsets of NK and CD8+ T cells with stem-like phenotype (CD27+Sca-1hi, CXCR3hi, CD127+TCF-1+T-bet+ Eomes+). Moreover, CD122-biased IL-2Cx combination therapy maintained long-term memory CD8+ T cells capable of potent antitumor protection. After the high dimensional profiling analysis of NK and CD8+T cells, principal component analysis revealed that the stem-like-NK cell and stem-like-CD8+T cell state in the combination were integrated in the same group. CONCLUSIONS CD122-biased IL-2Cx combined with the vaccine can induce a series of reactions in the immune cascade, including activation of not only NKT1 cells, but also NK and CD8+ T cells with a stem-like memory phenotype. Since it can also lead to a long-term, strong antitumor response, the combination of CD122-biased IL-2Cx with a vaccine may serve as a potential and competent strategy for patients with advanced cancer.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Program for Drug Discovery and Medical Technology Platforms, RIKEN, Yokohama, Japan
| | - Shogo Ueda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masami Kawamura
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Honoka Aoshima
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mikiko Satoh
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Jun Nakabayashi
- Department of Mathematics, Tokyo Medical and Dental University, Ichikawa, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Program for Drug Discovery and Medical Technology Platforms, RIKEN, Yokohama, Japan
| |
Collapse
|
26
|
Aru B, Pehlivanoğlu C, Dal Z, Dereli-Çalışkan NN, Gürlü E, Yanıkkaya-Demirel G. A potential area of use for immune checkpoint inhibitors: Targeting bone marrow microenvironment in acute myeloid leukemia. Front Immunol 2023; 14:1108200. [PMID: 36742324 PMCID: PMC9895857 DOI: 10.3389/fimmu.2023.1108200] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Acute myeloid leukemia (AML) arises from the cells of myeloid lineage and is the most frequent leukemia type in adulthood accounting for about 80% of all cases. The most common treatment strategy for the treatment of AML includes chemotherapy, in rare cases radiotherapy and stem cell and bone marrow transplantation are considered. Immune checkpoint proteins involve in the negative regulation of immune cells, leading to an escape from immune surveillance, in turn, causing failure of tumor cell elimination. Immune checkpoint inhibitors (ICIs) target the negative regulation of the immune cells and support the immune system in terms of anti-tumor immunity. Bone marrow microenvironment (BMM) bears various blood cell lineages and the interactions between these lineages and the noncellular components of BMM are considered important for AML development and progression. Administration of ICIs for the AML treatment may be a promising option by regulating BMM. In this review, we summarize the current treatment options in AML treatment and discuss the possible application of ICIs in AML treatment from the perspective of the regulation of BMM.
Collapse
Affiliation(s)
- Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Zeynep Dal
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | | | - Ege Gürlü
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Gülderen Yanıkkaya-Demirel
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye,*Correspondence: Gülderen Yanıkkaya-Demirel,
| |
Collapse
|
27
|
Zhang Q, Ma R, Chen H, Guo W, Li Z, Xu K, Chen W. CD86 Is Associated with Immune Infiltration and Immunotherapy Signatures in AML and Promotes Its Progression. JOURNAL OF ONCOLOGY 2023; 2023:9988405. [PMID: 37064861 PMCID: PMC10104747 DOI: 10.1155/2023/9988405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/21/2022] [Accepted: 07/25/2022] [Indexed: 04/18/2023]
Abstract
Background Cluster of differentiation 86 (CD86), also known as B7-2, is a molecule expressed on antigen-presenting cells that provides the costimulatory signals required for T cell activation and survival. CD86 binds to two ligands on the surface of T cells: the antigen CD28 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). By binding to CD28, CD86-together with CD80-promotes the participation of T cells in the antigen presentation process. However, the interrelationships among CD86, immunotherapy, and immune infiltration in acute myeloid leukemia (AML) are unclear. Methods The immunological effects of CD86 in various cancers (including on chemokines, immunostimulators, MHC, and receptors) were evaluated through a pan-cancer analysis using TCGA and GEO databases. The relationship between CD86 expression and mononucleotide variation, gene copy number variation, methylation, immune checkpoint blockers (ICBs), and T-cell inflammation score in AML was subsequently examined. ESTIMATE and limma packages were used to identify genes at the intersection of CD86 with StromalScore and ImmuneScore. Subsequently, GO/KEGG and PPI network analyses were performed. The immune risk score (IRS) model was constructed, and the validation set was used for verification. The predictive value was compared with the TIDE score. Results CD86 was overexpressed in many cancers, and its overexpression was associated with a poor prognosis. CD86 expression was positively correlated with the expression of CTLA4, PDCD1LG2, IDO1, HAVCR2, and other genes and negatively correlated with CD86 methylation. The expression of CD86 in AML cell lines was detected by QRT-PCR and Western blot, and the results showed that CD86 was overexpressed in AML cell lines. Immune infiltration assays showed that CD86 expression was positively correlated with CD8 T cell, Dendritic cell, macrophage, NK cell, and Th1_cell and also with immune examination site, immune regulation, immunotherapy response, and TIICs. ssGSEA showed that CD86 was enriched in immune-related pathways, and CD86 expression was correlated with mutations in the genes RB1, ERBB2, and FANCC, which are associated with responses to radiotherapy and chemotherapy. The IRS score performed better than the TIDE website score. Conclusion CD86 appears to participate in immune invasion in AML and is an important player in the tumor microenvironment in this malignancy. At the same time, the IRS score developed by us has a good effect and may provide some support for the diagnosis of AML. Thus, CD86 may serve as a potential target for AML immunotherapy.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruixue Ma
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huimin Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wentong Guo
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Wei Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
- Department of Hematology, The First People's Hospital of Suqian, Suqian, Jiangsu, China
| |
Collapse
|
28
|
Nuclear factor Nrf2 promotes glycosidase OGG1 expression by activating the AKT pathway to enhance leukemia cell resistance to cytarabine. J Biol Chem 2022; 299:102798. [PMID: 36528059 PMCID: PMC9823221 DOI: 10.1016/j.jbc.2022.102798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
Chemotherapy resistance is the dominant challenge in the treatment of acute myeloid leukemia (AML). Nuclear factor E2-related factor 2 (Nrf2) exerts a vital function in drug resistance of many tumors. Nevertheless, the potential molecular mechanism of Nrf2 regulating the base excision repair pathway that mediates AML chemotherapy resistance remains unclear. Here, in clinical samples, we found that the high expression of Nrf2 and base excision repair pathway gene encoding 8-hydroxyguanine DNA glycosidase (OGG1) was associated with AML disease progression. In vitro, Nrf2 and OGG1 were highly expressed in drug-resistant leukemia cells. Upregulation of Nrf2 in leukemia cells by lentivirus transfection could decrease the sensitivity of leukemia cells to cytarabine, whereas downregulation of Nrf2 in drug-resistant cells could enhance leukemia cell chemosensitivity. Meanwhile, we found that Nrf2 could positively regulate OGG1 expression in leukemia cells. Our chromatin immunoprecipitation assay revealed that Nrf2 could bind to the promoter of OGG1. Furthermore, the use of OGG1 inhibitor TH5487 could partially reverse the inhibitory effect of upregulated Nrf2 on leukemia cell apoptosis. In vivo, downregulation of Nrf2 could increase the sensitivity of leukemia cell to cytarabine and decrease OGG1 expression. Mechanistically, Nrf2-OGG1 axis-mediated AML resistance might be achieved by activating the AKT signaling pathway to regulate downstream apoptotic proteins. Thus, this study reveals a novel mechanism of Nrf2-promoting drug resistance in leukemia, which may provide a potential therapeutic target for the treatment of drug-resistant/refractory leukemia.
Collapse
|