1
|
Wang Y, Yang X, Liu Y, Li Y. A review of common immunotherapy and nano immunotherapy for acute myeloid leukemia. Front Immunol 2025; 16:1505247. [PMID: 40129984 PMCID: PMC11931025 DOI: 10.3389/fimmu.2025.1505247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy. Traditional chemotherapy methods not only bring serious side effects, but also lead to high recurrence rate and drug resistance in some patients. However, as an emerging therapeutic strategy, immunotherapy has shown great potential in the field of AML treatment in recent years. At present, common immunotherapy methods for AML include monoclonal antibodies, CAR-T cell therapy, and immune checkpoint inhibitors. With the deepening of research and technological progress, especially the application of nanotechnology in medicine, new immunotherapy is expected to become one of the important means for the treatment of acute myeloid leukemia in the future.
Collapse
Affiliation(s)
- Yaoyao Wang
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| | - Xiancong Yang
- Laboratory Department, Qilu Hospital of ShanDong University Dezhou Hospital, Dezhou, Shandong, China
| | - Yalin Liu
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
2
|
Tasis A, Spyropoulos T, Mitroulis I. The Emerging Role of CD8 + T Cells in Shaping Treatment Outcomes of Patients with MDS and AML. Cancers (Basel) 2025; 17:749. [PMID: 40075597 PMCID: PMC11898900 DOI: 10.3390/cancers17050749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). In this review, we focus on the implication of CD8+ T cells in the treatment response of patients with MDS and AML. First, we review reported studies of aberrant functionality and clonality of CD8+ T cells in MDS and AML, often driven by the immunosuppressive bone marrow microenvironment, which can hinder effective antitumor immunity. Additionally, we discuss the potential use of CD8+ T cell subpopulations, including memory and senescent-like subsets, as predictive biomarkers for treatment response to a variety of treatment regimens, such as hypomethylating agents, which is the standard of care for patients with higher-risk MDS, and chemotherapy which is the main treatment of patients with AML. Understanding the multifaceted role of CD8+ T cells and their interaction with malignant cells in MDS and AML will provide useful insights into their potential as prognostic/predictive biomarkers, but also uncover alternative approaches to novel treatment strategies that could reshape the therapeutic landscape, thus improving treatment efficacy, aiding in overcoming treatment resistance and improving patient survival in these challenging myeloid neoplasms.
Collapse
Affiliation(s)
- Athanasios Tasis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Theodoros Spyropoulos
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Ioannis Mitroulis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
3
|
Zhang M, Yang Y, Liu J, Guo L, Guo Q, Liu W. Bone marrow immune cells and drug resistance in acute myeloid leukemia. Exp Biol Med (Maywood) 2025; 250:10235. [PMID: 40008144 PMCID: PMC11851207 DOI: 10.3389/ebm.2025.10235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
In recent years, the relationship between the immunosuppressive niche of the bone marrow and therapy resistance in acute myeloid leukemia (AML) has become a research focus. The abnormal number and function of immunosuppressive cells, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), along with the dysfunction and exhaustion of immunological effector cells, including cytotoxic T lymphocytes (CTLs), dendritic cells (DCs) and natural killer cells (NKs), can induce immune escape of leukemia cells and are closely linked to therapy resistance in leukemia. This article reviews the research progress on the relationship between immune cells in the marrow microenvironment and chemoresistance in AML, aiming to provide new ideas for the immunotherapy of AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Drug Resistance, Neoplasm/immunology
- Tumor Microenvironment/immunology
- Dendritic Cells/immunology
- Myeloid-Derived Suppressor Cells/immunology
- Bone Marrow/immunology
- T-Lymphocytes, Regulatory/immunology
- Killer Cells, Natural/immunology
- Bone Marrow Cells/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Miao Zhang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - You Yang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qulian Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wenjun Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Rady M, Mostafa M, Dida G, Sabet F, Abou-Aisha K, Watzl C. Adoptive NK cell therapy in AML: progress and challenges. Clin Exp Med 2025; 25:41. [PMID: 39820676 PMCID: PMC11748472 DOI: 10.1007/s10238-025-01559-5] [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: 11/12/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Adoptive cell therapy (ACT) using natural killer (NK) cells has emerged as a promising therapeutic strategy for acute myeloid leukemia (AML), addressing challenges such as chemotherapy resistance and high relapse rates. Over the years, clinical trials and studies have explored various sources of NK cells, including ex vivo expanded NK cell lines, CAR-NK cells, peripheral blood-derived NK cells, and umbilical cord blood-derived NK cells. These therapies have demonstrated varying degrees of therapeutic efficacy, ranging from transient anti-leukemia activity to sustained remission in select patient groups. Toxicity profiles have generally shown favorable safety outcomes, with minimal incidence of severe adverse effects such as cytokine release syndrome (CRS) or graft-versus-host disease (GVHD). However, persistent challenges remain, including limited NK cell persistence, relapse, and heterogeneity in patient responses. This review provides a comprehensive analysis of clinical outcomes and toxicity profiles provided from clinical trials, clinical studies and case reports conducted in the last 15 years to judge on the efficacy, safety and applicability of using NK cells for ACT of AML. Our review highlights the significant potential of NK cell-based therapies for AML, while addressing the technical and biological challenges that must be overcome to enhance their efficacy and safety.
Collapse
Affiliation(s)
- Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt.
- Faculty of Biotechnology, German International University, New Administrative Capital, Egypt.
| | - Maha Mostafa
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| | - Gabriel Dida
- University of South Wales, Pontypridd, Wales, UK
- Department of Health Systems Management and Public Health, Technical University of Kenya, Nairobi, Kenya
| | - Fatima Sabet
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| | - Khaled Abou-Aisha
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| | - Carsten Watzl
- Immunology Department, Leibniz Research Center for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| |
Collapse
|
5
|
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
|
6
|
Luo Y, Sheikh TMM, Li X, Yuan Y, Yao F, Wang M, Guo X, Wu J, Shafiq M, Xie Q, Jiao X. Exploring the dynamics of gut microbiota, antibiotic resistance, and chemotherapy impact in acute leukemia patients: A comprehensive metagenomic analysis. Virulence 2024; 15:2428843. [PMID: 39620486 PMCID: PMC11622590 DOI: 10.1080/21505594.2024.2428843] [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: 05/07/2024] [Revised: 08/24/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024] Open
Abstract
Leukemia poses significant challenges to its treatment, and understanding its complex pathogenesis is crucial. This study used metagenomic sequencing to investigate the interplay between chemotherapy, gut microbiota, and antibiotic resistance in patients with acute leukemia (AL). Pre- and post-chemotherapy stool samples from patients revealed alterations in microbial richness, taxa, and antibiotic resistance genes (ARGs). The analysis revealed a decreased alpha diversity, increased dispersion in post-chemotherapy samples, and changes in the abundance of specific bacteria. Key bacteria such as Enterococcus, Klebsiella, and Escherichia coli have been identified as prevalent ARG carriers. Correlation analysis between gut microbiota and blood indicators revealed potential links between microbial species and inflammatory biomarkers, including C-reactive protein (CRP) and adenosine deaminase (ADA). This study investigated the impact of antibiotic dosage on microbiota and ARGs, revealing networks connecting co-occurring ARGs with microbial species (179 nodes, 206 edges), and networks associated with ARGs and antibiotic dosages (50 nodes, 50 edges). Antibiotics such as cephamycin and sulfonamide led to multidrug-resistant Klebsiella colonization. Our analyses revealed distinct microbial profiles with Salmonella enterica elevated post-chemotherapy in NF patients and Akkermansia muciniphila elevated pre-chemotherapy. These microbial signatures could inform strategies to modulate the gut microbiome, potentially mitigating the risk of neutropenic fever in patients undergoing chemotherapy. Finally, a comprehensive analysis of KEGG modules shed light on disrupted metabolic pathways after chemotherapy, providing insights into potential targets for managing side effects. Overall, this study revealed intricate relationships between gut microbiota, chemotherapy, and antibiotic resistance, providing new insights into improving therapy and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ying Luo
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | - Xin Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - YuMeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Fen Yao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Meimei Wang
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoling Guo
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Jilong Wu
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Shantou University Medical College, Shantou, China
| | - Qingdong Xie
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| |
Collapse
|
7
|
Zha C, Yang X, Yang J, Zhang Y, Huang R. Immunosuppressive microenvironment in acute myeloid leukemia: overview, therapeutic targets and corresponding strategies. Ann Hematol 2024; 103:4883-4899. [PMID: 39607487 DOI: 10.1007/s00277-024-06117-9] [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: 09/06/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Similar to other malignancies, immune dysregulation is a key feature of acute myeloid leukemia (AML), manifesting as suppressed anti-leukemia immune cells, immune evasion by leukemia blasts, and disease progression. Various immunosuppressive factors within the AML microenvironment contribute to the weakening of host immune responses and the efficacy of cellular immunotherapy. To address these challenges, strategies targeting immunosuppressive elements within the AML microenvironment aim to bolster host or adoptive immune effector cells, ultimately enhancing leukemia treatment. Additionally, the off-target effects of certain targeted drugs (venetoclax, sorafenib, ivosidenib, etc.) may also positively impact anti-AML immunity and immunotherapy. This review provides an overview of the immunosuppressive factors present in AML microenvironment and the strategies developed to rescue immune cells from immunosuppression. We also outline how targeted agents can alter the immune landscape in AML patients, and discuss the potential of targeted drugs to benefit host anti-leukemia immunity and immunotherapy for AML.
Collapse
Affiliation(s)
- Chenyu Zha
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinyu Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yujie Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Rui Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
8
|
Wang Z, Liu T, Li Y, Li Z, Bi K. Increased Th17 and Treg levels in peripheral blood positively correlate with minimal residual disease in acute myeloid leukaemia. Hematology 2024; 29:2346971. [PMID: 38682816 DOI: 10.1080/16078454.2024.2346971] [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: 04/24/2023] [Accepted: 04/18/2024] [Indexed: 05/01/2024] Open
Abstract
PURPOSE Immune dysregulation plays a key role in acute myeloid leukemia (AML). We aimed to explore the correlation between T helper cell 17 (Th17) and the regulatory cells (Tregs) in the peripheral blood of patients with newly diagnosed (ND) AML and bone marrow blast cells, as well as minimal residual disease (MRD) before and after treatment. METHODS Changes in Th17 and Treg cells in the peripheral blood of 32 patients with ND AML were observed before and after induction chemotherapy with cytarabine for seven days and anthracycline for three days. The levels of inflammatory cytokines were measured using an enzyme-linked immunosorbent assay. Correlation analysis between bone marrow blast cells and Th17 and Treg cell frequencies was performed using the Pearson's correlation test. Frequencies of Th17 and Treg cells and MRD were assessed using flow cytometry. RESULTS IL-6, IL-10, IL-17A, and GM-CSF levels gradually increased in patients with ND AML and CR and NR patients. The percentages of Th17 and Treg cells positively correlated with those of blast cells. In addition, the frequencies of Th17 and Treg cells in MRD-positive patients were higher than those in MRD-negative patients at the initial induction and after three months of chemotherapy. The frequencies of Tregs and Th17 cells positively correlated with MRD onset. CONCLUSION Increased Th17 and Treg cell levels were positively correlated with onset of AML, poor remission, and MRD.
Collapse
Affiliation(s)
- Zhimin Wang
- Department of Hematology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, People's Republic of China
- Department of Hematology, Binzhou People's Hospital, Binzhou City, People's Republic of China
| | - Tangxia Liu
- Department of Hematology, Binzhou People's Hospital, Binzhou City, People's Republic of China
| | - Yanru Li
- Department of Hematology, Binzhou People's Hospital, Binzhou City, People's Republic of China
| | - Zunchang Li
- Department of Hematology, Binzhou People's Hospital, Binzhou City, People's Republic of China
| | - Kehong Bi
- Department of Hematology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, People's Republic of China
| |
Collapse
|
9
|
Yu F, Jiang H, Gu Y. Causal relationship between immune cells and acute myeloid leukemia: a two-sample Mendelian randomization study. Discov Oncol 2024; 15:675. [PMID: 39560825 DOI: 10.1007/s12672-024-01565-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Immune cells are crucial in the etiology of acute myeloid leukemia (AML). Given the genetic, epigenetic, and clonal complexities of AML, pinpointing factors linked to immunotherapy presents a formidable challenge. Moreover, investigations into the connection between immune cells and AML are still in their infancy, necessitating further studies to decode the intricate connections involved. MATERIALS AND METHODS Based on Mendelian independent distribution law, Mendelian randomisation (MR) is an analytical method mainly used in epidemiological aetiology inference. This bidirectional two-sample MR study aims to investigate the causal link between immune cell phenotypes and AML. Pooled phenotypic data from 3,757 individuals in a Sardinian cohort, encompassing 731 immune cell phenotypes, were utilized. Aggregate data on AML were sourced from the FinnGen project of the Finnish Biobank. We analyzed the sensitivity of the results and evaluated heterogeneity, employing Cochran's Q test in conjunction with MR-Egger and MR-Presso to assess pleiotropy levels. RESULTS 26 distinct immune cell types were identified that potentially linked causally with AML. Furthermore, our analysis indicated a bidirectional causal link between Resting Treg % CD4 Treg, BAFF-R on memory B cells and AML. CONCLUSION This investigation delineates the causal link between immune cell phenotypes and the pathogenesis of AML, thereby unveiling potential therapeutic avenues to modulate immune cell functions in AML patients. It aims to discover innovative therapeutic strategies that target immune evasion tactics to reinstate immune responses against leukemia.
Collapse
Affiliation(s)
- Fanhua Yu
- Shaoxing Keqiao District Hospital of Traditional Chinese Medicine, Shaoxing, China
| | - Hao Jiang
- Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yena Gu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
| |
Collapse
|
10
|
Colonne CK, Kimble EL, Turtle CJ. Evolving strategies to overcome barriers in CAR-T cell therapy for acute myeloid leukemia. Expert Rev Hematol 2024; 17:797-818. [PMID: 39439295 DOI: 10.1080/17474086.2024.2420614] [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: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a complex and heterogeneous disease characterized by an aggressive clinical course and limited efficacious treatment options in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy is an investigational treatment strategy for R/R AML that has shown some promise. However, obstacles to successful CAR-T cell immunotherapy for AML remain. AREAS COVERED In analyses of clinical trials of CAR-T cell therapy for R/R AML, complete responses without measurable residual disease have been reported, but the durability of those responses remains unclear. Significant barriers to successful CAR-T cell therapy in AML include the scarcity of suitable tumor-target antigens (TTA), inherent T cell functional deficits, and the immunoinhibitory and hostile tumor microenvironment (TME). This review will focus on these barriers to successful CAR-T cell therapy in AML, and discuss scientific advancements and evolving strategies to overcome them. EXPERT OPINION Achieving durable remissions in R/R AML will likely require a multifaceted approach that integrates advancements in TTA selection, enhancement of the intrinsic quality of CAR-T cells, and development of strategies to overcome inhibitory mechanisms in the AML TME.
Collapse
Affiliation(s)
- Chanukya K Colonne
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erik L Kimble
- Translational Science and Therapeutic Division, Fred Hutchinson Cancer Center, Seattle, USA
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Cameron J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
11
|
Li Y, Jia Z, Liu X, Zhao H, Cui G, Luo J, Kong X. Single-cell sequencing technology to characterize stem T-cell subpopulations in acute T-lymphoblastic leukemia and the role of stem T-cells in the disease process. Aging (Albany NY) 2024; 16:13117-13131. [PMID: 39422621 PMCID: PMC11552640 DOI: 10.18632/aging.206123] [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: 03/01/2024] [Accepted: 07/17/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Precursor T-cell acute lymphoblastic leukemia (Pre-T ALL) is a malignant neoplastic disease in which T-cells proliferate in the bone marrow. Single-cell sequencing technology could identify characteristic cell types, facilitating the study of the therapeutic mechanisms in Pre-T ALL. METHODS The single-cell sequencing data (scRNA-seq) of Pre-T ALL were obtained from public databases. Key immune cell subpopulations involved in the progression of Pre-T ALL were identified by clustering and annotating the cellular data using AUCell. Next, pseudo-temporal analysis was performed to identify the differentiation trajectories of immune cell subpopulations using Monocle. Copy number mutation landscape of cell subpopulations was characterized by inferCNV. Finally, cellphoneDB was used to analyze intercellular communication relationships. RESULTS A total of 10 cellular subpopulations were classified, with Pre-T ALL showing a higher proportion of NK/T cells. NK/T cells were further clustered into two subpopulations. Stem T cells showed a high expression of marker genes related to hematopoietic stem cells, Naive T cells had a high expression of CCR7, CCR7, RCAN3, and NK cells high-expressed KLRD1, TRDC. The cell proliferation was reduced and the activation of T cell was increased during the differentiation of stem T cells to Naive T cells. We observed interaction between stem T cells with dendritic cells such as CD74-COPA, CD74-MIF as well as co-inhibition-related interactions such as LGALS9-HAVCR2, TGFB1-TGFBR3. CONCLUSION Stem T cells were involved in the development of Pre-T-ALL through the regulatory effects of transcription factors (TFs) KLF2 and FOS and multiple ligand-receptor pairs.
Collapse
Affiliation(s)
- Yan Li
- Department of Hematology, Handan First Hospital, Handan, Hebei 056001, China
| | - Zhenwei Jia
- Department of Hematology, Handan First Hospital, Handan, Hebei 056001, China
| | - Xiaoyan Liu
- Department of Hematology, Handan First Hospital, Handan, Hebei 056001, China
| | - Hongbo Zhao
- Department of Hematology, Handan First Hospital, Handan, Hebei 056001, China
| | - Guirong Cui
- Department of Hematology, Handan First Hospital, Handan, Hebei 056001, China
| | - Jianmin Luo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Xiaoyang Kong
- Department of Hematology, Handan First Hospital, Handan, Hebei 056001, China
| |
Collapse
|
12
|
Radpour R, Simillion C, Wang B, Abbas HA, Riether C, Ochsenbein AF. IL-9 secreted by leukemia stem cells induces Th1-skewed CD4+ T cells, which promote their expansion. Blood 2024; 144:888-903. [PMID: 38941612 DOI: 10.1182/blood.2024024000] [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: 02/12/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
ABSTRACT In acute myeloid leukemia (AML), leukemia stem cells (LSCs) and leukemia progenitor cells (LPCs) interact with various cell types in the bone marrow (BM) microenvironment, regulating their expansion and differentiation. To study the interaction of CD4+ and CD8+ T cells in the BM with LSCs and LPCs, we analyzed their transcriptome and predicted cell-cell interactions by unbiased high-throughput correlation network analysis. We found that CD4+ T cells in the BM of patients with AML were activated and skewed toward T-helper (Th)1 polarization, whereas interleukin-9 (IL-9)-producing (Th9) CD4+ T cells were absent. In contrast to normal hematopoietic stem cells, LSCs produced IL-9, and the correlation modeling predicted IL9 in LSCs as a main hub gene that activates CD4+ T cells in AML. Functional validation revealed that IL-9 receptor signaling in CD4+ T cells leads to activation of the JAK-STAT pathway that induces the upregulation of KMT2A and KMT2C genes, resulting in methylation on histone H3 at lysine 4 to promote genome accessibility and transcriptional activation. This induced Th1-skewing, proliferation, and effector cytokine secretion, including interferon gamma (IFN-γ) and tumor necrosis factor α (TNF-α). IFN-γ and, to a lesser extent, TNF-α produced by activated CD4+ T cells induced the expansion of LSCs. In accordance with our findings, high IL9 expression in LSCs and high IL9R, TNF, and IFNG expression in BM-infiltrating CD4+ T cells correlated with worse overall survival in AML. Thus, IL-9 secreted by AML LSCs shapes a Th1-skewed immune environment that promotes their expansion by secreting IFN-γ and TNF-α.
Collapse
MESH Headings
- Interleukin-9/genetics
- Interleukin-9/metabolism
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/immunology
- Th1 Cells/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Tumor Microenvironment/immunology
- Receptors, Interleukin-9/genetics
- Receptors, Interleukin-9/metabolism
- Interferon-gamma/metabolism
- Histone-Lysine N-Methyltransferase/genetics
Collapse
Affiliation(s)
- Ramin Radpour
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Bofei Wang
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Hussein A Abbas
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX
| | - Carsten Riether
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F Ochsenbein
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Khattab S, El Sorady M, El-Ghandour A, Visani G, Piccaluga PP. Hematopoietic and leukemic stem cells homeostasis: the role of bone marrow niche. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1027-1055. [PMID: 39351440 PMCID: PMC11438561 DOI: 10.37349/etat.2024.00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024] Open
Abstract
The bone marrow microenvironment (BMM) has highly specialized anatomical characteristics that provide a sanctuary place for hematopoietic stem cells (HSCs) that allow appropriate proliferation, maintenance, and self-renewal capacity. Several cell types contribute to the constitution and function of the bone marrow niche. Interestingly, uncovering the secrets of BMM and its interaction with HSCs in health paved the road for research aiming at better understanding the concept of leukemic stem cells (LSCs) and their altered niche. In fact, they share many signals that are responsible for interactions between LSCs and the bone marrow niche, due to several biological similarities between LSCs and HSCs. On the other hand, LSCs differ from HSCs in their abnormal activation of important signaling pathways that regulate survival, proliferation, drug resistance, invasion, and spread. Targeting these altered niches can help in better treatment choices for hematological malignancies and bone marrow disorders in general and acute myeloid leukemia (AML) in particular. Moreover, targeting those niches may help in decreasing the emergence of drug resistance and lower the relapse rate. In this article, the authors reviewed the most recent literature on bone marrow niches and their relations with either normal HSCs and AML cells/LSC, by focusing on pathogenetic and therapeutic implications.
Collapse
Affiliation(s)
- Shaimaa Khattab
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
- Medical Research Institute, Hematology department, Alexandria University, Alexandria 21561, Egypt
| | - Manal El Sorady
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Ashraf El-Ghandour
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, Azienda Ospedaliera Marche Nord, 61121 Pesaro, Italy
| | - Pier Paolo Piccaluga
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
| |
Collapse
|
14
|
Perzolli A, Koedijk JB, Zwaan CM, Heidenreich O. Targeting the innate immune system in pediatric and adult AML. Leukemia 2024; 38:1191-1201. [PMID: 38459166 PMCID: PMC11147779 DOI: 10.1038/s41375-024-02217-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
While the introduction of T cell-based immunotherapies has improved outcomes in many cancer types, the development of immunotherapies for both adult and pediatric AML has been relatively slow and limited. In addition to the need to identify suitable target antigens, a better understanding of the immunosuppressive tumor microenvironment is necessary for the design of novel immunotherapy approaches. To date, most immune characterization studies in AML have focused on T cells, while innate immune lineages such as monocytes, granulocytes and natural killer (NK) cells, received less attention. In solid cancers, studies have shown that innate immune cells, such as macrophages, myeloid-derived suppressor cells and neutrophils are highly plastic and may differentiate into immunosuppressive cells depending on signals received in their microenvironment, while NK cells appear to be functionally impaired. Hence, an in-depth characterization of the innate immune compartment in the TME is urgently needed to guide the development of immunotherapeutic interventions for AML. In this review, we summarize the current knowledge on the innate immune compartment in AML, and we discuss how targeting its components may enhance T cell-based- and other immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alicia Perzolli
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Joost B Koedijk
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Olaf Heidenreich
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands.
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
15
|
Corradi G, Forte D, Cristiano G, Polimeno A, Ciciarello M, Salvestrini V, Bandini L, Robustelli V, Ottaviani E, Cavo M, Ocadlikova D, Curti A. Ex vivo characterization of acute myeloid leukemia patients undergoing hypomethylating agents and venetoclax regimen reveals a venetoclax-specific effect on non-suppressive regulatory T cells and bona fide PD-1 +TIM3 + exhausted CD8 + T cells. Front Immunol 2024; 15:1386517. [PMID: 38812504 PMCID: PMC11133521 DOI: 10.3389/fimmu.2024.1386517] [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: 02/15/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive heterogeneous disease characterized by several alterations of the immune system prompting disease progression and treatment response. The therapies available for AML can affect lymphocyte function, limiting the efficacy of immunotherapy while hindering leukemia-specific immune reactions. Recently, the treatment based on Venetoclax (VEN), a specific B-cell lymphoma 2 (BCL-2) inhibitor, in combination with hypomethylating agents (HMAs) or low-dose cytarabine, has emerged as a promising clinical strategy in AML. To better understand the immunological effect of VEN treatment, we characterized the phenotype and immune checkpoint (IC) receptors' expression on CD4+ and CD8+ T cells from AML patients after the first and second cycle of HMA in combination with VEN. HMA and VEN treatment significantly increased the percentage of naïve CD8+ T cells and TIM-3+ CD4+ and CD8+ T cells and reduced cytokine-secreting non-suppressive T regulatory cells (Tregs). Of note, a comparison between AML patients treated with HMA only and HMA in combination with VEN revealed the specific contribution of VEN in modulating the immune cell repertoire. Indeed, the reduction of cytokine-secreting non-suppressive Tregs, the increased TIM-3 expression on CD8+ T cells, and the reduced co-expression of PD-1 and TIM-3 on both CD4+ and CD8+ T cells are all VEN-specific. Collectively, our study shed light on immune modulation induced by VEN treatment, providing the rationale for a novel therapeutic combination of VEN and IC inhibitors in AML patients.
Collapse
Affiliation(s)
- Giulia Corradi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Dorian Forte
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Gianluca Cristiano
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Andrea Polimeno
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Marilena Ciciarello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Consiglio Nazionale delle Ricerche (CNR) Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, Bologna, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Valentina Salvestrini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Lorenza Bandini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Valentina Robustelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Emanuela Ottaviani
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Michele Cavo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Darina Ocadlikova
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Antonio Curti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| |
Collapse
|
16
|
Zhong F, He S, Guo N, Shi L, Zhang L, Jin H, Kong G. A novel immunogenic cell death-related classification indicates the immune landscape and predicts clinical outcome and treatment response in acute myeloid leukemia. Cancer Cell Int 2024; 24:139. [PMID: 38627685 PMCID: PMC11022379 DOI: 10.1186/s12935-024-03326-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/12/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) is closely related to anti-tumor therapy and regulates the tumor microenvironment (TME). This study aims to explore the molecular characteristics of ICD in acute myeloid leukemia (AML) and to analyze the value of ICD-related biomarkers in TME indication, prognosis prediction, and treatment response evaluation in AML. METHODS Single-sample gene set enrichment analysis was used to calculate the ICD score. LASSO regression was used to construct a prognostic risk score model. We also analyzed differences in clinical characteristics, immune landscape, immunotherapy response, and chemotherapy sensitivity between high-risk and low-risk patients. RESULTS This study identified two ICD-related subtypes and found significant heterogeneity in clinical prognosis, TME, and immune landscape between different ICD subtypes. Subsequently, a novel ICD-related prognostic risk score model was developed, which accurately predicted the prognosis of AML patients and was validated in nine AML cohorts. Moreover, there were significant correlations between risk scores and clinicopathological factors, somatic mutations, TME characteristics, immune cell infiltration, immunotherapy response, and chemosensitivity. We further validated the model gene expression in a clinically real-world cohort. CONCLUSIONS The novel ICD-related signatures identified and validated by us can serve as promising biomarkers for predicting clinical outcomes, chemotherapy sensitivity, and immunotherapy response in AML patients, guiding the establishment of personalized and accurate treatment strategies for AML.
Collapse
Affiliation(s)
- Fangmin Zhong
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shuyang He
- Queen Mary School of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Ni Guo
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Luyi Shi
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Linlin Zhang
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Hua Jin
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Guangyao Kong
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.
- Key Laboratory of Surgical Critical Care and Life Support, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
17
|
Slezak AJ, Chang K, Beckman TN, Refvik KC, Alpar AT, Lauterbach AL, Solanki A, Kwon JW, Gomes S, Mansurov A, Hubbell JA. Cysteine-binding adjuvant enhances survival and promotes immune function in a murine model of acute myeloid leukemia. Blood Adv 2024; 8:1747-1759. [PMID: 38324726 PMCID: PMC10985806 DOI: 10.1182/bloodadvances.2023012529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
ABSTRACT Therapeutic vaccination has long been a promising avenue for cancer immunotherapy but is often limited by tumor heterogeneity. The genetic and molecular diversity between patients often results in variation in the antigens present on cancer cell surfaces. As a result, recent research has focused on personalized cancer vaccines. Although promising, this strategy suffers from time-consuming production, high cost, inaccessibility, and targeting of a limited number of tumor antigens. Instead, we explore an antigen-agnostic polymeric in situ cancer vaccination platform for treating blood malignancies, in our model here with acute myeloid leukemia (AML). Rather than immunizing against specific antigens or targeting adjuvant to specific cell-surface markers, this platform leverages a characteristic metabolic and enzymatic dysregulation in cancer cells that produces an excess of free cysteine thiols on their surfaces. These thiols increase in abundance after treatment with cytotoxic agents such as cytarabine, the current standard of care in AML. The resulting free thiols can undergo efficient disulfide exchange with pyridyl disulfide (PDS) moieties on our construct and allow for in situ covalent attachment to cancer cell surfaces and debris. PDS-functionalized monomers are incorporated into a statistical copolymer with pendant mannose groups and TLR7 agonists to target covalently linked antigen and adjuvant to antigen-presenting cells in the liver and spleen after IV administration. There, the compound initiates an anticancer immune response, including T-cell activation and antibody generation, ultimately prolonging survival in cancer-bearing mice.
Collapse
Affiliation(s)
- Anna J. Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Kevin Chang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Taryn N. Beckman
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL
| | - Kirsten C. Refvik
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Aaron T. Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | | | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL
| | - Jung Woo Kwon
- Department of Pathology, University of Chicago, Chicago, IL
| | - Suzana Gomes
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
- Committee on Immunology, University of Chicago, Chicago, IL
- Committee on Cancer Biology, University of Chicago, Chicago, IL
| |
Collapse
|
18
|
Jing L, Zhang B, Sun J, Feng J, Fu D. Prognostic insights and immune microenvironment delineation in acute myeloid leukemia by ferroptosis-derived signature. Heliyon 2024; 10:e28237. [PMID: 38532996 PMCID: PMC10963645 DOI: 10.1016/j.heliyon.2024.e28237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Acute myeloid leukemia (AML) represents as a prevalent and formidable hematological malignancy, characterized by notably low 5-year survival rates. Ferroptosis has been found to be correlated with cancer initiation, therapeutic response, and clinical outcome. Nevertheless, the involvement of Ferroptosis-related genes (FRGs) in AML remains ambiguous. Five independent AML cohorts totaling 1,470 (GSE37642, GSE12417, GSE10358, Beat-AML, and TCGA-AML) patients with clinical information were used to systematically investigated the influence of these FRGs expression on outcome and tumor microenvironment. The integration of these datasets led to the subdivision into training and validation sets. Nineteen FRGs were identified as correlated with the overall survival (OS) of AML patients, primarily enriched in ferroptosis, fatty acid metabolism, and leukemia-related signaling pathways. The prognostic signature, consisting of 11 FRGs, was formulated using LASSO-Cox stepwise regression analysis. Patients with high-risk scores exhibited reduced survival compared to those in the low-risk group. The receiver operating characteristic (ROC) analysis underscored the signature's robust predictive accuracy. The high predictive efficacy was confirmed by both internal and external validation datasets. Leukemia and signaling related to immune regulation were mainly enriched pathways of the differentially expressed genes by comparing high- and low-risk groups. The immune composition deconvolution might indicate an immunosuppressive niche in the high-risk patients. The pRRophetic algorithm exploration unveiled chemical drugs with potentially sensitivity among patients in both groups. Collectively, our study developed a ferroptosis-derived prognostic signature that provides the OS prediction and identifies the immune microenvironment for AML patients on large-scale AML cohorts.
Collapse
Affiliation(s)
- Lijun Jing
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Biyu Zhang
- School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Jinghui Sun
- College of Medicine, Medical University of South Carolina, Charleston, SC, 29425, United States
| | - Jueping Feng
- Department of Oncology, Wuhan Fourth Hospital, Wuhan, 430033, China
| | - Denggang Fu
- College of Medicine, Medical University of South Carolina, Charleston, SC, 29425, United States
| |
Collapse
|
19
|
Zhang Y, Huang Y, Hong Y, Lin Z, Zha J, Zhu Y, Li Z, Wang C, Fang Z, Zhou Z, Peng Y, Yu X, Liu L, Xu B. Lactate acid promotes PD-1 + Tregs accumulation in the bone marrow with high tumor burden of Acute myeloid leukemia. Int Immunopharmacol 2024; 130:111765. [PMID: 38447414 DOI: 10.1016/j.intimp.2024.111765] [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: 01/05/2024] [Revised: 02/18/2024] [Accepted: 02/24/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) displayed poor response to programmed death-1 (PD-1) blockade therapy. Regulatory T cells (Tregs) was one of major immunosuppressive components in Tumor microenvironment and plays a vital role in the resistance of immunotherapy. Coinhibitory receptors regulate function of regulatory Tregs and are associated with resistance of PD-1 blockade. However, the coinhibitory receptors expression and differentiated status of Tregs in AML patients remain to be unclear. METHODS Phenotypic determination of Tregs and CD8+ T cells in bone marrow of healthy donors and AML patients was performed by flow cytometry. Coculture experiments of AML and Tregs in vitro were performed and the concentrations of lactate acid (LA) in the supernatant were examined by ELISA. RESULTS More Tregs differentiated into effector subsets in AML patients. However, PD-1 and T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) expression on Tregs were comparable in healthy donors and AML patients. Further analysis showed that PD-1+ and PD-1+TIGIT+Tregs are more abundant in the bone marrow of patients with higher leukemic load. Moreover, PD-1+ Tregs accumulation was associated with higher level of senescent CD4+ T cells and increased frequencies of exhausted CD4+ as well as CD8+ T cells. Notably, neither Tregs nor their effector subsets were decreased among patients in complete remission. PD-1 expression was significantly downregulated in Tregs after achieving complete remission. Mechanistically, both AML cell line (KG-1α) and primary AML blasts produced high concentration of LA. Blockade of LA by lactate transporter inhibitor abrogated the upregulation of PD-1 by AML cells. CONCLUSION PD-1+ Tregs accumulation in bone marrow in higher leukemic burden setting was linked to lactate acid secreted by AML blasts and decreased after disease remission. Our findings provided a novel insight into Tregs in AML and possible mechanism for resistance of PD-1 blockade in AML.
Collapse
Affiliation(s)
- Yining Zhang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Yueting Huang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Yan Hong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Zhijuan Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Yuwen Zhu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Zhifeng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Caiyan Wang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Zhihong Fang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Ziwei Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China
| | - Yun Peng
- Department of Rheumatology and Clinical Immunology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xingxing Yu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China.
| | - Long Liu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China.
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China; Key laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancies, Xiamen, 361003, China.
| |
Collapse
|
20
|
Zhong F, Yao F, Jiang J, Yu X, Liu J, Huang B, Wang X. CD8 + T cell-based molecular subtypes with heterogeneous immune landscapes and clinical significance in acute myeloid leukemia. Inflamm Res 2024; 73:329-344. [PMID: 38195768 DOI: 10.1007/s00011-023-01839-4] [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: 10/18/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy. Although high-dose chemotherapy is the primary treatment option, it cannot cure the disease, and new approaches need to be developed. The tumor microenvironment (TME) plays a crucial role in tumor biology and immunotherapy. CD8 + T cells are the main anti-tumor immune effector cells, and it is essential to understand their relationship with the TME and the clinicopathological characteristics of AML. METHODS In this study, we conducted a systematic analysis of CD8 + T cell infiltration through multi-omics data and identified molecular subtypes with significant differences in CD8 + T cell infiltration and prognosis. We aimed to provide a comprehensive evaluation of the pathological factors affecting the prognosis of AML patients and to offer theoretical support for the precise treatment of AML. RESULTS Our results indicate that CD8 + T cell infiltration is accompanied by immunosuppression, and that there are two molecular subtypes, with the C2 subtype having a significantly worse prognosis than the C1 subtype, as well as less CD8 + T cell infiltration. We developed a signature to distinguish molecular subtypes using multiple machine learning algorithms and validated the prognostic predictive power of molecular subtypes in nine AML cohorts including 2059 AML patients. Our findings suggest that there are different immunosuppressive characteristics between the two subtypes. The C1 subtype has up-regulated expression of immune checkpoints such as CTLA4, PD-1, LAG3, and TIGITD, while the C2 subtype infiltrates more immunosuppressive cells such as Tregs and M2 macrophages. The C1 subtype is more responsive to anti-PD-1 immunotherapy and induction chemotherapy, as well as having higher immune and cancer-promoting variant-related pathway activity. Patients with the C2 subtype had a higher FLT3 mutation rate, higher WBC counts, and a higher percentage of blasts, as indicated by increased activity of signaling pathways involved in energy metabolism and cell proliferation. Analysis of data from ex vivo AML cell drug assays has identified a group of drugs that differ in therapeutic sensitivity between molecular subtypes. CONCLUSIONS Our results suggest that the molecular subtypes we constructed have potential application value in the prognosis evaluation and treatment guidance of AML patients.
Collapse
Affiliation(s)
- Fangmin Zhong
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fangyi Yao
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Junyao Jiang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiajing Yu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jing Liu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
21
|
You E, Park CJ, Cho YU, Jang S, Lee MY, Kim H, Koh KN, Im HJ, Choi EJ, Lee JH, Lee KH. Increased PD-1 expression of bone marrow T-cells in acute myeloid leukaemia patients after stem cell transplantation, and its association with overall survival. Ann Clin Biochem 2024; 61:79-89. [PMID: 37314798 DOI: 10.1177/00045632231184716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Immune checkpoints are involved in mechanisms by which tumours escape from the host immune system. Our aim was to evaluate acute myeloid leukaemia (AML) patients to determine expression levels of checkpoint molecules according to diagnosis and treatments, and to identify optimal candidates for checkpoint blockade. METHODS Bone marrow (BM) samples were obtained from 279 AML patients at different disease status and from 23 controls. Flow cytometric analyses of PD-1 and PD-L1/PD-L2 expression were performed. RESULTS Programmed death-1 (PD-1) expression levels on CD8+ T-cells at AML diagnosis were increased compared to controls. PD-L1 and PD-L2 expression levels on leukaemic cells at diagnosis were significantly higher in secondary AML than in de novo AML. PD-1 levels on CD8+ and CD4+ T-cells after allo-SCT were significantly higher than those at diagnosis and after CTx. PD-1 expression on CD8+ T-cells increased in the acute GVHD group than in the non-GVHD group. The overall survival of patients with high PD-1 expression on CD8+ T-cells was significantly shorter than that of patients with low PD-1 expression. CONCLUSIONS In conclusion, patients who underwent allo-SCT exhibited high PD-1 expression, suggesting that allo-SCT increases PD-1 expression on T-cells, and the patients with high PD-1 expression on CD8+ T-cells after allo-SCT showed the poor prognosis. For these patients, PD-1 blockade could be an immunotherapeutic strategy.
Collapse
Affiliation(s)
- Eunkyoung You
- Department of Laboratory Medicine, Inje University College of Medicine, Busan Paik Hospital, Busan, Korea
| | - Chan-Jeoung Park
- Department of Laboratory Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Young-Uk Cho
- Department of Laboratory Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Seongsoo Jang
- Department of Laboratory Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Min Young Lee
- Department of Laboratory Medicine, Kyung Hee University School of Medicine and Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Hery Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea
| | - Kyung Nam Koh
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea
| | - Ho Joon Im
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children's Hospital, Seoul, Korea
| | - Eun-Ji Choi
- Department of Hematology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Je-Hwan Lee
- Department of Hematology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyoo-Hyung Lee
- Department of Hematology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
22
|
Lu W, Yu G, Li Y, Yin C, Long J, Chen X, Chen Y, Zheng Z, Lai Y, Zhou X, Xu D. Identifying prognostic biomarker related to immune infiltration in acute myeloid leukemia. Clin Exp Med 2023; 23:4553-4562. [PMID: 37561221 DOI: 10.1007/s10238-023-01164-4] [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: 07/05/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
The immune cells of tumor microenvironment (TME) constitute a vital element of the tumor tissue. There is increasing evidence for their clinical significance in predicting prognosis and therapeutic outcomes. However, the TME immune cell infiltrating pattern of the bone marrow in acute myeloid leukemia (AML) patients remains unclear. Here, RNA-sequencing results of AML patients from TCGA database were used to quantify the abundance of 28 types of immune cells in the TME using the single-sample gene set enrichment analysis algorithm. We comprehensively evaluated the immune infiltration status in the TCGA-LAML cohort and defined two immunophenotypes: the immune hot and immune cold subtypes. Additionally, we constructed a TME score reflecting the immune infiltrating pattern of the patients using Cox regression algorithm. Subtypes with high TME score were characterized by over-activation of immune inflammation-related pathways, release of inflammatory factors, T-cell dysfunction, and poor prognosis. Subtypes with a low TME score were characterized by relatively low immune infiltration and immune exclusion. Our analysis indicated that patients in the low TME score group were more sensitive to chemotherapeutic drugs, and those in high TME score were more likely to respond to immunotherapy. Our study provides a new direction to evaluate anti-tumor therapy from immune infiltration of the TME, and the individualized scoring system in this study has important clinical significance in identifying patients who respond to immunotherapy.
Collapse
Affiliation(s)
- Weixiang Lu
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Guopan Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yanlin Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Changxin Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jiaxin Long
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiaofan Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yanxiao Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Zhongxin Zheng
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yujie Lai
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524000, China
| | - Xinyu Zhou
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524000, China
| | - Dan Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
23
|
Solomon SR, Solh M, Morris LE, Holland HK, Bachier-Rodriguez L, Zhang X, Guzowski C, Jackson KC, Brown S, Bashey A. Phase 2 study of PD-1 blockade following autologous transplantation for patients with AML ineligible for allogeneic transplant. Blood Adv 2023; 7:5215-5224. [PMID: 37379271 PMCID: PMC10500475 DOI: 10.1182/bloodadvances.2023010477] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/25/2023] [Accepted: 06/11/2023] [Indexed: 06/30/2023] Open
Abstract
Allogeneic transplant remains the best postremission therapy for patients with nonfavorable risk acute myeloid leukemia (AML). However, some patients are ineligible because of psychosocial barriers, such as lack of appropriate caregiver support. We hypothesized that immune checkpoint inhibition after autologous transplant might represent effective postremission therapy in such patients. We conducted a phase 2 study of autologous transplantation followed by administration of pembrolizumab (8 cycles starting day +1). Twenty patients with nonfavorable AML in complete remission were treated (median age, 64 years; CR1, 80%); 55% were non-White and adverse-risk AML was present in 40%. Treatment was well tolerated, with only 1 nonrelapse death. Immune-related adverse events occurred in 9 patients. After a median follow-up of 80 months, 14 patients remain alive, with 10 patients in continuous remission. The estimated 2-year LFS was 48.4%, which met the primary end point of 2-year LFS >25%; the 2-year overall survival (OS), nonrelapse mortality, and cumulative incidences of relapse were 68%, 5%, and 46%, respectively. In comparison with a propensity score-matched cohort group of patients with AML receiving allogeneic transplant, the 3-year OS was similar (73% vs 76%). Patients in the study had inferior LFS (51% vs 75%) but superior postrelapse survival (45% vs 14%). In conclusion, programmed cell death protein-1 blockade after autologous transplant is a safe and effective alternative postremission strategy in patients with nonfavorable risk AML who are ineligible for allogeneic transplant, a context in which there is significant unmet need. This trial was registered at www.clinicaltrials.gov as #NCT02771197.
Collapse
Affiliation(s)
- Scott R. Solomon
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Melhem Solh
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Lawrence E. Morris
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - H. Kent Holland
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | | | - Xu Zhang
- Center for Clinical and Transitional Sciences, University of Texas Health Science Center, Houston, TX
| | - Caitlin Guzowski
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Katelin C Jackson
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Stacey Brown
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| | - Asad Bashey
- Blood and Marrow Transplant Program, Northside Hospital Cancer Institute, Atlanta, GA
| |
Collapse
|
24
|
Bakhtiyari M, Liaghat M, Aziziyan F, Shapourian H, Yahyazadeh S, Alipour M, Shahveh S, Maleki-Sheikhabadi F, Halimi H, Forghaniesfidvajani R, Zalpoor H, Nabi-Afjadi M, Pornour M. The role of bone marrow microenvironment (BMM) cells in acute myeloid leukemia (AML) progression: immune checkpoints, metabolic checkpoints, and signaling pathways. Cell Commun Signal 2023; 21:252. [PMID: 37735675 PMCID: PMC10512514 DOI: 10.1186/s12964-023-01282-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a multifarious and heterogeneous array of illnesses characterized by the anomalous proliferation of myeloid cells in the bone marrow microenvironment (BMM). The BMM plays a pivotal role in promoting AML progression, angiogenesis, and metastasis. The immune checkpoints (ICs) and metabolic processes are the key players in this process. In this review, we delineate the metabolic and immune checkpoint characteristics of the AML BMM, with a focus on the roles of BMM cells e.g. tumor-associated macrophages, natural killer cells, dendritic cells, metabolic profiles and related signaling pathways. We also discuss the signaling pathways stimulated in AML cells by BMM factors that lead to AML progression. We then delve into the roles of immune checkpoints in AML angiogenesis, metastasis, and cell proliferation, including co-stimulatory and inhibitory ICs. Lastly, we discuss the potential therapeutic approaches and future directions for AML treatment, emphasizing the potential of targeting metabolic and immune checkpoints in AML BMM as prognostic and therapeutic targets. In conclusion, the modulation of these processes through the use of directed drugs opens up new promising avenues in combating AML. Thereby, a comprehensive elucidation of the significance of these AML BMM cells' metabolic and immune checkpoints and signaling pathways on leukemic cells can be undertaken in the future investigations. Additionally, these checkpoints and cells should be considered plausible multi-targeted therapies for AML in combination with other conventional treatments in AML. Video Abstract.
Collapse
Affiliation(s)
- Maryam Bakhtiyari
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hooriyeh Shapourian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Alipour
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Shaghayegh Shahveh
- American Association of Naturopath Physician (AANP), Washington, DC, USA
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Halimi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Forghaniesfidvajani
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA.
| |
Collapse
|
25
|
Huang W, Zheng S, Wang Q, Zhao N, Long Z. Identification and validation of a prognostic risk-scoring model based on the level of TIM-3 expression in acute myeloid leukemia. Sci Rep 2023; 13:15658. [PMID: 37730831 PMCID: PMC10511414 DOI: 10.1038/s41598-023-42700-2] [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: 06/24/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterized by an unfavorable prognosis due to the presence of self-renewing leukemic stem cells (LSCs). The presence of T-cell immunoglobulin mucin-3 (TIM-3) on the surface of LSCs has been observed in various types of human AML, exerting an impact on the prognostic outcome. Exploring the hub genes associated with varying levels of TIM-3 expression offers a valuable approach to enhance our understanding of the underlying mechanisms involving TIM-3 and to identify potential prognostic indicators in AML. Nevertheless, to date, no research studies have reported a prognostic model that relies on the level of TIM-3 expression. In our study, we screen the hub-genes based on different expression level of TIM-3 through WGCNA. The prognostic risk-scoring model was constructed based on hub-genes. The results show the risk prognostic model has extraordinary ability to predict prognosis in both the training and validation sets. The high-risk group present poor prognosis with mutation of NPM1, TP53 (Multiple Hit) and FLT3(multiple hit), while IDH2 (Missense Mutation), MUC16 (Multiple Hit/Missense Mutation) occur mutation in low-risk group presenting favorite prognosis than high-risk group. Leukocyte cell-cell adhesion, regulation of T cell activation and I-κB kinase/NF-κB signaling enriched in high-risk group, involving in HSCs or LSCs anchoring to BM, which implicated in LSCs survival and chemotherapy resistance. B7-H3 (CD276) and CD276 would be the potential immune targets in high-risk group. The risk score model may help in distinguishing immune and molecular characteristics, predicting patient outcomes.
Collapse
Affiliation(s)
- Wanxue Huang
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
| | - Shasha Zheng
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Wang
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
| | - Na Zhao
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China.
| | - Zhiguo Long
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China.
| |
Collapse
|
26
|
Chen EC, Garcia JS. Immunotherapy for Acute Myeloid Leukemia: Current Trends, Challenges, and Strategies. Acta Haematol 2023; 147:198-218. [PMID: 37673048 DOI: 10.1159/000533990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND In the past decade, there have been significant breakthroughs in immunotherapies for B-cell lymphoid malignancies and multiple myeloma, but progress has been much less for acute myeloid leukemia (AML). Nevertheless, challenge begets innovation and several therapeutic strategies are under investigation. SUMMARY In this review, we review the state of the art in AML immunotherapy including CD33- and CD123-targeted agents, immune checkpoint inhibition, and adoptive cell therapy strategies. We also share conceptual frameworks for approaching the growing catalog of investigational AML immunotherapies and propose future directions for the field. KEY MESSAGES Immunotherapies for AML face significant challenges but novel strategies are in development.
Collapse
Affiliation(s)
- Evan C Chen
- Department of Medical Oncology, Division of Leukemia, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jacqueline S Garcia
- Department of Medical Oncology, Division of Leukemia, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Ghetti M, Vannini I, Bochicchio MT, Azzali I, Ledda L, Marconi G, Melloni M, Fabbri F, Rondoni M, Chicchi R, Angeli D, Ghelli Luserna di Rorà A, Giannini B, Zacheo I, Biguzzi R, Lanza F, Martinelli G, Simonetti G. Uncovering the expression of circPVT1 in the extracellular vesicles of acute myeloid leukemia patients. Biomed Pharmacother 2023; 165:115235. [PMID: 37536029 DOI: 10.1016/j.biopha.2023.115235] [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: 05/18/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023] Open
Abstract
Extracellular vesicles (EVs) act as molecular mediators in the tumor microenvironment, by shuttling information contained within malignant cells and functioning as regulators of the immune system. Circular (circ)RNAs are characterized by a closed loop-like structure that makes them more stable in the extracellular milieu and suitable to be packaged inside EVs. circPVT1 (hsa_circ_0001821) showed an oncogenic role in several cancer types and immunosuppressive properties in myeloid and lymphoid cell subsets. In this study, we characterized EVs from acute myeloid leukemia (AML) patients in terms of size, concentrations, surface markers and circPVT1 cargo. We showed that circPVT1 is overexpressed by primary blast cells from newly-diagnosed AML patients compared with hematopoietic stem-progenitor cells and is released as cell-free RNA in the plasma. We isolated EVs from the plasma of AML patients and healthy subjects by size exclusion chromatography and characterized them by nanoparticle tracking analysis. EVs from patients' plasma are larger compared with those from healthy subjects and their surface profile is characterized by higher levels of the leukemic cell markers CD133, CD105, CD49e and other immune-related epitopes, with differences according to AML molecular profile. Moreover, digital PCR analysis revealed that circPVT1 is more abundant inside EVs from the plasma of AML patients compared with healthy subjects. Our findings provide new insights on the features and content of AML EVs and suggest a role of circPVT1 in the crosstalk between AML cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Martina Ghetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ivan Vannini
- Pathology Unit, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Irene Azzali
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Lorenzo Ledda
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Marconi
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Mattia Melloni
- Laboratory of Biomarkers, Biomolecular Targets and Personalized Medicine in Oncology, Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Rondoni
- Hematology Unit & Romagna Transplant Network, Ravenna Hospital, Ravenna, Italy
| | - Roberta Chicchi
- Laboratorio Unico AUSL della Romagna, U.O. Medicina Trasfusionale di Forlì-Cesena e Officina Trasfusionale della Romagna, Pievesestina di Cesena, Italy
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Andrea Ghelli Luserna di Rorà
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy; Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
| | - Barbara Giannini
- Laboratorio Unico AUSL della Romagna, U.O. Genetica Medica, Pievesestina di Cesena, Italy
| | - Irene Zacheo
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Rino Biguzzi
- Laboratorio Unico AUSL della Romagna, U.O. Medicina Trasfusionale di Forlì-Cesena e Officina Trasfusionale della Romagna, Pievesestina di Cesena, Italy
| | - Francesco Lanza
- Hematology Unit & Romagna Transplant Network, Ravenna Hospital, Ravenna, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| |
Collapse
|
28
|
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
|
29
|
Khelfa M, Leclerc M, Kerbrat S, Boudjemai YNS, Benchouaia M, Neyrinck-Leglantier D, Cagnet L, Berradhia L, Tamagne M, Croisille L, Pirenne F, Maury S, Vingert B. Divergent CD4 + T-cell profiles are associated with anti-HLA alloimmunization status in platelet-transfused AML patients. Front Immunol 2023; 14:1165973. [PMID: 37701444 PMCID: PMC10493329 DOI: 10.3389/fimmu.2023.1165973] [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: 02/14/2023] [Accepted: 07/17/2023] [Indexed: 09/14/2023] Open
Abstract
Introduction Acute myeloid leukemia (AML) is one of the commonest hematologic disorders. Due to the high frequency of disease- or treatment-related thrombocytopenia, AML requires treatment with multiple platelet transfusions, which can trigger a humoral response directed against platelets. Some, but not all, AML patients develop an anti-HLA immune response after multiple transfusions. We therefore hypothesized that different immune activation profiles might be associated with anti-HLA alloimmunization status. Methods We tested this hypothesis, by analyzing CD4+ T lymphocyte (TL) subsets and their immune control molecules in flow cytometry and single-cell multi-omics. Results A comparison of immunological status between anti-HLA alloimmunized and non-alloimmunized AML patients identified differences in the phenotype and function of CD4+ TLs. CD4+ TLs from alloimmunized patients displayed features of immune activation, with higher levels of CD40 and OX40 than the cells of healthy donors. However, the most notable differences were observed in non-alloimmunized patients. These patients had lower levels of CD40 and OX40 than alloimmunized patients and higher levels of PD1. Moreover, the Treg compartment of non-alloimmunized patients was larger and more functional than that in alloimmunized patients. These results were supported by a multi-omics analysis of immune response molecules in conventional CD4+ TLs, Tfh circulating cells, and Tregs. Discussion Our results thus reveal divergent CD4+ TL characteristics correlated with anti-HLA alloimmunization status in transfused AML patients. These differences, characterizing CD4+ TLs independently of any specific antigen, should be taken into account when considering the immune responses of patients to infections, vaccinations, or transplantations.
Collapse
Affiliation(s)
- Mehdi Khelfa
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Mathieu Leclerc
- Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor, Service d’Hématologie clinique, Créteil, France
| | - Stéphane Kerbrat
- Univ Paris Est Creteil, INSERM, IMRB, Plateforme de Génomique, Créteil, France
| | | | - Médine Benchouaia
- Univ Paris Est Creteil, INSERM, IMRB, Plateforme de Génomique, Créteil, France
| | - Déborah Neyrinck-Leglantier
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Léonie Cagnet
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Lylia Berradhia
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Marie Tamagne
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | | | - France Pirenne
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Sébastien Maury
- Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor, Service d’Hématologie clinique, Créteil, France
| | - Benoît Vingert
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
30
|
Larson JH, Jin S, Loschi M, Bolivar Wagers S, Thangavelu G, Zaiken MC, McDonald-Hyman C, Saha A, Aguilar EG, Koehn B, Osborn MJ, Panoskaltsis-Mortari A, Macdonald KPA, Hill GR, Murphy WJ, Serody JS, Maillard I, Kean LS, Kim SV, Littman DR, Blazar BR. Enforced gut homing of murine regulatory T cells reduces early graft-versus-host disease severity. Am J Transplant 2023; 23:1102-1115. [PMID: 36878433 PMCID: PMC10475494 DOI: 10.1016/j.ajt.2023.01.030] [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: 12/19/2022] [Accepted: 01/31/2023] [Indexed: 03/07/2023]
Abstract
Damage to the gastrointestinal tract following allogeneic hematopoietic stem cell transplantation is a significant contributor to the severity and perpetuation of graft-versus-host disease. In preclinical models and clinical trials, we showed that infusing high numbers of regulatory T cells reduces graft-versus-host disease incidence. Despite no change in in vitro suppressive function, transfer of ex vivo expanded regulatory T cells transduced to overexpress G protein-coupled receptor 15 or C-C motif chemokine receptor 9, specific homing receptors for colon or small intestine, respectively, lessened graft-versus-host disease severity in mice. Increased regulatory T cell frequency and retention within the gastrointestinal tissues of mice that received gut homing T cells correlated with lower inflammation and gut damage early post-transplant, decreased graft-versus-host disease severity, and prolonged survival compared with those receiving control transduced regulatory T cells. These data provide evidence that enforced targeting of ex vivo expanded regulatory T cells to the gastrointestinal tract diminishes gut injury and is associated with decreased graft-versus-host disease severity.
Collapse
Affiliation(s)
- Jemma H Larson
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sujeong Jin
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Loschi
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sara Bolivar Wagers
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael C Zaiken
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cameron McDonald-Hyman
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Asim Saha
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ethan G Aguilar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brent Koehn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J Osborn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kelli P A Macdonald
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Immunology Department, Brisbane, Queensland, Australia
| | - Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA; Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, California, USA
| | - Jonathan S Serody
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sangwon V Kim
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Dan R Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, USA; Howard Hughes Medical Institute, New York University School of Medicine, New York, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
31
|
Damiani D, Tiribelli M. Checkpoint Inhibitors in Acute Myeloid Leukemia. Biomedicines 2023; 11:1724. [PMID: 37371818 PMCID: PMC10295997 DOI: 10.3390/biomedicines11061724] [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: 04/09/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The prognosis of acute myeloid leukemia (AML) remains unsatisfactory. Among the reasons for the poor response to therapy and high incidence of relapse, there is tumor cell immune escape, as AML blasts can negatively influence various components of the immune system, mostly weakening T-cells. Since leukemic cells can dysregulate immune checkpoints (ICs), receptor-based signal transductors that lead to the negative regulation of T-cells and, eventually, to immune surveillance escape, the inhibition of ICs is a promising therapeutic strategy and has led to the development of so-called immune checkpoint inhibitors (ICIs). ICIs, in combination with conventional chemotherapy, hypomethylating agents or targeted therapies, are being increasingly tested in cases of AML, but the results reported are often conflicting. Here, we review the main issues concerning the immune system in AML, the main pathways leading to immune escape and the results obtained from clinical trials of ICIs, alone or in combination, in newly diagnosed or relapsed/refractory AML.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| |
Collapse
|
32
|
Romine KA, Bottomly D, Yashar W, Long N, Viehdorfer M, McWeeney SK, Tyner JW. Immune cell proportions correlate with clinicogenomic features and ex vivo drug responses in acute myeloid leukemia. Front Oncol 2023; 13:1192829. [PMID: 37361575 PMCID: PMC10285384 DOI: 10.3389/fonc.2023.1192829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction The implementation of small-molecule and immunotherapies in acute myeloid leukemia (AML) has been challenging due to genetic and epigenetic variability amongst patients. There are many potential mechanisms by which immune cells could influence small-molecule or immunotherapy responses, yet, this area remains understudied. Methods Here we performed cell type enrichment analysis from over 560 AML patient bone marrow and peripheral blood samples from the Beat AML dataset to describe the functional immune landscape of AML. Results We identify multiple cell types that significantly correlate with AML clinical and genetic features, and we also observe significant correlations of immune cell proportions with ex vivo small-molecule and immunotherapy responses. Additionally, we generated a signature of terminally exhausted T cells (Tex) and identified AML with high monocytic proportions as strongly correlating with increased proportions of these immunosuppressive T cells. Discussion Our work, which is accessible through a new "Cell Type" module in our visualization platform (Vizome; http://vizome.org/), can be leveraged to investigate potential contributions of different immune cells on many facets of the biology of AML.
Collapse
Affiliation(s)
- Kyle A. Romine
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, United States
| | - William Yashar
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Nicola Long
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Matthew Viehdorfer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Shannon K. McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, United States
| | - Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
33
|
Mu X, Chen C, Dong L, Kang Z, Sun Z, Chen X, Zheng J, Zhang Y. Immunotherapy in leukaemia. Acta Biochim Biophys Sin (Shanghai) 2023; 55:974-987. [PMID: 37272727 PMCID: PMC10326417 DOI: 10.3724/abbs.2023101] [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: 04/07/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023] Open
Abstract
Leukaemia is the common name for a group of malignant diseases of the haematopoietic system with complex classifications and characteristics. Remarkable progress has been made in basic research and preclinical studies for acute leukaemia compared to that of the many other types/subtypes of leukaemia, especially the exploration of the biological basis and application of immunotherapy in acute myeloid leukaemia (AML) and B-cell acute lymphoblastic leukaemia (B-ALL). In this review, we summarize the basic approaches to immunotherapy for leukaemia and focus on the research progress made in immunotherapy development for AML and ALL. Importantly, despite the advances made to date, big challenges still exist in the effectiveness of leukaemia immunotherapy, especially in AML. Therefore, we use AML as an example and summarize the mechanisms of tumour cell immune evasion, describe recently reported data and known therapeutic targets, and discuss the obstacles in finding suitable treatment targets and the results obtained in recent clinical trials for several types of single and combination immunotherapies, such as bispecific antibodies, cell therapies (CAR-T-cell treatment), and checkpoint blockade. Finally, we summarize novel immunotherapy strategies for treating lymphocytic leukaemia and clinical trial results.
Collapse
Affiliation(s)
- Xingmei Mu
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Chumao Chen
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Loujie Dong
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhaowei Kang
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhixian Sun
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Xijie Chen
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Junke Zheng
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yaping Zhang
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| |
Collapse
|
34
|
D’Silva SZ, Singh M, Pinto AS. NK cell defects: implication in acute myeloid leukemia. Front Immunol 2023; 14:1112059. [PMID: 37228595 PMCID: PMC10203541 DOI: 10.3389/fimmu.2023.1112059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is a complex disease with rapid progression and poor/unsatisfactory outcomes. In the past few years, the focus has been on developing newer therapies for AML; however, relapse remains a significant problem. Natural Killer cells have strong anti-tumor potential against AML. This NK-mediated cytotoxicity is often restricted by cellular defects caused by disease-associated mechanisms, which can lead to disease progression. A stark feature of AML is the low/no expression of the cognate HLA ligands for the activating KIR receptors, due to which these tumor cells evade NK-mediated lysis. Recently, different Natural Killer cell therapies have been implicated in treating AML, such as the adoptive NK cell transfer, Chimeric antigen receptor-modified NK (CAR-NK) cell therapy, antibodies, cytokine, and drug treatment. However, the data available is scarce, and the outcomes vary between different transplant settings and different types of leukemia. Moreover, remission achieved by some of these therapies is only for a short time. In this mini-review, we will discuss the role of NK cell defects in AML progression, particularly the expression of different cell surface markers, the available NK cell therapies, and the results from various preclinical and clinical trials.
Collapse
Affiliation(s)
- Selma Z. D’Silva
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Meenakshi Singh
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Andrea S. Pinto
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| |
Collapse
|
35
|
Liu Z, Xu X, Liu K, Zhang J, Ding D, Fu R. Immunogenic Cell Death in Hematological Malignancy Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207475. [PMID: 36815385 PMCID: PMC10161053 DOI: 10.1002/advs.202207475] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/09/2023] [Indexed: 05/06/2023]
Abstract
Although the curative effect of hematological malignancies has been improved in recent years, relapse or drug resistance of hematological malignancies will eventually recur. Furthermore, the microenvironment disorder is an important mechanism in the pathogenesis of hematological malignancies. Immunogenic cell death (ICD) is a unique mechanism of regulated cell death (RCD) that triggers an intact antigen-specific adaptive immune response by firing a set of danger signals or damage-associated molecular patterns (DAMPs), which is an immunotherapeutic modality with the potential for the treatment of hematological malignancies. This review summarizes the existing knowledge about the induction of ICD in hematological malignancies and the current research on combining ICD inducers with other treatment strategies for hematological malignancies.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Xintong Xu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Kaining Liu
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Jingtian Zhang
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Rong Fu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| |
Collapse
|
36
|
Romine KA, MacPherson K, Cho HJ, Kosaka Y, Flynn PA, Byrd KH, Coy JL, Newman MT, Pandita R, Loo CP, Scott J, Adey AC, Lind EF. BET inhibitors rescue anti-PD1 resistance by enhancing TCF7 accessibility in leukemia-derived terminally exhausted CD8 + T cells. Leukemia 2023; 37:580-592. [PMID: 36681742 PMCID: PMC9991923 DOI: 10.1038/s41375-023-01808-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/08/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
Many acute myeloid leukemia (AML) patients exhibit hallmarks of immune exhaustion, such as increased myeloid-derived suppressor cells, suppressive regulatory T cells and dysfunctional T cells. Similarly, we have identified the same immune-related features, including exhausted CD8+ T cells (TEx) in a mouse model of AML. Here we show that inhibitors that target bromodomain and extra-terminal domain (BET) proteins affect tumor-intrinsic factors but also rescue T cell exhaustion and ICB resistance. Ex vivo treatment of cells from AML mice and AML patients with BET inhibitors (BETi) reversed CD8+ T cell exhaustion by restoring proliferative capacity and expansion of the more functional precursor-exhausted T cells. This reversal was enhanced by combined BETi and anti-PD1 treatment. BETi synergized with anti-PD1 in vivo, resulting in the reduction of circulating leukemia cells, enrichment of CD8+ T cells in the bone marrow, and increase in expression of Tcf7, Slamf6, and Cxcr5 in CD8+ T cells. Finally, we profiled the epigenomes of in vivo JQ1-treated AML-derived CD8+ T cells by single-cell ATAC-seq and found that JQ1 increases Tcf7 accessibility specifically in Tex cells, suggesting that BETi likely acts mechanistically by relieving repression of progenitor programs in Tex CD8+ T cells and maintaining a pool of anti-PD1 responsive CD8+ T cells.
Collapse
Affiliation(s)
- Kyle A Romine
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Kevin MacPherson
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Hyun-Jun Cho
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Yoko Kosaka
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Patrick A Flynn
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Kaelan H Byrd
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jesse L Coy
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Matthew T Newman
- School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ravina Pandita
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Christopher P Loo
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jaime Scott
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Andrew C Adey
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
- Center for Early Detection Advanced Research, Oregon Health & Science University, Portland, OR, USA
| | - Evan F Lind
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
37
|
Arellano-Ballestero H, Sabry M, Lowdell MW. A Killer Disarmed: Natural Killer Cell Impairment in Myelodysplastic Syndrome. Cells 2023; 12:633. [PMID: 36831300 PMCID: PMC9954109 DOI: 10.3390/cells12040633] [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: 01/27/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Myelodysplastic syndrome (MDS) treatment remains a big challenge due to the heterogeneous nature of the disease and its ability to progress to acute myeloid leukemia (AML). The only curative option is allogeneic hematopoietic stem cell transplantation (HSCT), but most patients are unfit for this procedure and are left with only palliative treatment options, causing a big unmet need in the context of this disease. Natural killer (NK) cells are attractive candidates for MDS immunotherapy due to their ability to target myeloid leukemic cells without prior sensitization, and in recent years we have seen an arising number of clinical trials in AML and, recently, MDS. NK cells are reported to be highly dysfunctional in MDS patients, which can be overcome by adoptive NK cell immunotherapy or activation of endogenous NK cells. Here, we review the role of NK cells in MDS, the contribution of the tumor microenvironment (TME) to NK cell impairment, and the most recent data from NK cell-based clinical trials in MDS.
Collapse
Affiliation(s)
| | - May Sabry
- Department of Haematology, University College London, London NW3 5PF, UK
- InmuneBio Inc., Boca Raton, FL 33432, USA
- Novamune Ltd., London WC2R 1DJ, UK
| | - Mark W. Lowdell
- Department of Haematology, University College London, London NW3 5PF, UK
- InmuneBio Inc., Boca Raton, FL 33432, USA
| |
Collapse
|
38
|
N6-methyladenosine in hematological malignancies: a concise review. Curr Opin Hematol 2023; 30:4-13. [PMID: 36165537 DOI: 10.1097/moh.0000000000000741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE OF REVIEW Hematological malignancies are a kind of systemic cancers mostly related to abnormal differentiation of blood stem cells. Because of the poor prognosis, chemotherapy resistance and common recurrence, new mechanisms and treatment therapies are looking forward to be discovered. RECENT FINDINGS Over the years, epigenetic abnormalities have been known to act a key part in occurrence and development of hematological tumors. In the internal modifications on long noncoding eukaryotic mRNA, there is a common type called N6-methyladenosine that can change the expression of target genes and participate in the translation, degradation and splicing of mRNA. M6A is related to a wealth of cancers, such as HNRNPA2B1's elevation in multiple myeloma, METTLE3's elevation in acute myeloid leukemia and lung cancer. Immune cells, playing a significant role in hematological cancers, can also be regulated by m6A. SUMMARY In the review, we summarized the recent progress on hematological malignancies associating with m6A and immune cells, which may offer a new road for the treatment of them.
Collapse
|
39
|
Pousse L, Korfi K, Medeiros BC, Berrera M, Kumpesa N, Eckmann J, Hutter IK, Griesser V, Karanikas V, Klein C, Amann M. CD25 targeting with the afucosylated human IgG1 antibody RG6292 eliminates regulatory T cells and CD25+ blasts in acute myeloid leukemia. Front Oncol 2023; 13:1150149. [PMID: 37205201 PMCID: PMC10185852 DOI: 10.3389/fonc.2023.1150149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Background Acute Myeloid leukemia is a heterogeneous disease that requires novel targeted treatment options tailored to the patients' specific microenvironment and blast phenotype. Methods We characterized bone marrow and/or blood samples of 37 AML patients and healthy donors by high dimensional flow cytometry and RNA sequencing using computational analysis. In addition, we performed ex vivo ADCC assays using allogeneic NK cells isolated from healthy donors and AML patient material to test the cytotoxic potential of CD25 Mab (also referred to as RG6292 and RO7296682) or isotype control antibody on regulatory T cells and CD25+ AML cells. Results Bone marrow composition, in particular the abundance of regulatory T cells and CD25 expressing AML cells, correlated strongly with that of the blood in patients with time-matched samples. In addition, we observed a strong enrichment in the prevalence of CD25 expressing AML cells in patients bearing a FLT3-ITD mutation or treated with a hypomethylating agent in combination with venetoclax. We adopted a patient-centric approach to study AML clusters with CD25 expression and found it most highly expressed on immature phenotypes. Ex vivo treatment of primary AML patient samples with CD25 Mab, a human CD25 specific glycoengineered IgG1 antibody led to the specific killing of two different cell types, CD25+ AML cells and regulatory T cells, by allogeneic Natural Killer cells. Conclusion The in-depth characterization of patient samples by proteomic and genomic analyses supported the identification of a patient population that may benefit most by harnessing CD25 Mab's dual mode of action. In this pre-selected patient population, CD25 Mab could lead to the specific depletion of regulatory T cells, in addition to leukemic stem cells and progenitor-like AML cells that are responsible for disease progression or relapse.
Collapse
Affiliation(s)
- Laurène Pousse
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
- *Correspondence: Laurène Pousse, ; Maria Amann,
| | - Koorosh Korfi
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Bruno C. Medeiros
- Genentech, Inc. Hematology Department, South San Francisco, CA, United States
| | - Marco Berrera
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Nadine Kumpesa
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Jan Eckmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Münich (RICM), Penzberg, Germany
| | - Idil Karakoc Hutter
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Vera Griesser
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Vaios Karanikas
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Christian Klein
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
| | - Maria Amann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Zurich (RICZ), Schlieren, Switzerland
- *Correspondence: Laurène Pousse, ; Maria Amann,
| |
Collapse
|
40
|
Hino C, Xu Y, Xiao J, Baylink DJ, Reeves ME, Cao H. The potential role of the thymus in immunotherapies for acute myeloid leukemia. Front Immunol 2023; 14:1102517. [PMID: 36814919 PMCID: PMC9940763 DOI: 10.3389/fimmu.2023.1102517] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023] Open
Abstract
Understanding the factors which shape T-lymphocyte immunity is critical for the development and application of future immunotherapeutic strategies in treating hematological malignancies. The thymus, a specialized central lymphoid organ, plays important roles in generating a diverse T lymphocyte repertoire during the infantile and juvenile stages of humans. However, age-associated thymic involution and diseases or treatment associated injury result in a decline in its continuous role in the maintenance of T cell-mediated anti-tumor/virus immunity. Acute myeloid leukemia (AML) is an aggressive hematologic malignancy that mainly affects older adults, and the disease's progression is known to consist of an impaired immune surveillance including a reduction in naïve T cell output, a restriction in T cell receptor repertoire, and an increase in frequencies of regulatory T cells. As one of the most successful immunotherapies thus far developed for malignancy, T-cell-based adoptive cell therapies could be essential for the development of a durable effective treatment to eliminate residue leukemic cells (blasts) and prevent AML relapse. Thus, a detailed cellular and molecular landscape of how the adult thymus functions within the context of the AML microenvironment will provide new insights into both the immune-related pathogenesis and the regeneration of a functional immune system against leukemia in AML patients. Herein, we review the available evidence supporting the potential correlation between thymic dysfunction and T-lymphocyte impairment with the ontogeny of AML (II-VI). We then discuss how the thymus could impact current and future therapeutic approaches in AML (VII). Finally, we review various strategies to rejuvenate thymic function to improve the precision and efficacy of cancer immunotherapy (VIII).
Collapse
Affiliation(s)
- Christopher Hino
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Mark E Reeves
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| |
Collapse
|
41
|
Ren Z, Huang X, Lv Q, Lei Y, Shi H, Wang F, Wang M. High expression of B4GALT1 is associated with poor prognosis in acute myeloid leukemia. Front Genet 2022; 13:882004. [PMID: 36568388 PMCID: PMC9780537 DOI: 10.3389/fgene.2022.882004] [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: 02/23/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia is the most prevalent type of leukemia in adults and is prone to relapse and chemoresistance, with a low long-term survival rate. Therefore, the identification of quality biomarkers constitutes an urgent unmet need. High expression of beta-1,4-galactosyltransferase 1 (B4GALT1) has been observed in several cancer types; however, its function in acute myeloid leukemia has rarely been studied. Therefore, our study obtained gene expression data from The Cancer Genome Atlas (TCGA) database to analyze the relationship between B4GALT1 and LAML. We compared the expression of B4GALT1 in LAML and healthy samples using the Wilcoxon rank-sum test. Furthermore, the association between B4GALT1 and survival rates was investigated using Kaplan-Meier analysis and Cox regression. The nomogram obtained by Cox analysis predicts the effect of B4GALT1 on the prognosis. To assess B4GALT1-related genes' enrichment pathway and function and the correlation between B4GALT1 and immune features, GO/KEGG, protein-protein interaction network, and single sample gene set enrichment analysis were used. In addition, B4GALT1-specific siRNAs were used to verify the effect of B4GALT1 on apoptosis. The results showed that B4GALT1 is overexpressed in LAML and has some reference value in the diagnostic and prognostic assessment of LAML. Moreover, functional enrichment showed that B4GALT1 and its 63 associated genes were closely associated with the negative regulation of the apoptotic signaling pathway. Silencing B4GALT1 significantly promoted apoptosis. In addition, B4GALT1 expression was positively correlated with the infiltration levels of macrophages, regulatory T-cell (Tregs), and Th17 cells; in contrast, B4GALT1 expression was negatively correlated with the infiltration levels of T helper cells, Mast cells, and NK cells. In conclusion, our study shows that B4GALT1 may play a vital role in the occurrence of LAML.
Collapse
Affiliation(s)
- Zhihong Ren
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Xiaoyu Huang
- Henan Key Laboratory of Immunology and Targeted Drug, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Qing Lv
- School of Nursing and Health, Hennan University, Kaifeng, China
| | - Yiming Lei
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Haiqiang Shi
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Fanping Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China,*Correspondence: Fanping Wang, ; Mingyong Wang,
| | - Mingyong Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China,Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang Medical University, Xinxiang, China,*Correspondence: Fanping Wang, ; Mingyong Wang,
| |
Collapse
|
42
|
Rahmani S, Yazdanpanah N, Rezaei N. Natural killer cells and acute myeloid leukemia: promises and challenges. Cancer Immunol Immunother 2022; 71:2849-2867. [PMID: 35639116 PMCID: PMC10991240 DOI: 10.1007/s00262-022-03217-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
Acute myeloid leukemia (AML) is considered as one of the most malignant conditions of the bone marrow. Over the past few decades, despite substantial progresses in the management of AML, relapse remission remains a major problem. Natural killer cells (NK cells) are known as a unique component of the innate immune system. Due to swift tumor detection, distinct cytotoxic action, and extensive immune interaction, NK cells have been used in various cancer settings for decades. It has been a growing knowledge of therapeutic magnitudes ranging from adoptive NK cell transfer to chimeric antigen receptor NK cells, aiming to achieve better therapeutic responses in patients with AML. In this article, the potentials of NK cells for treatment of AML are highlighted, and challenges for such therapeutic methods are discussed. In addition, the clinical application of NK cells, mainly in patients with AML, is pictured according to the existing evidence.
Collapse
Affiliation(s)
- Shayan Rahmani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
43
|
Reale A, Khong T, Spencer A. Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment. J Clin Med 2022; 11:jcm11236892. [PMID: 36498469 PMCID: PMC9737553 DOI: 10.3390/jcm11236892] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
Abstract
Tumor cells actively incorporate molecules (e.g., proteins, lipids, RNA) into particles named extracellular vesicles (EVs). Several groups have demonstrated that EVs can be transferred to target (recipient) cells, making EVs an important means of intercellular communication. Indeed, EVs are able to modulate the functions of target cells by reprogramming signaling pathways. In a cancer context, EVs promote the formation of a supportive tumor microenvironment (TME) and (pre)metastatic niches. Recent studies have revealed that immune cells, tumor cells and their secretome, including EVs, promote changes in the TME and immunosuppressive functions of immune cells (e.g., natural killer, dendritic cells, T and B cells, monocytes, macrophages) that allow tumor cells to establish and propagate. Despite the growing knowledge on EVs and on their roles in cancer and as modulators of the immune response/escape, the translation into clinical practice remains in its early stages, hence requiring improved translational research in the EVs field. Here, we comprehensively review the current knowledge and most recent research on the roles of EVs in tumor immune evasion and immunosuppression in both solid tumors and hematological malignancies. We also highlight the clinical utility of EV-mediated immunosuppression targeting and EV-engineering. Importantly, we discuss the controversial role of EVs in cancer biology, current limitations and future perspectives to further the EV knowledge into clinical practice.
Collapse
Affiliation(s)
- Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne, VIC 3004, Australia
- Correspondence: (A.R.); (A.S.)
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne, VIC 3004, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne, VIC 3004, Australia
- Malignant Haematology and Stem Cell Transplantation, Department of Haematology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Clinical Hematology, Monash University, Melbourne, VIC 3004, Australia
- Correspondence: (A.R.); (A.S.)
| |
Collapse
|
44
|
Riether C. Regulation of hematopoietic and leukemia stem cells by regulatory T cells. Front Immunol 2022; 13:1049301. [PMID: 36405718 PMCID: PMC9666425 DOI: 10.3389/fimmu.2022.1049301] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/20/2022] [Indexed: 01/25/2023] Open
Abstract
Adult bone marrow (BM) hematopoietic stem cells (HSCs) are maintained in a quiescent state and sustain the continuous production of all types of blood cells. HSCs reside in a specialized microenvironment the so-called HSC niche, which equally promotes HSC self-renewal and differentiation to ensure the integrity of the HSC pool throughout life and to replenish hematopoietic cells after acute injury, infection or anemia. The processes of HSC self-renewal and differentiation are tightly controlled and are in great part regulated through cellular interactions with classical (e.g. mesenchymal stromal cells) and non-classical niche cells (e.g. immune cells). In myeloid leukemia, some of these regulatory mechanisms that evolved to maintain HSCs, to protect them from exhaustion and immune destruction and to minimize the risk of malignant transformation are hijacked/disrupted by leukemia stem cells (LSCs), the malignant counterpart of HSCs, to promote disease progression as well as resistance to therapy and immune control. CD4+ regulatory T cells (Tregs) are substantially enriched in the BM compared to other secondary lymphoid organs and are crucially involved in the establishment of an immune privileged niche to maintain HSC quiescence and to protect HSC integrity. In leukemia, Tregs frequencies in the BM even increase. Studies in mice and humans identified the accumulation of Tregs as a major immune-regulatory mechanism. As cure of leukemia implies the elimination of LSCs, the understanding of these immune-regulatory processes may be of particular importance for the development of future treatments of leukemia as targeting major immune escape mechanisms which revolutionized the treatment of solid tumors such as the blockade of the inhibitory checkpoint receptor programmed cell death protein 1 (PD-1) seems less efficacious in the treatment of leukemia. This review will summarize recent findings on the mechanisms by which Tregs regulate stem cells and adaptive immune cells in the BM during homeostasis and in leukemia.
Collapse
Affiliation(s)
- Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland,*Correspondence: Carsten Riether,
| |
Collapse
|
45
|
Guo X, Zhou X. Risk stratification of acute myeloid leukemia: Assessment using a novel prediction model based on ferroptosis-immune related genes. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:11821-11839. [PMID: 36653976 DOI: 10.3934/mbe.2022551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In acute myeloid leukemia (AML), the link between ferroptosis and the immune microenvironment has profound clinical significance. The objective of this study was to investigate the role of ferroptosis-immune related genes (FIRGs) in predicting the prognosis and therapeutic sensitivity in patients with AML. Using The Cancer Genome Atlas dataset, single sample gene set enrichment analysis was performed to calculate the ferroptosis score of AML samples. To search for FIRGs, differentially expressed genes between the high- and low-ferroptosis score groups were identified and then cross-screened with immune related genes. Univariate Cox and LASSO regression analyses were performed on the FIRGs to establish a prognostic risk score model with five signature FIRGs (BMP2, CCL3, EBI3, ELANE, and S100A6). The prognostic risk score model was then used to divide the patients into high- and low-risk groups. For external validation, two Gene Expression Omnibus cohorts were employed. Overall survival was poorer in the high-risk group than in the low-risk group. The novel risk score model was an independent prognostic factor for overall survival in patients with AML. Infiltrating immune cells were also linked to high-risk scores. Treatment targeting programmed cell death protein 1 may be more effective in high-risk patients. This FIRG-based prognostic risk model may aid in optimizing prognostic risk stratification and treatment of AML.
Collapse
Affiliation(s)
- Xing Guo
- Department of Hematology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Xiaogang Zhou
- Department of Hematology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| |
Collapse
|
46
|
Ding H, Feng Y, Xu J, Lin Z, Huang J, Wang F, Luo H, Gao Y, Zhai X, Wang X, Zhang L, Niu T, Zheng Y. A novel immune prognostic model of non-M3 acute myeloid leukemia. Am J Transl Res 2022; 14:5308-5325. [PMID: 36105048 PMCID: PMC9452334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
Acute myeloid leukemia (AML) is a common hematological malignancy in adults. AML patients exhibit clinical heterogeneity with complications of molecular basis. The leukemogenesis of AML involves immune escape, and the immunosuppression status of the patient might have great impact on AML treatment outcome. In this study, we established an immune prognostic model of AML using bioinformatics tools. With the data in the TCGA and GTEx datasets, we analyzed differentially expressed genes (DEGs) in non-M3 AML and identified 420 immune-related DEGs. Among which, 49 genes' expression was found to be related to AML prognosis based on univariate Cox regression analysis. Next, we established a prognostic model with these 49 genes in AML by LASSO regression and multivariate Cox regression analyses. In our model, the expressions of 5 immune genes, MIF, DEF6, OSM, MPO, AVPR1B, were used to stratify non-M3 AML patients' treatment outcome. A patient's risk score could be calculated as Risk Score=0.40081 × MIF (MIF expression) - 0.15201 × MPO + 0.78073 × DEF6 - 0.45192 × AVPR1B + 0.25912 × OSM. The area under the curve of the risk score signature was 0.8, 0.8, and 0.96 at 1 year, 3 years, and 5 years, respectively. The prognostic model was then validated internally by TCGA data and externally by GEO data. At last, the result of single-sample gene-set enrichment analysis demonstrated that compared with healthy samples, the abundance of non-turmeric immune cells was significantly repressed in AML. To summarize, we presented an immune-related 5-gene signature prognostic model in AML.
Collapse
Affiliation(s)
- Hong Ding
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Yu Feng
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Juan Xu
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Zhimei Lin
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
- Department of Hematology, The Affiliated Hospital of Chengdu UniversityChengdu 610081, Sichuan, China
| | - Jingcao Huang
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Hongmei Luo
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Yuhan Gao
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Xinyu Zhai
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Xin Wang
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Li Zhang
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| |
Collapse
|
47
|
Jacobsen N, Frisch T, Keiding N, Heilmann C, Sengeløv H, Madsen HO, Dickmeiss E, Ryder LP. Higher recipient pre-transplant FOXP3 mRNA expression is associated with acute leukaemia relapse after HSCT. EJHAEM 2022; 3:975-979. [PMID: 36051013 PMCID: PMC9421961 DOI: 10.1002/jha2.438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 06/15/2023]
Abstract
The effect of higher FOXP3 mRNA expression by recipient pre-transplant CD4+ T cells on leukaemia relapse was analysed in a series of 106 patients who received allogeneic haematopoietic stem cell transplantation after myeloablative conditioning with or without antithymocyte globulin (ATG) due to acute leukaemia in 1st or 2nd complete remission. FOXP3 mRNA was measured by qPCR in purified CD4+ T cells from blood obtained before conditioning. Higher FOXP3 mRNA expression was associated with an increased relapse risk when conditioning included ATG (n = 43, hazard ratio [HR] 11.0 [2.50-48.4], p = 0.00001). No effect was observed in patients not receiving ATG (HR 0.95 [0.53-1.81]).
Collapse
Affiliation(s)
- Niels Jacobsen
- Department of HematologyRigshospitalet University HospitalCopenhagenDenmark
| | - Tina Frisch
- Department of Clinical ImmunologyTissue Typing LaboratoryRigshospitalet University HospitalCopenhagenDenmark
| | - Niels Keiding
- Department of BiostatisticsFaculty of Health SciencesCopenhagen UniversityCopenhagenDenmark
| | - Carsten Heilmann
- Paediatric and Adolescence MedicineRigshospitalet University HospitalCopenhagenDenmark
| | - Henrik Sengeløv
- Department of HematologyRigshospitalet University HospitalCopenhagenDenmark
| | - Hans O. Madsen
- Department of Clinical ImmunologyTissue Typing LaboratoryRigshospitalet University HospitalCopenhagenDenmark
| | - Ebbe Dickmeiss
- Department of Clinical ImmunologyTissue Typing LaboratoryRigshospitalet University HospitalCopenhagenDenmark
| | - Lars P. Ryder
- Department of Clinical ImmunologyTissue Typing LaboratoryRigshospitalet University HospitalCopenhagenDenmark
| |
Collapse
|
48
|
Hino C, Pham B, Park D, Yang C, Nguyen MH, Kaur S, Reeves ME, Xu Y, Nishino K, Pu L, Kwon SM, Zhong JF, Zhang KK, Xie L, Chong EG, Chen CS, Nguyen V, Castillo DR, Cao H. Targeting the Tumor Microenvironment in Acute Myeloid Leukemia: The Future of Immunotherapy and Natural Products. Biomedicines 2022; 10:biomedicines10061410. [PMID: 35740430 PMCID: PMC9219790 DOI: 10.3390/biomedicines10061410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays an essential role in the development, proliferation, and survival of leukemic blasts in acute myeloid leukemia (AML). Within the bone marrow and peripheral blood, various phenotypically and functionally altered cells in the TME provide critical signals to suppress the anti-tumor immune response, allowing tumor cells to evade elimination. Thus, unraveling the complex interplay between AML and its microenvironment may have important clinical implications and are essential to directing the development of novel targeted therapies. This review summarizes recent advancements in our understanding of the AML TME and its ramifications on current immunotherapeutic strategies. We further review the role of natural products in modulating the TME to enhance response to immunotherapy.
Collapse
Affiliation(s)
- Christopher Hino
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Bryan Pham
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Daniel Park
- Department of Internal Medicine, School of Medicine, University of California San Francisco–Fresno, Fresno, CA 93701, USA;
| | - Chieh Yang
- Department of Internal Medicine, School of Medicine, University of California Riverside, Riverside, CA 92521, USA;
| | - Michael H.K. Nguyen
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Simmer Kaur
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Mark E. Reeves
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Yi Xu
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Kevin Nishino
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Lu Pu
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Sue Min Kwon
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Jiang F. Zhong
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA;
| | - Ke K. Zhang
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA; (K.K.Z.); (L.X.)
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA; (K.K.Z.); (L.X.)
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Esther G. Chong
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Chien-Shing Chen
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Vinh Nguyen
- Department of Biology, University of California Riverside, Riverside, CA 92521, USA;
| | - Dan Ran Castillo
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
- Correspondence: (D.R.C.); (H.C.)
| | - Huynh Cao
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
- Correspondence: (D.R.C.); (H.C.)
| |
Collapse
|
49
|
Pouya FD, Gazouli M, Rasmi Y, Lampropoulou DI, Nemati M. MicroRNAs and drug resistance in colorectal cancer with special focus on 5-fluorouracil. Mol Biol Rep 2022; 49:5165-5178. [PMID: 35212928 DOI: 10.1007/s11033-022-07227-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023]
Abstract
Colorectal cancer is globally one of the most common cancers in all age groups. The current chemotherapy combinations for colorectal cancer treatment include 5-fluorouracil-based regimens; however, drug resistance remains one of the main reasons for chemotherapy failure and disease recurrence. Many studies have determined colorectal cancer chemoresistance mechanisms such as drug efflux, cell cycle arrest, DNA damage repair, apoptosis, autophagy, vital enzymes, epigenetic, epithelial-mesenchymal transition, stem cells, and immune system suppression. Several microRNAs affect drug resistance by regulating the drug resistance-related target genes in colorectal cancer. These drug resistance-related miRNAs may be used as promising biomarkers for predicting drug response or as potential therapeutic targets for treating patients with colorectal cancer. This work reviews and discuss the role of selected microRNAs in 5-fluorouracil resistance and their molecular mechanisms in colorectal cancer.
Collapse
Affiliation(s)
- Fahima Danesh Pouya
- Department of Biochemistry, Faculty of Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Dimitra Ioanna Lampropoulou
- Second Department of Medical Oncology, General Oncology Hospital of Kifissia "Agioi Anargiroi", Nea Kifissia, Athens, Greece
| | - Mohadeseh Nemati
- Department of Biochemistry, Faculty of Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
50
|
Wang H, You Y, Zhu X. The Role of Exosomes in the Progression and Therapeutic Resistance of Hematological Malignancies. Front Oncol 2022; 12:887518. [PMID: 35692747 PMCID: PMC9178091 DOI: 10.3389/fonc.2022.887518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022] Open
Abstract
Exosomes are membrane limited structures which derive from cell membranes and cytoplasm. When released into extracellular space, they circulate through the extracellular fluid, including the peripheral blood and tissue fluid. Exosomes surface molecules mediate their targeting to specific recipient cells and deliver their contents to recipient cells by receptor-ligand interaction and/or phagocytosis and/or endocytosis or direct fusion with cell membrane. Exosomes contain many functional molecules, including nucleic acids (DNAs, mRNAs, non-coding RNAs), proteins (transcription factors, enzymes), and lipids which have biological activity. By passing these cargos, exosomes can transfer information between cells. In this way, exosomes are extensively involved in physiological and pathological processes, such as angiogenesis, matrix reprogramming, coagulation, tumor progression. In recent years, researcher have found that exosomes from malignant tumors can mediate information exchange between tumor cells or between tumor cells and non-tumor cells, thereby promoting tumor survival, progression, and resistance to therapy. In this review, we discuss the pro-tumor and anti-therapeutic effects of exosomes in hematological malignancies, hoping to contribute to the early conquest of hematological malignancy.
Collapse
Affiliation(s)
- Haobing Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong You
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiaojian Zhu,
| |
Collapse
|