1
|
Zou J, Ding W, Chen Y, Pan X, Fu X, Zheng S, Lin Y, Xia C, Ma Y, Zhang X, Wu S, Gao F. Incorporating zinc coordination driven nanozyme into chitosan and hyaluronic acid based nanoplatform for scavenging H 2S/ROS in managing inflammatory bowel disease. Carbohydr Polym 2025; 361:123501. [PMID: 40368544 DOI: 10.1016/j.carbpol.2025.123501] [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: 11/18/2024] [Revised: 02/25/2025] [Accepted: 03/12/2025] [Indexed: 05/16/2025]
Abstract
Inflammatory bowel disease (IBD) causes damage to patients, and therapy remains a crucial challenge owing to the presence of excessive reactive oxygen species (ROS) and hydrogen sulfide (H2S) in colonic inflammatory circulation. Herein, we report a shikonin (SK) and zinc (Zn) driven coordination nanozyme (SK-Zn, S-Z), which effectively scavenged ROS and H2S. Furthermore, a pH responsive targeted polymer nanoplatform (S-Z@ChF), consisting of S-Z, chitosan, hyaluronic acid and poly ferulic acid, was developed for the oral treatment of colitis. In vivo experiments revealed that the nanoplatform could target colonic inflammatory lesions, relieve inflammatory index and restore the colonic mechanical barrier. The transcriptomics and pharmacodynamic mechanism analysis revealed that S-Z@ChF scavenged ROS, inhibited nuclear factor kappa-B (NF-κB) and pyruvate kinase isozyme type M2 (PKM2) and signal transducer and activator of transcription 3 (STAT3) pathways, balancing the level of macrophage polarization and pro-inflammatory enzyme. Additionally, S-Z@ChF reliably regulated the gut microbiota during H2S scavenging. In conclusion, S-Z@ChF, which provides multiple anti-inflammatory pathways and microenvironment reprogramming support for blocking inflammatory circulation, was found to be biologically safe, with significant potential for clinical application in IBD.
Collapse
Affiliation(s)
- Jiafeng Zou
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenxing Ding
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - You Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xier Pan
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiuzhi Fu
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Shulei Zheng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yiting Lin
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Chuanhe Xia
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ying Ma
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaochun Zhang
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Shuang Wu
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Feng Gao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
2
|
Aakko S, Ylitalo A, Kuusanmäki H, Rannikko JH, Björkman M, Mandelin J, Heckman CA, Kontro M, Hollmén M. CLEVER-1 targeting antibody, bexmarilimab, supports HLA-DR expression and alters ex vivo responsiveness to azacitidine and venetoclax in myeloid malignancies. Sci Rep 2025; 15:16775. [PMID: 40369178 PMCID: PMC12078569 DOI: 10.1038/s41598-025-01675-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 05/07/2025] [Indexed: 05/16/2025] Open
Abstract
Treatment of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) requires new therapy options, especially for patients uneligible for intense chemotherapy or with relapsed or refractory disease. CLEVER-1 is a myeloid checkpoint protein, which can be targeted with a therapeutic function blocking antibody, bexmarilimab. Bexmarilimab has shown clinical efficacy in different solid tumors. Here, we show preclinical data demonstrating expression of CLEVER-1 on immature malignant myeloid cells and their derivates in MDS and AML bone marrow samples and AML cell lines. Highest CLEVER-1 levels were observed in AML with monocytic differentiation. Ex vivo treatment of AML/MDS bone marrow samples with bexmarilimab led to an increase in antigen-presenting human leukocyte antigen DR isotype (HLA-DR) molecule expression. Combination of bexmarilimab with current standard-of-care (SoC) drugs, azacitidine and venetoclax, showed potential for HLA-DR induction and enhanced killing of leukemic cells, respectively. Our non-clinical findings support the feasibility of CLEVER-1 inhibition in AML/MDS to induce antigen presentating molecule expression and potentially, an anti-leukemic effect together with SoC. Therapeutic targeting of CLEVER-1 with bexmarilimab is currently undergoing clinical investigation in the BEXMAB trial (NCT05428969).
Collapse
MESH Headings
- Humans
- Azacitidine/pharmacology
- Azacitidine/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/metabolism
- Myelodysplastic Syndromes/pathology
- Antibodies, Monoclonal, Humanized/pharmacology
- HLA-DR Antigens/metabolism
- Cell Line, Tumor
Collapse
Affiliation(s)
- Sofia Aakko
- Faron Pharmaceuticals Ltd, Turku, Finland
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arno Ylitalo
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre (BRIC) and Novo Nordisk Foundation Center For Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Jenna H Rannikko
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | | | | | - Caroline A Heckman
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mika Kontro
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Maija Hollmén
- Faron Pharmaceuticals Ltd, Turku, Finland.
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland.
| |
Collapse
|
3
|
Guan Z, Zhang Z, Wang K, Qiao S, Ma T, Wu L. Targeting myeloid cells for hematological malignancies: the present and future. Biomark Res 2025; 13:59. [PMID: 40205623 PMCID: PMC11983845 DOI: 10.1186/s40364-025-00775-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
Hematological malignancies are a diverse group of cancers that originate in the blood and bone marrow and are characterized by the abnormal proliferation and differentiation of hematopoietic cells. Myeloid blasts, which are derived from normal myeloid progenitors, play a central role in these diseases by disrupting hematopoiesis and driving disease progression. In addition, other myeloid cells, including tumor-associated macrophages and myeloid-derived suppressor cells, adapt dynamically to the tumor microenvironment, where they can promote immune evasion and resistance to treatment. This review explores the unique characteristics and pathogenic mechanisms of myeloid blasts, the immunosuppressive roles of myeloid cells, and their complex interactions within the TME. Furthermore, we highlight emerging therapeutic approaches targeting myeloid cells, focusing on strategies to reprogram their functions, inhibit their suppressive effects, or eliminate pathological populations altogether, as well as the latest preclinical and clinical trials advancing these approaches. By integrating insights from these studies, we aim to provide a comprehensive understanding of the roles of myeloid cells in hematological malignancies and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Zihui Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Peking University First Hospital, Beijing, 100034, China
| | - Zhengqi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Kaiyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shukai Qiao
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Teng Ma
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
| | - Lina Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
4
|
Yin J, Luo J, Wang L, Liu L, Liu L. STAB1 Promotes Acute Myeloid Leukemia Progression by Activating the IKK/NF-κB Pathway and Increasing M2 Macrophage Polarization. Cancer Sci 2025. [PMID: 40083109 DOI: 10.1111/cas.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/10/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
As a multifunctional scavenger receptor, stabilin-1 (STAB1) has been identified to induce chronic inflammation and promote cancer progression. Although in silico studies from multiple data sets showed that STAB1 might facilitate the progression of acute myeloid leukemia (AML) and drug resistance, the real impacts of STAB1 expression on AML patients and the detailed mechanisms remain unclear. Herein, we found that a higher expression of STAB1 is associated with a worse prognosis in AML patients. Subsequent in vitro experiments demonstrated that STAB1 knockdown suppressed proliferation and promoted apoptosis through regulating the IKK/NF-κB pathway in human AML cell lines HEL and NB4. In addition, in vivo studies showed that STAB1 silencing prolonged survival, reduced proliferation, and inhibited aggressiveness of AML cells in xenograft mouse models. Moreover, we investigated the impact of STAB1 expression in AML cells on macrophage differentiation and found that co-culture of macrophages with conditioned medium from STAB1-knockdown AML cells reduced M2 polarization of macrophages. Taken together, our study suggests that STAB1 promotes growth and aggressiveness of AML cells through activating the IKK/NF-κB pathway while also regulating M2 macrophage polarization within the chronic inflammatory environment. Therefore, targeting STAB1 could be a potential therapeutic strategy for treating AML.
Collapse
Affiliation(s)
- Jiaxiu Yin
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Oncology and Hematology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Lanxiang Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Haubner S, Subklewe M, Sadelain M. Honing CAR T cells to tackle acute myeloid leukemia. Blood 2025; 145:1113-1125. [PMID: 39630061 DOI: 10.1182/blood.2024024063] [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: 09/18/2024] [Accepted: 11/15/2024] [Indexed: 03/14/2025] Open
Abstract
ABSTRACT Acute myeloid leukemia (AML) remains a dismal disease with poor prognosis, particularly in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR) therapy has yielded remarkable clinical results in other leukemias and thus has, in principle, the potential to achieve similar outcomes in R/R AML. Redirecting the approved CD19-specific CAR designs against the myeloid antigens CD33, CD123, or CLEC12A has occasionally yielded morphologic leukemia-free states but has so far been marred by threatening myeloablation and early relapses. These safety and efficacy limitations are largely due to the challenge of identifying suitable target antigens and designing adequate receptors for effective recognition and safe elimination of AML. Building on lessons learned from the initial clinical attempts, a new wave of CAR strategies relying on alternative target antigens and innovative CAR designs is about to enter clinical evaluation. Adapted multiantigen targeting, logic gating, and emerging cell engineering solutions offer new possibilities to better direct T-cell specificity and sensitivity toward AML. Pharmacologic modulation and genetic epitope engineering may extend these approaches by augmenting target expression in AML cells or minimizing target expression in normal hematopoietic cells. On/off switches or CAR T-cell depletion may curb excessive or deleterious CAR activity. Investigation of AML-intrinsic resistance and leukemic microenvironmental factors is poised to reveal additional targetable AML vulnerabilities. We summarize here the findings, challenges, and new developments of CAR therapy for AML. These illustrate the need to specifically adapt CAR strategies to the complex biology of AML to achieve better therapeutic outcomes.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- T-Lymphocytes/immunology
- Animals
- Antigens, Neoplasm/immunology
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Sascha Haubner
- Columbia Initiative in Cell Engineering and Therapy, Department of Medicine, Columbia University, New York, NY
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Michel Sadelain
- Columbia Initiative in Cell Engineering and Therapy, Department of Medicine, Columbia University, New York, NY
| |
Collapse
|
6
|
Lecornec N, Duchmann M, Itzykson R. Single-cell sequencing applications in acute myeloid leukemia. Leuk Lymphoma 2025; 66:175-189. [PMID: 39496597 DOI: 10.1080/10428194.2024.2422833] [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: 07/23/2024] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of malignancies with poor prognosis. AML result from the proliferation of immature myeloid cells blocked at a variable stage of differentiation. Beyond inter-patient heterogeneity, AMLs are characterized by genetic and phenotypic intra-patient heterogeneity. Despite major advances in deciphering AML biology with bulk sequencing studies, pivotal questions remain unanswered. Analyses at the single-cell level could thus transform our understanding of these neoplasms. We review recent progresses in single-cell sequencing technologies from cell processing to bioinformatic pipelines. We next discuss how single-cell applications have helped understand the genetic and functional intra-leukemic heterogeneity, emphasizing aspects related to leukemic stem cells, clonal evolution and measurable residual disease (MRD) monitoring. We finally delineate how single-cell technologies could be implemented in routine clinical practice to improve patient management.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Single-Cell Analysis/methods
- Neoplasm, Residual/genetics
- Neoplasm, Residual/diagnosis
- Biomarkers, Tumor/genetics
- High-Throughput Nucleotide Sequencing/methods
- Clonal Evolution
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Computational Biology/methods
- Prognosis
Collapse
Affiliation(s)
- Nicolas Lecornec
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département d'Immuno-Hématologie Pédiatrique, Hôpital Robert-Debré, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Matthieu Duchmann
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Raphael Itzykson
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
7
|
Zhou D, Zheng Z, Li Y, Zhang J, Lu X, Zheng H, Dai J. Integrated multi-omics and machine learning reveal a gefitinib resistance signature for prognosis and treatment response in lung adenocarcinoma. IUBMB Life 2025; 77:e2930. [PMID: 39612355 DOI: 10.1002/iub.2930] [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: 08/03/2024] [Accepted: 09/11/2024] [Indexed: 12/01/2024]
Abstract
Gefitinib resistance (GR) presents a significant challenge in treating lung adenocarcinoma (LUAD), highlighting the need for alternative therapies. This study explores the genetic basis of GR to improve prediction, prevention, and treatment strategies. We utilized public databases to obtain GR gene sets, single-cell data, and transcriptome data, applying univariate and multivariate regression analyses alongside machine learning to identify key genes and develop a predictive signature. The signature's performance was evaluated using survival analysis and time-dependent ROC curves on internal and external datasets. Enrichment and tumor immune microenvironment analyses were conducted to understand the mechanistic roles of the signature genes in GR. Our analysis identified a robust 22-gene signature with strong predictive performance across validation datasets. This signature was significantly associated with chromosomal processes, DNA replication, immune cell infiltration, and various immune scores based on enrichment and tumor microenvironment analyses. Importantly, the signature also showed potential in predicting the efficacy of immunotherapy in LUAD patients. Moreover, we identified alternative agents to gefitinib that could offer improved therapeutic outcomes for high-risk and low-risk patient groups, thereby guiding treatment strategies for gefitinib-resistant patients. In conclusion, the 22-gene signature not only predicts prognosis and immunotherapy efficacy in gefitinib-resistant LUAD patients but also provides novel insights into non-immunotherapy treatment options.
Collapse
Affiliation(s)
- Dong Zhou
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhi Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanqi Li
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiao Lu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jigang Dai
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
8
|
Chen T, Sun W, Xu ZJ. The immune mechanisms of acute exacerbations of idiopathic pulmonary fibrosis. Front Immunol 2024; 15:1450688. [PMID: 39737178 PMCID: PMC11682984 DOI: 10.3389/fimmu.2024.1450688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Acute exacerbations of idiopathic pulmonary fibrosis (AE-IPF) are the leading cause of mortality among patients with IPF. There is still a lack of effective treatments for AE-IPF, resulting in a hospitalization mortality rate as high as 70%-80%. To reveal the complicated mechanism of AE-IPF, more attention has been paid to its disturbed immune environment, as patients with IPF exhibit deficiencies in pathogen defense due to local immune dysregulation. During the development of AE-IPF, the classical stimulatory signals in adaptive immunity are inhibited, while the nonclassical immune reactions (Th17) are activated, attracting numerous neutrophils and monocytes to lung tissues. However, there is limited information about the specific changes in the immune response of AE-IPF. We summarized the immune mechanisms of AE-IPF in this review.
Collapse
Affiliation(s)
- Tao Chen
- Department of Respiratory and Critical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Sun
- Department of Respiratory and Critical Medicine, The second hospital of Tianjin Medical University, Tianjin, China
| | - Zuo-jun Xu
- Department of Respiratory and Critical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
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
|
10
|
Yoon C, Kim HK, Ham YS, Gil WJ, Mun SJ, Cho E, Yuk JM, Yang CS. Toxoplasma gondii macrophage migration inhibitory factor shows anti- Mycobacterium tuberculosis potential via AZIN1/STAT1 interaction. SCIENCE ADVANCES 2024; 10:eadq0101. [PMID: 39453997 PMCID: PMC11506136 DOI: 10.1126/sciadv.adq0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
Mycobacterium tuberculosis (MTB) is a pathogenic bacterium, belonging to the family Mycobacteriaceae, that causes tuberculosis (TB). Toxoplasma gondii macrophage migration inhibitory factor (TgMIF), a protein homolog of macrophage migration inhibitory factor, has been explored for its potential to modulate immune responses during MTB infections. We observed that TgMIF that interacts with CD74, antizyme inhibitor 1 (AZIN1), and signal transducer and activator of transcription 1 (STAT1) modulates endocytosis, restoration of mitochondrial function, and macrophage polarization, respectively. These interactions promote therapeutic efficacy in mice infected with MTB, thereby presenting a potential route to host-directed therapy development. Furthermore, TgMIF, in combination with first-line TB drugs, significantly inhibited drug-resistant MTB strains, including multidrug-resistant TB. These results demonstrate that TgMIF is potentially a multifaceted therapeutic agent against TB, acting through immune modulation, enhancement of mitochondrial function, and dependent on STAT1 and AZIN1 pathways.
Collapse
Affiliation(s)
- Chanjin Yoon
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588, South Korea
| | - Hyo Keun Kim
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Yu Seong Ham
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Woo Jin Gil
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Seok-Jun Mun
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Euni Cho
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Department of Medicinal and Life Science, Hanyang University, Ansan 15588, South Korea
| |
Collapse
|
11
|
Zhang D, Jiang Y, Wang M, Zhao J, Wan J, Li Z, Huang D, Yu J, Li J, Liu J, Huang F, Hao S. A novel costimulatory molecule gene-modified leukemia cell-derived exosome enhances the anti-leukemia efficacy of DC vaccine in mouse models. Vaccine 2024; 42:126097. [PMID: 38960787 DOI: 10.1016/j.vaccine.2024.06.064] [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: 12/18/2023] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVES Leukemia cell-derived exosomes (LEXs), carrying leukemia cell-specific antigens, can serve as a source of antigen for dendritic cell (DC) vaccine loading. However, LEX-targeted DC-based vaccines have demonstrated limited antitumor immune effects in clinical trials, attributed to the low immunogenicity of LEXs and the scant levels of costimulatory molecules on DCs. The costimulatory molecules CD80 and CD86, which are crucial to DC function, play a significant role in enhancing immune efficacy. In this study, we explored the anti-leukemia immune response of costimulatory molecule gene-modified LEX-targeted DCs (LEX-8086) in vitro and in animal models. METHODS DCs were incubated with LEX-8086 to produce LEX-8086-targeted DCs (DCsLEX-8086). ELISA, cytotoxicity assays and flow cytometry utilized to assess the antitumor efficacy of DCsLEX8086 in vitro. Flow cytometry was used to evaluate the immunomodulatory function of DCsLEX8086 in animal models. RESULTS Our findings indicated that LEX-8086 enhanced the maturation and antigen-presenting ability of DCs. Immunization with DCsLEX8086 significantly activated CD8+ T cells and boosted the CTL response in vitro. More importantly, DCsLEX-8086 effectively suppressed tumor growth and exerted anti-leukemia effects in both prophylactic and therapeutic animal models. Furthermore, DCsLEX-8086 promoted the proportion of CD4+ T cells, CD8+ T cells and M1 macrophages in the tumor environments both prophylactically and therapeutically. Treatment with DCsLEX-8086 showed no significant difference in the levels of M2 macrophages but decreased the proportion of Tregs within the tumor bed during therapeutic experiments. CONCLUSION The results suggested that DCsLEX-8086 induces a more effective anti-leukemia immunity compared to DCsLEX-null in vivo and in vitro. DCsLEX-8086 might achieve antitumor effects by elevating the numbers of CD4+ T cells, CD8+ T cells, and M1 macrophages in tumors. Our findings indicate that DCsLEX-8086 could be leveraged to develop a new, highly effective vaccine for anti-leukemia immunity.
Collapse
Affiliation(s)
- Difan Zhang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Jiang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minghui Wang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhao
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiangbo Wan
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhichao Li
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Li
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayu Liu
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Huang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Siguo Hao
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Mesaros O, Onciul M, Matei E, Joldes C, Jimbu L, Neaga A, Serban O, Zdrenghea M, Nanut AM. Macrophages as Potential Therapeutic Targets in Acute Myeloid Leukemia. Biomedicines 2024; 12:2306. [PMID: 39457618 PMCID: PMC11505058 DOI: 10.3390/biomedicines12102306] [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: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous malignant hemopathy, and although new drugs have emerged recently, current treatment options still show limited efficacy. Therapy resistance remains a major concern due to its contribution to treatment failure, disease relapse, and increased mortality among patients. The underlying mechanisms of resistance to therapy are not fully understood, and it is crucial to address this challenge to improve therapy. Macrophages are immune cells found within the bone marrow microenvironment (BMME), of critical importance for leukemia development and progression. One defining feature of macrophages is their plasticity, which allows them to adapt to the variations in the microenvironment. While this adaptability is advantageous during wound healing, it can also be exploited in cancer scenarios. Thus, clinical and preclinical investigations that target macrophages as a therapeutic strategy appear promising. Existing research indicates that targeting macrophages could enhance the effectiveness of current AML treatments. This review addresses the importance of macrophages as therapeutic targets including relevant drugs investigated in clinical trials such as pexidartinib, magrolimab or bexmarilimab, but also provides new insights into lesser-known therapies, like macrophage receptor with a collagenous structure (MACRO) inhibitors and Toll-like receptor (TLR) agonists.
Collapse
Affiliation(s)
- Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Madalina Onciul
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
| | - Emilia Matei
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Pathology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Corina Joldes
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, 19-21 Croitorilor Str., 400162 Cluj-Napoca, Romania
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Alexandra Neaga
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Oana Serban
- Regina Maria” Regional Laboratory in Cluj-Napoca, 109 Observatorului Str., 400363 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Ana Maria Nanut
- Regina Maria” Regional Laboratory in Cluj-Napoca, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| |
Collapse
|
13
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [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: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
14
|
Boët E, Saland E, Skuli S, Griessinger E, Sarry JE. [ Mitohormesis: a key driver of the therapy resistance in cancer cells]. C R Biol 2024; 347:59-75. [PMID: 39171610 DOI: 10.5802/crbiol.154] [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: 03/21/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 08/23/2024]
Abstract
A large body of literature highlights the importance of energy metabolism in the response of haematological malignancies to therapy. In this review, we are particularly interested in acute myeloid leukaemia, where mitochondrial metabolism plays a key role in response and resistance to treatment. We describe the new concept of mitohormesis in the response to therapy-induced stress and in the initiation of relapse in this disease.
Collapse
|
15
|
Yu S, Jiang J. Immune infiltration-related genes regulate the progression of AML by invading the bone marrow microenvironment. Front Immunol 2024; 15:1409945. [PMID: 39072320 PMCID: PMC11272452 DOI: 10.3389/fimmu.2024.1409945] [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: 03/31/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
In this study, we try to find the pathogenic role of immune-related genes in the bone marrow microenvironment of AML. Through WGCNA, seven modules were obtained, among which the turquoise module containing 1793 genes was highly correlated with the immune infiltration score. By unsupervised clustering, the turquoise module was divided into two clusters: the intersection of clinically significant genes in the TCGA and DEGs to obtain 178 genes for mutation analysis, followed by obtaining 17 genes with high mutation frequency. Subsequently, these 17 genes were subjected to LASSO regression analysis to construct a riskscore model of 8 hub genes. The TIMER database, ImmuCellAI portal website, and ssGSEA elucidate that the hub genes and risk scores are closely related to immune cell infiltration into the bone marrow microenvironment. In addition, we also validated the relative expression levels of hub genes using the TCGA database and GSE114868, and additional expression levels of hub genes in AML cell lines in vitro. Therefore, we constructed an immune infiltration-related gene model that identify 8 hub genes with good risk stratification and predictive prognosis for AML.
Collapse
Affiliation(s)
- Shuangmei Yu
- Department of Radio-immunity, Heilongjiang Provincial Hospital, Harbin, China
| | - Jiquan Jiang
- Department of Laboratory Diagnosis, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
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
|
17
|
Li K, Nie H, Jin R, Wu X. Mesenchymal stem cells-macrophages crosstalk and myeloid malignancy. Front Immunol 2024; 15:1397005. [PMID: 38779660 PMCID: PMC11109455 DOI: 10.3389/fimmu.2024.1397005] [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: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
As major components of the tumor microenvironment, both mesenchymal stem cells (MSCs) and macrophages can be remodelled and exhibit different phenotypes and functions during tumor initiation and progression. In recent years, increasing evidence has shown that tumor-associated macrophages (TAMs) play a crucial role in the growth, metastasis, and chemotherapy resistance of hematological malignancies, and are associated with poor prognosis. Consequently, TAMs have emerged as promising therapeutic targets. Notably, MSCs exert a profound influence on modulating immune cell functions such as macrophages and granulocytes, thereby playing a crucial role in shaping the immunosuppressive microenvironment surrounding tumors. However, in hematological malignancies, the cellular and molecular mechanisms underlying the interaction between MSCs and macrophages have not been clearly elucidated. In this review, we provide an overview of the role of TAMs in various common hematological malignancies, and discuss the latest advances in understanding the interaction between MSCs and macrophages in disease progression. Additionally, potential therapeutic approaches targeting this relationship are outlined.
Collapse
Affiliation(s)
- Kun Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Nie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Burk AC, Apostolova P. Metabolic instruction of the graft-versus-leukemia immunity. Front Immunol 2024; 15:1347492. [PMID: 38500877 PMCID: PMC10944922 DOI: 10.3389/fimmu.2024.1347492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/05/2024] [Indexed: 03/20/2024] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is frequently performed to cure hematological malignancies, such as acute myeloid leukemia (AML), through the graft-versus-leukemia (GVL) effect. In this immunological process, donor immune cells eliminate residual cancer cells in the patient and exert tumor control through immunosurveillance. However, GVL failure and subsequent leukemia relapse are frequent and associated with a dismal prognosis. A better understanding of the mechanisms underlying AML immune evasion is essential for developing novel therapeutic strategies to boost the GVL effect. Cellular metabolism has emerged as an essential regulator of survival and cell fate for both cancer and immune cells. Leukemia and T cells utilize specific metabolic programs, including the orchestrated use of glucose, amino acids, and fatty acids, to support their growth and function. Besides regulating cell-intrinsic processes, metabolism shapes the extracellular environment and plays an important role in cell-cell communication. This review focuses on recent advances in the understanding of how metabolism might affect the anti-leukemia immune response. First, we provide a general overview of the mechanisms of immune escape after allo-HCT and an introduction to leukemia and T cell metabolism. Further, we discuss how leukemia and myeloid cell metabolism contribute to an altered microenvironment that impairs T cell function. Next, we review the literature linking metabolic processes in AML cells with their inhibitory checkpoint ligand expression. Finally, we focus on recent findings concerning the role of systemic metabolism in sustained GVL efficacy. While the majority of evidence in the field still stems from basic and preclinical studies, we discuss translational findings and propose further avenues for bridging the gap between bench and bedside.
Collapse
Affiliation(s)
- Ann-Cathrin Burk
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, Freiburg, Germany
- Department of Medicine I, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Petya Apostolova
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
19
|
Chen C, Ding J, Ma Z, Xie Y, Zhang L, Zhu D. Exosome-Delivered EGFR Induced by Acidic Bile Salts Regulates Macrophage M2 Polarization to Promote Esophageal Adenocarcinoma Cell Proliferation. Onco Targets Ther 2024; 17:113-128. [PMID: 38384996 PMCID: PMC10879628 DOI: 10.2147/ott.s437560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/05/2024] [Indexed: 02/23/2024] Open
Abstract
Purpose Chronic gastroesophageal reflux disease (GERD) causes the abnormal reflux of acid and bile salts, which would induce Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC). EGFR, as one of main components of the exosome, plays an important role in cancer progression. Here, we investigated the role of acidic bile salts (ABS)-induced exosomal EGFR in EAC cell proliferation. Methods Electronic microscopic examination and Western blot were used to identify exosomes. Western blot, siRNA transfection, enzyme-linked immunosorbent assay, qRT-PCR, cell viability detection, mouse xenograft tumor models, and immunohistochemical staining were performed to study the function of ABS-induced exosomal EGFR in cell proliferation. Results We found that ABS improved the exosomal EGFR level of normal human esophageal epithelial cells, BE cells, and BE-associated adenocarcinoma cells. The results were confirmed in the serum-derived exosomes from healthy persons and patients suffering from GERD, BE with or without GERD, and EAC with or without GERD. Moreover, cell line-derived exosomal EGFR was found to promote macrophage M2 polarization through the PI3K-AKT pathway. The co-incubation medium of macrophages and exosomes improved cell proliferation and tumor growth, which depended on the exosomal EGFR level. CCL18 was identified as the most effective component of the co-incubation medium to promote EAC cell proliferation by binding to its receptor PITPNM3 in vitro and in vivo. Conclusion Our findings demonstrate that ABS-induced exosomal EGFR regulates macrophage M2 polarization to promote EAC proliferation. This study provides an important insight into the role of ABS in EAC development.
Collapse
Affiliation(s)
- Chuangui Chen
- Department of Minimally Invasive Esophagus Surgery, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People’s Republic of China
- Beijing Viewsolid Biotechnology Co., LTD, Beijing, 102200, People’s Republic of China
| | - Jinsheng Ding
- Department of Pancreatic Cancer, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People’s Republic of China
| | - Zhao Ma
- Department of Minimally Invasive Esophagus Surgery, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People’s Republic of China
| | - Yongjie Xie
- Department of Pancreatic Cancer, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People’s Republic of China
| | - Linhua Zhang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, People’s Republic of China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, People’s Republic of China
| |
Collapse
|
20
|
Weinhäuser I, Pereira-Martins DA, Hilberink JR, Brouwers-Vos A, Rego EM, Huls G, Schuringa JJ. Thiostrepton induces cell death of acute myeloid leukemia blasts and the associated macrophage population. Haematologica 2024; 109:639-645. [PMID: 37646656 PMCID: PMC10828769 DOI: 10.3324/haematol.2023.283621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Affiliation(s)
- Isabel Weinhäuser
- Department of Experimental Hematology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Department of Internal Medicine, Medical School of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil; Center for Cell Based Therapy, University of São Paulo, Ribeirao Preto, Brazil; Authors contributed equally to this study
| | - Diego A Pereira-Martins
- Department of Experimental Hematology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Department of Internal Medicine, Medical School of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil; Center for Cell Based Therapy, University of São Paulo, Ribeirao Preto, Brazil; Authors contributed equally to this study
| | - Jacobien R Hilberink
- Department of Experimental Hematology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Authors contributed equally to this study
| | - Annet Brouwers-Vos
- Department of Experimental Hematology, University Medical Centre Groningen, University of Groningen, Groningen
| | - Eduardo M Rego
- Center for Cell Based Therapy, University of São Paulo, Ribeirao Preto
| | - Gerwin Huls
- Department of Experimental Hematology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Lead contact authors
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Lead contact authors.
| |
Collapse
|
21
|
Liu J, Jiang P, Lu Z, Yu Z, Qian P. Decoding leukemia at the single-cell level: clonal architecture, classification, microenvironment, and drug resistance. Exp Hematol Oncol 2024; 13:12. [PMID: 38291542 PMCID: PMC10826069 DOI: 10.1186/s40164-024-00479-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
Leukemias are refractory hematological malignancies, characterized by marked intrinsic heterogeneity which poses significant obstacles to effective treatment. However, traditional bulk sequencing techniques have not been able to effectively unravel the heterogeneity among individual tumor cells. With the emergence of single-cell sequencing technology, it has bestowed upon us an unprecedented resolution to comprehend the mechanisms underlying leukemogenesis and drug resistance across various levels, including the genome, epigenome, transcriptome and proteome. Here, we provide an overview of the currently prevalent single-cell sequencing technologies and a detailed summary of single-cell studies conducted on leukemia, with a specific focus on four key aspects: (1) leukemia's clonal architecture, (2) frameworks to determine leukemia subtypes, (3) tumor microenvironment (TME) and (4) the drug-resistant mechanisms of leukemia. This review provides a comprehensive summary of current single-cell studies on leukemia and highlights the markers and mechanisms that show promising clinical implications for the diagnosis and treatment of leukemia.
Collapse
Affiliation(s)
- Jianche Liu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- International Campus, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, 718 East Haizhou Road, Haining, 314400, China
| | - Penglei Jiang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, 310058, China
| | - Zezhen Lu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- International Campus, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, 718 East Haizhou Road, Haining, 314400, China
| | - Zebin Yu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, 310058, China
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
22
|
Dancik GM, Varisli L, Tolan V, Vlahopoulos S. Aldehyde Dehydrogenase Genes as Prospective Actionable Targets in Acute Myeloid Leukemia. Genes (Basel) 2023; 14:1807. [PMID: 37761947 PMCID: PMC10531322 DOI: 10.3390/genes14091807] [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: 07/13/2023] [Revised: 08/29/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
It has been previously shown that the aldehyde dehydrogenase (ALDH) family member ALDH1A1 has a significant association with acute myeloid leukemia (AML) patient risk group classification and that AML cells lacking ALDH1A1 expression can be readily killed via chemotherapy. In the past, however, a redundancy between the activities of subgroup members of the ALDH family has hampered the search for conclusive evidence to address the role of specific ALDH genes. Here, we describe the bioinformatics evaluation of all nineteen member genes of the ALDH family as prospective actionable targets for the development of methods aimed to improve AML treatment. We implicate ALDH1A1 in the development of recurrent AML, and we show that from the nineteen members of the ALDH family, ALDH1A1 and ALDH2 have the strongest association with AML patient risk group classification. Furthermore, we discover that the sum of the expression values for RNA from the genes, ALDH1A1 and ALDH2, has a stronger association with AML patient risk group classification and survival than either one gene alone does. In conclusion, we identify ALDH1A1 and ALDH2 as prospective actionable targets for the treatment of AML in high-risk patients. Substances that inhibit both enzymatic activities constitute potentially effective pharmaceutics.
Collapse
Affiliation(s)
- Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey; (L.V.); (V.T.)
| | - Veysel Tolan
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey; (L.V.); (V.T.)
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| |
Collapse
|