1
|
Lv J, Zhang Y, Wu Q, Jiang P, Lin Y. Inhibition of SIRT4 promotes bladder cancer progression and immune escape via attenuating CD8 + T cells function. Int Immunopharmacol 2025; 147:113906. [PMID: 39756164 DOI: 10.1016/j.intimp.2024.113906] [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: 11/04/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Bladder cancer (BCa) is one of the most common malignancies of the urinary system and is characterized by a high recurrence rate and significant mortality. Sirtuin 4 (SIRT4), a member of the NAD+-dependent deacetylase and ADP-ribosyltransferase family, is involved in regulating cellular metabolism, DNA repair, and longevity, potentially influencing tumor progression and immune escape. This study aimed to elucidate the role of SIRT4 in BCa. METHODS The correlation between the sirtuin family and immunotherapy sensitivity in BCa patients was analyzed via IMvigor210 data. The clinical significance and immunological role of SIRT4 across multiple cancer types were assessed by evaluating its associations with clinicopathologic features, prognosis, tumor mutation burden (TMB), microsatellite instability (MSI), immune cell infiltration, and immune response genes across 33 datasets from The Cancer Genome Atlas (TCGA). SIRT4 expression was confirmed in BCa tissues, and its functions were examined via proliferation and migration assays. CD8+ T cells were isolated from the peripheral blood of healthy individuals and activated with CD3 and CD28 antibodies and recombinant IL2. Coculture assays involving BCa cells and activated CD8+ T cells, alongside ELISA, were conducted to evaluate the immunological function of SIRT4. RESULTS SIRT4 was positively associated with the immunotherapy response of BCa patients on the basis of IMvigor210 data. Its expression was downregulated in 11 tumor types but upregulated in 3. SIRT4 was significantly correlated with tumor stage in 2 tumor types and showed varying associations with overall survival, progression-free survival, and disease-specific survival. Additionally, SIRT4 was correlated with TMB in 10 tumor types and with MSI in 8. GSEA indicated that SIRT4 was negatively associated with the immune response in 9 tumor types, excluding BCa. It was positively correlated with immune cell infiltration in 2 tumor types and negatively correlated in 6. The TCGA data revealed that SIRT4 was positively associated with activated NK cell infiltration but negatively associated with M1 macrophages, neutrophils, resting NK cells, and activated memory CD4 T cells. Enrichment analyses revealed positive correlations with various chemokines, immunoinhibitors, immunostimulators, lymphocytes, MHC molecules, and MHC receptors, suggesting that SIRT4 may enhance the immune response in BCa. Further experiments confirmed that SIRT4 was downregulated in BCa tissues compared with adjacent normal tissues. Inhibition of SIRT4 promoted BCa cell proliferation and migration, whereas knockdown of SIRT4 impaired the chemotaxis and tumor-killing ability of CD8+ T cells in the BCa tumor microenvironment. CONCLUSIONS In summary, SIRT4 inhibits the progression and immune escape of BCa, indicating its potential as a novel biomarker and immune checkpoint for immunotherapy.
Collapse
Affiliation(s)
- Jiancheng Lv
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Urology, Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Qikai Wu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peng Jiang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yiwei Lin
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Gao Z, Gong Z, Huang H, Ren X, Li Z, Gao P. Transcriptomic analysis of key genes and signaling pathways in sepsis-associated intestinal mucosal barrier damage. Gene 2025; 936:149137. [PMID: 39617276 DOI: 10.1016/j.gene.2024.149137] [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: 05/12/2024] [Revised: 10/19/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024]
Abstract
OBJECTIVES The aim is to analyze differentially expressed genes (DEGs) in mice with sepsis-related intestinal mucosal barrier damage and to explore the diagnostic and protective mechanisms of this condition at the transcriptome level. METHODS Small intestinal tissues from healthy male C57BL/6J mice subjected to Cecal ligation and puncture (CLP) and sham operation were collected. High-throughput sequencing was performed using the paired-end sequencing mode of the Illumina HiSeq platform. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted on the differentially expressed genes (DEGs). A protein-protein interaction (PPI) network was constructed using the STRING database, and hub genes were identified with Cytoscape. These hub genes were then validated using quantitative real-time polymerase chain reaction (RT-qPCR). RESULTS A total of 239 DEGs were identified, with 49 upregulated and 130 downregulated genes. KEGG enrichment analysis showed that these DEGs were primarily involved in cytokine-cytokine receptor interaction, Th1 and Th2 cell differentiation, viral protein interactions with cytokines and their receptors, and the IL-17 signaling pathway. The top 10 hub genes were selected using the cytoHubba plugin. Experimental validation confirmed that the expression levels of TBX21, CSF3, IL-6, CXCR3, and CXCL9 matched the sequencing results. CONCLUSION TBX21, CSF3, IL-6,CXCR3, and CXCL9 may be potential biological markers for the diagnosis and treatment the sepsis-associated intestinal mucosal barrier.
Collapse
Affiliation(s)
- Zhao Gao
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, PR China
| | | | - Hai Huang
- Department of Emergency Medicine, Changzhou Wujin People's Hospital, 2 Yongningbei Road, Changzhou 213000, PR China
| | - Xuemeng Ren
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, PR China
| | - Zhenlu Li
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, PR China.
| | - Peng Gao
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, PR China.
| |
Collapse
|
3
|
Qi L, Wang J, Hou S, Liu S, Zhang Q, Zhu S, Liu S, Zhang S. Unraveling the tumor microenvironment of esophageal squamous cell carcinoma through single-cell sequencing: A comprehensive review. Biochim Biophys Acta Rev Cancer 2025; 1880:189264. [PMID: 39805342 DOI: 10.1016/j.bbcan.2025.189264] [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: 08/27/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly heterogeneous and aggressive malignancy. The progression, invasiveness, and metastatic potential of ESCC are shaped by a multitude of cells within the tumor microenvironment (TME), including tumor cells, immune cells, endothelial cells, as well as fibroblasts and other cell types. Recent advancements in single-cell sequencing technologies have significantly enhanced our comprehension of the diverse landscape of ESCC. Single-cell multi-omics technology, particularly single-cell transcriptome sequencing, have shed light on the expression profiles of individual cells and the molecular characteristics of distinct tumor cell populations. This review summarizes the latest literature on single-cell research in the field of ESCC, aiming to elucidate the heterogeneity of tumor cells, immune cells, and stromal cells at the single-cell level. Furthermore, it explores the impact of cellular interactions within the TME on the progression of ESCC. By compiling a comprehensive overview of single-cell omics research on ESCC, this article aims to enhance our understanding of ESCC diagnosis and treatment by elucidating the intricate interplay within the TME. It explores the cellular composition, spatial arrangement, and functional attributes of the ESCC TME, offering potential therapeutic targets and biomarkers for personalized treatment strategies.
Collapse
Affiliation(s)
- Lingyu Qi
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Jiaxin Wang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Songyuan Hou
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Siying Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Qian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Shengtao Zhu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Si Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| | - Shutian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| |
Collapse
|
4
|
Zhang G, Yu Q, Chen X, Zhao X, Xu Y, Yang X. Unraveling the complexities of immunotherapy for thymic epithelial tumors via bioinformatics and experimental analyses. Comput Biol Med 2025; 185:109488. [PMID: 39631109 DOI: 10.1016/j.compbiomed.2024.109488] [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: 03/12/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Thymic epithelial tumors (TETs) are rare neoplasms typically located in the anterior mediastinum. While immune checkpoint inhibitors (ICIs) show promise for advanced or refractory TETs, their clinical application is hindered by heterogeneous responses across TET subtypes, lack of reliable predictive markers, and the risk of immune-related adverse events (irAEs). METHODS We analyzed TCGA, GEO, and GTEx databases to identify differentially expressed genes (DEGs) among three TET subtypes. Comprehensive enrichment analysis determined gene functions and pathways. CIBERSORT analysis revealed subtype-specific immune infiltration profiles. We assessed immune-related genes using immune/stromal scores, TIDE scores, and immune checkpoint gene correlation analysis. Immunohistochemistry was performed to evaluate FGF17 and PD-L1 protein expression levels and their correlation in TET samples. RESULTS Our findings revealed distinctive molecular and immune infiltration patterns across TET subtypes. Pathway analysis showed upregulation of immune-related pathways in type C. CIBERSORT analysis revealed higher fractions of plasma cells and activated CD4 T cells in type C and increased resting dendritic cells in type A or B3. Furthermore, we identified 1,100 DEGs between responders and non-responders to pembrolizumab. FGF17 emerged as a potential predictive marker for immunotherapy response, showing significantly lower expression in type C and a strong negative correlation with PD-L1 expression (P < 0.001). We identified 115 genes potentially linked to irAEs, with CXCL8, IL17A, and CD40LG among the top hub genes in the protein-protein interaction network. CONCLUSIONS This study provides insights into subtype-specific molecular and immune characteristics of TETs, identifies FGF17 as a potential negative biomarker for immunotherapy response (with lower expression potentially indicating better response), and elucidates mechanisms of irAEs. These findings contribute to the development of targeted immunotherapeutic approaches for managing TETs, particularly in predicting response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Gaowen Zhang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China; Department of Thoracic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266000, China.
| | - Qian Yu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Xiaotong Chen
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, 110001, China.
| | - Xitong Zhao
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Yang Xu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Xueying Yang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| |
Collapse
|
5
|
Chen X, Liang W, Lin C, Lin Y. Identification of a Novel Three-immunogene Diagnostic Signature for Alopecia Areata. Ann Dermatol 2025; 37:22-31. [PMID: 39894670 DOI: 10.5021/ad.24.053] [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: 05/02/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Autoimmune mechanisms have important roles in the pathogenesis of alopecia areata (AA). OBJECTIVE This study aimed to evaluate the exact biological and clinical importance of immunogenes in AA patients using bioinformatic methods. METHODS Five AA scalp gene expression profiles were obtained from the Gene Expression Omnibus database. Differentially-expressed genes (DEGs) between AA and control groups were identified. An immune-related gene diagnostic signature (IRGDS) was established by protein-protein interaction network analysis, least absolute shrinkage and selection operator and logistic regression analysis. RESULTS A total of 102 immune-related DEGs were identified. We developed an IRGDS composed of CD8A, CSF1R and CXCL10 for AA molecular pathological assessment and diagnosis (area under the receiver operating characteristic curve [AUC]=0.962). We also validated the diagnostic value of the IRGDS in an external cohort (AUC=0.955). Patients with high IRGDS scores presented with a higher abundance of immune cell infiltration and expression of genes associated with immune recruitment and immune activation, suggesting adverse biological alterations. CONCLUSION In our study, an IRGDS model with accurately diagnostic capacity for AA was established, and biological alterations were deciphered in AA. The IRGDS may be used as an auxiliary diagnostic marker for AA.
Collapse
Affiliation(s)
- Xiuwen Chen
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Wenzi Liang
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Changmin Lin
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Yike Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
6
|
Ristić D, Bärnthaler T, Gruden E, Kienzl M, Danner L, Herceg K, Sarsembayeva A, Kargl J, Schicho R. GPR55 in the tumor microenvironment of pancreatic cancer controls tumorigenesis. Front Immunol 2025; 15:1513547. [PMID: 39885986 PMCID: PMC11779727 DOI: 10.3389/fimmu.2024.1513547] [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: 10/18/2024] [Accepted: 12/31/2024] [Indexed: 02/01/2025] Open
Abstract
Background The G protein-coupled receptor 55 (GPR55) is part of an expanded endocannabinoid system (ECS), and plays a pro-tumorigenic role in different cancer models, including pancreatic cancer. Next to cancer cells, various cells of the immune tumor microenvironment (TME) express receptors of the ECS that critically determine tumor growth. The role of GPR55 in cancer cells has been widely described, but its role in the immune TME is not well understood. Methods We intended to uncover the role of GPR55 in tumor immunity in a model of pancreatic ductal adenocarcinoma (PDAC). To this end, a KPCY tumor cell line or a GPR55-overexpressing KPCY cell line (KPCY55) from murine PDAC were subcutaneously injected into wildtype (WT) and GPR55 knockout (KO) mice, and immune cell populations were evaluated by flow cytometry. Results Deficiency of GPR55 in the TME led to reduced tumor weight and volume, and altered the immune cell composition of tumors, favoring an anti-tumorigenic environment by increasing the number of CD3+ T cells, particularly CD8+ T cells, and the expression of PDL1 on macrophages. RNA-seq pathway analysis revealed higher T cell activity in KPCY55 tumors of GPR55 KO vs. WT mice. In addition, tumors from GPR55 KO mice displayed increased levels of T cell chemokines Cxcl9 and Cxcl10. Migration of T cells from GPR55 KO mice towards CXCL9 was increased in comparison to T cells from WT mice, suggesting that a CXCR3/CXCL9 axis was involved in T cell influx into tumors of GPR55 KO mice. Notably, anti-PD-1 immunotherapy increased tumor burden in WT mice, while this effect was absent in the GPR55 KO mice. Conclusion Our study indicates that GPR55 in TME cells may drive tumor growth by suppressing T cell functions, such as migration, in a model of PDAC, making it an interesting target for immunotherapies.
Collapse
MESH Headings
- Animals
- Tumor Microenvironment/immunology
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/genetics
- Mice
- Receptors, Cannabinoid/metabolism
- Receptors, Cannabinoid/genetics
- Mice, Knockout
- Cell Line, Tumor
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinogenesis/immunology
- Mice, Inbred C57BL
- Humans
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
Collapse
|
7
|
Li Q, Zhang C, Li L. N6-Methyladenosine (m6A) Reader LRPPRC-Mediated CXCL11 Induces Cell Inflammation to Drive Breast Cancer Cell Malignancy. Crit Rev Immunol 2025; 45:81-92. [PMID: 39612279 DOI: 10.1615/critrevimmunol.2024053166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Breast cancer (BC) is among the most prevalent malignant cancers in women. We examined the function and regulatory mechanism of the N6-methyladenosine (m6A) modification reader leucine-rich pentatricopeptide repeat containing (LRPPRC) in BC inflammation and progression. LRPPRC and C-X-C motif chemokine ligand 11 (CXCL11) levels were measured by quantitative real-time polymerase chain reaction. The regulatory mechanisms of LRPPRC and CXCL11 were determined by RNA binding protein immunoprecipitation, methylated RNA immunoprecipitation, and mRNA stability assays. Moreover, the function of LRPPRC and CXCL11 in BC cells was explored by cell counting kit-8, wound healing, and Transwell assays. Enzyme-linked immunosorbent assay was used to measure proinflammatory cytokine [tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and IL-1β) levels. LRPPRC was expressed at considerably higher levels in BC samples compared with normal tissue samples, and its overexpression predicted a poor prognosis. Reduced LRPPRC decreased BC cell viability, migration, and invasion, whereas overexpression promoted a malignant phenotype. LRPPRC exerted its stimulative effect through CXCL11 m6A modification. CXCL11 upregulation suppressed the antitumor silencing effect of LRPPRC on BC cells. CXCL11 upregulation enhanced the secretion of inflammatory factors by BC cells. LRPPRC aggravates BC inflammation and malignancy by increasing the m6A modification of CXCL11. These findings offer a potential target for BC therapy.
Collapse
Affiliation(s)
- Qing Li
- Department of Oncology, The Affiliated Hospital of Jianghan University, Wuhan 430015, Hubei, China
| | - Changchun Zhang
- Department of Oncology, The Affiliated Hospital of Jianghan University, Wuhan 430015, Hubei, China
| | - Li Li
- The Affiliated Hospital of Jianghan University
| |
Collapse
|
8
|
Fu C, Guo H, Wang M, Ni C, Wu X, Chen X, Hou J, Wang L. Manganese improves CD8 + T cell recruitment via cGAS-STING in hepatocellular carcinoma. Int Immunopharmacol 2024; 143:113591. [PMID: 39549546 DOI: 10.1016/j.intimp.2024.113591] [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: 06/02/2024] [Revised: 10/11/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths worldwide. Chemotherapy using cisplatin, a drug that damages deoxyribonucleic acid (DNA), is not very effective in treating HCC due to its side effects and drug resistance. Manganese (Mn2+), a trace element, has been shown to enhance immune responses, but its ability to improve cisplatin-induced antitumor immunity in HCC remains unclear. The present study found that treatment with Mn2+ in combination with cisplatin promoted cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling activation and C-X-C motif chemokine ligand 10 (CXCL10) production in tumor and dendritic cells. CXCL10 is associated with CD8A levels, and its high expression is linked to better prognosis in patients with HCC. In addition, Mn2+ and cisplatin co-treatment enhanced the recruitment of CD8+ T cells through the CXCL10/CXCR3 axis. Similarly, in an orthotopic transplantation tumor model, STING activation, CD8+ T cell infiltration, and tumor cell killing levels were higher in the combined treatment group. The above findings suggest that utilizing Mn2+ in combination with cisplatin could be a potential treatment option for HCC.
Collapse
Affiliation(s)
- Chunxue Fu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Hanrui Guo
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China; Department of Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Meiling Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China; Department of Pathology, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Caiya Ni
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiangwei Wu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xueling Chen
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jun Hou
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| | - Lianghai Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital/Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| |
Collapse
|
9
|
Buzaglo GBB, Telles GD, Araújo RB, Junior GDS, Ruberti OM, Ferreira MLV, Derchain SFM, Vechin FC, Conceição MS. The Therapeutic Potential of Physical Exercise in Cancer: The Role of Chemokines. Int J Mol Sci 2024; 25:13740. [PMID: 39769501 PMCID: PMC11678861 DOI: 10.3390/ijms252413740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/11/2025] Open
Abstract
The global increase in cancer cases and mortality has been associated with inflammatory processes, in which chemokines play crucial roles. These molecules, a subfamily of cytokines, are essential for the migration, adhesion, interaction, and positioning of immune cells throughout the body. Chemokines primarily originate in response to pathogenic stimuli and inflammatory cytokines. They are expressed by lymphocytes in the bloodstream and are divided into four classes (CC, CXC, XC, and CX3C), playing multifaceted roles in the tumor environment (TME). In the TME, chemokines regulate immune behavior by recruiting cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which promote tumor survival. Additionally, they directly influence tumor behavior, promoting pathological angiogenesis, invasion, and metastasis. On the other hand, chemokines can also induce antitumor responses by mobilizing CD8+ T cells and natural killer (NK) cells to the tumor, reducing pro-inflammatory chemokines and enhancing essential antitumor responses. Given the complex interaction between chemokines, the immune system, angiogenic factors, and metastasis, it becomes evident how important it is to target these pathways in therapeutic interventions to counteract cancer progression. In this context, physical exercise emerges as a promising strategy due to its role modulating the expression of anti-inflammatory chemokines and enhancing the antitumor response. Aerobic and resistance exercises have been associated with a beneficial inflammatory profile in cancer, increased infiltration of CD8+ T cells in the TME, and improvement of intratumoral vasculature. This creates an environment less favorable to tumor growth and supports the circulation of antitumor immune cells and chemokines. Therefore, understanding the impact of exercise on the expression of chemokines can provide valuable insights for therapeutic interventions in cancer treatment and prevention.
Collapse
Affiliation(s)
- Glenda B. B. Buzaglo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Guilherme D. Telles
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Rafaela B. Araújo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Gilmar D. S. Junior
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Olivia M. Ruberti
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Marina L. V. Ferreira
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Sophie F. M. Derchain
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-881, Brazil;
| | - Felipe C. Vechin
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Miguel S. Conceição
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| |
Collapse
|
10
|
Szymczak M, Heidecke H, Żabińska M, Janek Ł, Wronowicz J, Kujawa K, Bukowiec-Marek K, Gołębiowski T, Skalec K, Schulze-Forster K, Konieczny A, Banasik M. The Influence of Anti-ETAR and Anti-CXCR3 Antibody Levels on the Course of Specific Glomerulonephritis Types. J Clin Med 2024; 13:7752. [PMID: 39768675 PMCID: PMC11679591 DOI: 10.3390/jcm13247752] [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: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Anti-ETAR (endothelin A receptor) antibodies and anti-CXCR3 (C-X-C motif chemokine receptor 3) antibodies are types of non-HLA (human leukocyte antigens) antibodies that could have some influence on the course of glomerulonephritis. The authors aimed to study the influence of these antibodies' levels on the course of specific glomerulonephritis types. Methods: We evaluated the anti-ETAR and anti-CXCR3 antibody levels in the serum of patients with membranous nephropathy (n = 18), focal and segmental glomerulosclerosis (FSGS) (n = 25), systemic lupus erythematosus (n = 17), IgA nephropathy (n = 14), mesangiocapillary glomerulonephritis (n = 6), anti-neutrophil cytoplasmic antibodies (c-ANCA) vasculitis (n = 40), and perinuclear anti-neutrophil cytoplasmic antibodies (p-ANCA) vasculitis (n = 16), and we compared their levels with the control group (n = 22). Next, we observed the patients' clinical parameters (serum creatinine, albumin, total protein) for 2 years and checked the correlation of the clinical course markers with basic receptor antibody level. Results: Our results indicate lower anti-ETAR antibody levels in patients with FSGS and IgA nephropathy compared to the control group. Both types of antibodies correlated with creatinine levels after 2 years of observation in IgA nephropathy. Both types of antibodies seemed to negatively influence the total protein and albumin levels in systemic lupus erythematosus. Conclusions: This prospective observation showed that anti-ETAR and anti-CXCR 3 antibody levels are connected with the course of IgA nephropathy and lupus nephritis.
Collapse
Affiliation(s)
- Maciej Szymczak
- Clinical Department of Nephrology, Transplantation Medicine and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.B.-M.); (T.G.); (A.K.); (M.B.)
| | - Harald Heidecke
- CellTrendGmbh, Im Biotechnologiepark 3 TGZ II, 14943 Luckenwalde, Germany; (H.H.); (K.S.-F.)
| | - Marcelina Żabińska
- Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Faculty of Medicine, Wrocław University of Science and Technology, 58-376 Wroclaw, Poland;
| | - Łucja Janek
- Statistical Analysis Center, Wroclaw Medical University, 50-368 Wroclaw, Poland; (Ł.J.); (J.W.); (K.K.)
| | - Jakub Wronowicz
- Statistical Analysis Center, Wroclaw Medical University, 50-368 Wroclaw, Poland; (Ł.J.); (J.W.); (K.K.)
| | - Krzysztof Kujawa
- Statistical Analysis Center, Wroclaw Medical University, 50-368 Wroclaw, Poland; (Ł.J.); (J.W.); (K.K.)
| | - Karolina Bukowiec-Marek
- Clinical Department of Nephrology, Transplantation Medicine and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.B.-M.); (T.G.); (A.K.); (M.B.)
| | - Tomasz Gołębiowski
- Clinical Department of Nephrology, Transplantation Medicine and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.B.-M.); (T.G.); (A.K.); (M.B.)
| | - Karolina Skalec
- Clinical Department of Nephrology, Transplantation Medicine and Internal Diseases, University Clinical Hospital, 50-556 Wroclaw, Poland;
| | - Kai Schulze-Forster
- CellTrendGmbh, Im Biotechnologiepark 3 TGZ II, 14943 Luckenwalde, Germany; (H.H.); (K.S.-F.)
| | - Andrzej Konieczny
- Clinical Department of Nephrology, Transplantation Medicine and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.B.-M.); (T.G.); (A.K.); (M.B.)
| | - Mirosław Banasik
- Clinical Department of Nephrology, Transplantation Medicine and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.B.-M.); (T.G.); (A.K.); (M.B.)
| |
Collapse
|
11
|
Schirm S, Nouailles G, Kirsten H, Trimpert J, Wyler E, Teixeira Alves LG, Landthaler M, Ahnert P, Suttorp N, Witzenrath M, Scholz M. A biomathematical model of SARS-CoV-2 in Syrian hamsters. Sci Rep 2024; 14:30541. [PMID: 39695178 DOI: 10.1038/s41598-024-80498-9] [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: 04/12/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
When infected with SARS-CoV-2, Syrian hamsters (Mesocricetus auratus) develop moderate disease severity presenting key features of human COVID-19. We here develop a biomathematical model of the disease course by translating known biological mechanisms of virus-host interactions and immune responses into ordinary differential equations. We explicitly describe the dynamics of virus population, affected alveolar epithelial cells, and involved relevant immune cells comprising for example CD4+ T cells, CD8+ T cells, macrophages, natural killer cells and B cells. We also describe the humoral response dynamics of neutralising antibodies and major regulatory cytokines including CCL8 and CXCL10. The model is developed and parametrized based on experimental data collected at days 2, 3, 5, and 14 post infection. Pulmonary cell composition and their transcriptional profiles were obtained by lung single-cell RNA (scRNA) sequencing analysis. Parametrization of the model resulted in a good agreement of model and data. The model can be used to predict, for example, the time course of the virus population, immune cell dynamics, antibody production and regeneration of alveolar cells for different therapy scenarios or after multiple-infection events. We aim to translate this model to the human situation in the future.
Collapse
Affiliation(s)
- Sibylle Schirm
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, 04107, Leipzig, Germany.
| | - Geraldine Nouailles
- Department of Infectious Diseases and Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, 04107, Leipzig, Germany
| | - Jakob Trimpert
- Institute of Virology, Freie Universität Berlin, 14163, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Luiz Gustavo Teixeira Alves
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
- Institute for Biology, Humboldt-Universität zu Berlin, 10099, Berlin, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, 04107, Leipzig, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, 04107, Leipzig, Germany
| |
Collapse
|
12
|
Daoudlarian D, Segot A, Latifyan S, Bartolini R, Joo V, Mederos N, Bouchaab H, Demicheli R, Abdelhamid K, Ferahta N, Doms J, Stalder G, Noto A, Mencarelli L, Mosimann V, Berthold D, Stravodimou A, Sartori C, Shabafrouz K, Thompson JA, Wang Y, Peters S, Pantaleo G, Obeid M. Tocilizumab and immune signatures for targeted management of cytokine release syndrome in immune checkpoint therapy. Ann Oncol 2024:S0923-7534(24)04979-2. [PMID: 39701282 DOI: 10.1016/j.annonc.2024.12.004] [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: 10/09/2024] [Revised: 11/26/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND This study aimed to identify specific biomarkers in oncology patients experiencing immune-related cytokine release syndrome (irCRS)-like symptoms during immune checkpoint inhibitor (ICI) therapy, including severe cases like hemophagocytic lymphohistiocytosis (irHLH), and to distinguish these from sepsis. A secondary objective was to retrospectively analyze the efficacy of tocilizumab (TCZ) in treating corticosteroid (CS)-refractory high-grade irCRS. PATIENTS AND METHODS A cohort of 35 patients presenting with irCRS-like symptoms was studied, including 9 with irHLH-like manifestations and 8 with sepsis. Immune profiling was carried out using 48 mass cytometry markers, along with an analysis of 45 serum biomarkers, including 27 cytokines and 18 additional markers from the HScore. Twelve patients with high-grade irCRS refractory to CS were treated with TCZ. RESULTS Twenty-four biomarkers significantly distinguished between irHLH and grade 3 irCRS (P = 0.0027-0.0455). Hepatocyte growth factor (HGF) and ferritin had superior predictive values compared with the traditional HScore, both with a positive predictive value (PPV) and negative predictive value (NPV) of 100%. CXCL9 differentiated irHLH from grade 3 irCRS and predicted the need for TCZ treatment intensification (PPV = 90%, NPV = 100%). Additional biomarkers, including leukocyte count, neutrophils, ferritin, interleukin (IL)-6, IL-7, epidermal growth factor, fibrinogen, and granulocyte-macrophage colony-stimulating factor (GM-CSF), discriminated sepsis from high-grade irCRS (PPV = 75%-80%, NPV = 100%). Elevated frequencies of CXCR5+ or CCR4+ CD8 memory cells, CD38+ intermediate monocytes, and CD62L+ neutrophils were observed in high-grade irCRS compared with sepsis. All 12 patients with high-grade irCRS refractory to CS treated with TCZ experienced complete resolution. CONCLUSIONS This study highlights the importance of specific immunologic biomarkers in determining irCRS severity, predicting outcomes, and distinguishing between irHLH, irCRS, and sepsis. It also demonstrates the efficacy of TCZ in managing high-grade irCRS, underscoring the need for personalized therapeutic strategies based on these biomarkers.
Collapse
Affiliation(s)
- D Daoudlarian
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - A Segot
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Service and Central Laboratory of Hematology, University of Lausanne, Lausanne, Switzerland
| | - S Latifyan
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - R Bartolini
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - V Joo
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - N Mederos
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - H Bouchaab
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - R Demicheli
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - K Abdelhamid
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - N Ferahta
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - J Doms
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - G Stalder
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Service and Central Laboratory of Hematology, University of Lausanne, Lausanne, Switzerland; Service of Hematology, Institut Central des Hôpitaux, Hôpital du Valais, Sion, Switzerland
| | - A Noto
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - L Mencarelli
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - V Mosimann
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - D Berthold
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - A Stravodimou
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - C Sartori
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Internal Medicine Service, University of Lausanne, Lausanne, Switzerland
| | - K Shabafrouz
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - J A Thompson
- Department of Medicine, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, USA
| | - Y Wang
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Peters
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - G Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - M Obeid
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
13
|
Tsang CH, Kozielewicz P. Exploring G Protein-Coupled Receptors in Hematological Cancers. ACS Pharmacol Transl Sci 2024; 7:4000-4009. [PMID: 39698279 PMCID: PMC11651347 DOI: 10.1021/acsptsci.4c00473] [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: 08/07/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 12/20/2024]
Abstract
Hematological cancers, such as lymphomas and leukemias, pose significant challenges in oncology, necessitating a deeper understanding of their molecular landscape to enhance therapeutic strategies. This article critically examines and discusses recent research on the roles of G protein-coupled receptors (GPCRs) in myeloma, lymphomas, and leukemias with a particular focus on pediatric acute lymphoblastic (lymphocytic) leukemia (ALL). By utilizing RNA sequencing (RNA-seq), we analyzed GPCR expression patterns in pediatric ALL samples (aged 3-12 years old), with a further focus on Class A orphan GPCRs. Our analysis revealed distinct GPCR expression profiles in pediatric ALL, identifying several candidates with aberrant upregulated expression compared with healthy counterparts. Among these GPCRs, GPR85, GPR65, and GPR183 have varying numbers of studies in the field of hematological cancers and pediatric ALL. Furthermore, we explored missense mutations of pediatric ALL in relation to the RNA gene expression findings, providing insights into the genetic underpinnings of this disease. By integrating both RNA-seq and missense mutation data, this article aims to provide an insightful and broader perspective on the potential correlations between specific GPCR and their roles in pediatric ALL.
Collapse
Affiliation(s)
- Choi Har Tsang
- Molecular Pharmacology of GPCRs, Department Physiology & Pharmacology,
Karolinska Institutet, Biomedicum, 171 65 Stockholm,
Sweden
| | - Pawel Kozielewicz
- Molecular Pharmacology of GPCRs, Department Physiology & Pharmacology,
Karolinska Institutet, Biomedicum, 171 65 Stockholm,
Sweden
| |
Collapse
|
14
|
Cuiffo B, Maxwell M, Yan D, Guemiri R, Boone A, Bellet D, Rivest B, Cardia J, Robert C, Fricker SP. Self-delivering RNAi immunotherapeutic PH-762 silences PD-1 to generate local and abscopal antitumor efficacy. Front Immunol 2024; 15:1501679. [PMID: 39697325 PMCID: PMC11652358 DOI: 10.3389/fimmu.2024.1501679] [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: 09/25/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024] Open
Abstract
Objective Immunotherapeutic inhibition of PD-1 by systemically administered monoclonal antibodies is widely used in cancer treatment, but it may cause severe immune-related adverse events (irSAEs). Neoadjuvant PD-1 inhibition before surgery has shown promise in reducing recurrence by stimulating durable antitumor immunity. Local intratumoral (IT) immunotherapy is a potential strategy to minimize irSAEs, but antibodies have limited tumor penetration, making them less suitable for this approach. Therapeutic self-delivering RNAi (INTASYL) is an emerging modality well-suited for neoadjuvant immunotherapy. This study presents preclinical proof-of-concept for PH-762, an INTASYL designed to silence PD-1, currently in clinical development for advanced cutaneous malignancies (ClinicalTrials.gov#NCT06014086). Methods and analysis PH-762 pharmacology was characterized in vitro, and in vivo antitumor efficacy was evaluated using a murine analogue (mPH-762) in syngeneic tumor models with varying PD-1 responsiveness. Bilateral Hepa1-6 models assessed abscopal effects of local treatment. Ex vivo analyses explored mechanisms of direct and abscopal efficacy. Results PH-762 was rapidly internalized by human T cells, silencing PD-1 mRNA and decreasing PD-1 surface protein, enhancing TCR-stimulated IFN-γ and CXCL10 secretion. In vivo, IT mPH-762 provided robust antitumor efficacy, local and lymphatic biodistribution, and was well tolerated. Ex vivo analyses revealed that IT mPH-762 depleted PD-1 protein, promoted leukocyte and T cell infiltration, and correlated with tumor control. IT mPH-762 also demonstrated efficacy against untreated distal tumors (abscopal effect) by priming systemic antitumor immunity. Conclusion These data support PH-762 as a promising candidate for neoadjuvant immunotherapy in clinical studies.
Collapse
Affiliation(s)
| | | | - Dingxue Yan
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Ramdane Guemiri
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | - Andrew Boone
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Deborah Bellet
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | | - James Cardia
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Caroline Robert
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | |
Collapse
|
15
|
Taylor MH, Naing A, Powderly J, Woodard P, Chung L, Lin WH, Tian H, Siemers N, Xiang H, Deng R, Hong K, Valencia D, Huang T, Zhu Y, Liao XC, Schebye XM, Patel MR. Phase I dose escalation study of IO-108, an anti-LILRB2 antibody, in patients with advanced solid tumors. J Immunother Cancer 2024; 12:e010006. [PMID: 39567210 PMCID: PMC11580248 DOI: 10.1136/jitc-2024-010006] [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] [Accepted: 09/16/2024] [Indexed: 11/22/2024] Open
Abstract
PURPOSE In this first-in-human dose escalation study, the safety and efficacy of IO-108, a fully human monoclonal antibody targeting leukocyte immunoglobulin-like receptor B2 (LILRB2), was investigated in patients with advanced solid tumors as monotherapy and in combination with pembrolizumab, an anti-programmed cell death protein 1 (PD-1) antibody. METHODS The study included patients with histologically or cytologically confirmed advanced and relapsed solid tumors, with measurable disease by Response Evaluation Criteria In Solid Tumors (RECIST) V.1.1. Patients were treated with escalating doses of IO-108 every 3 weeks (Q3W) as monotherapy and in combination with pembrolizumab. Safety and tolerability were the primary objectives. Secondary and exploratory objectives included: pharmacokinetics, clinical efficacy, immunogenicity and biomarkers. RESULTS Of 25 patients enrolled, 12 were treated with IO-108 monotherapy and 13 received combination therapy. IO-108 was well-tolerated up to the maximally administered dose of 1,800 mg every 3 weeks (Q3W) as monotherapy and in combination with pembrolizumab. No dose-limiting toxicity was observed, and a maximum tolerated dose was not reached. Treatment-related adverse events (TRAEs) occurred in 6 (50.0%) patients treated with IO-108 monotherapy and 6 (46.2%) patients treated with IO-108+pembrolizumab. All TRAEs were mild or moderate (Grade 1 or 2), and no TRAEs led to treatment discontinuation or death. IO-108 exhibited a dose-proportional increase in exposure. Full receptor occupancy (RO) in peripheral blood was achieved at doses ≥600 mg. The overall response rate was 9% (1/11) in the monotherapy and 23% (3/13) in the combination therapy. A patient with treatment-refractory Merkel cell carcinoma treated with IO-108 monotherapy achieved a durable complete response (CR) for more than 2 years. Pharmacodynamic gene expression changes reflecting increased tumor infiltration of T cells were associated with clinical benefits in both monotherapy and combination therapy. Additionally, baseline tumor inflammation gene signature (TIS) scores correlated with clinical benefit. CONCLUSION IO-108 is well tolerated and has led to objective response as monotherapy and in combination with pembrolizumab. The complete response and the pharmacodynamic changes in the monotherapy cohort demonstrate single agent activity of IO-108 and provide proof of concept that targeting myeloid-suppressive pathways through LILRB2 inhibition may potentiate the clinical efficacy of anti-PD-1 immune checkpoint inhibitors. TRIAL REGISTRATION NUMBER NCT05054348.
Collapse
MESH Headings
- Humans
- Male
- Female
- Neoplasms/drug therapy
- Neoplasms/immunology
- Middle Aged
- Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Receptors, Immunologic/antagonists & inhibitors
- Adult
- Membrane Glycoproteins/antagonists & inhibitors
- Aged, 80 and over
Collapse
Affiliation(s)
- Matthew H Taylor
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Aung Naing
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John Powderly
- Carolina BioOncology Institute, Huntersville, North Carolina, USA
| | - Paul Woodard
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Luke Chung
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Wen Hong Lin
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Hongyu Tian
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Nathan Siemers
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Hong Xiang
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Rong Deng
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Kyu Hong
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Donna Valencia
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Tao Huang
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | - Ying Zhu
- Immune-Onc Therapeutics, Inc, Palo Alto, California, USA
| | | | | | - Manish R Patel
- Florida Cancer Specialists/ Sarah Cannon Research Institute, Sarasota, Florida, USA
| |
Collapse
|
16
|
Leveque E, Battut L, Petitfils C, Valitutti S, Cenac N, Dietrich G, Espinosa E. Alternative activation of mast cells by CD4+ T helper cells. J Leukoc Biol 2024; 116:1127-1141. [PMID: 38916515 DOI: 10.1093/jleuko/qiae139] [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: 12/30/2023] [Revised: 05/16/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024] Open
Abstract
Effector CD4+ T (Teff) lymphocytes infiltrate sites of inflammation and orchestrate the immune response by instructing local leukocytes. Mast cells (MCs) are tissue sentinel cells strategically located near blood vessels and T cell-rich areas. MC/Teff cell interactions shape Teff cell responses, but in turn, Teff cell action on MCs is still poorly understood. Here, we analyzed the human MC/Teff cell interplay through both the application of RNA sequencing and functional assays. We showed that activated Teff cells induce a specific transcriptomic program in MCs including production of both inflammatory cytokines and chemokines, prostaglandin, and a FcεRI-dependent degranulation facilitation, thereby driving them toward an inflammatory phenotype. Moreover, Teff cells induce in MCs the capacity to interact with CD4+ T cells through a wide range of dedicated soluble and membrane ligands and to play the role of antigen-presenting cells.
Collapse
Affiliation(s)
- Edouard Leveque
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1037, Centre de Recherche en Cancérologie de Toulouse, 2 Avenue H. Curien, F-31037, France
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
| | - Louise Battut
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Camille Petitfils
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Salvatore Valitutti
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1037, Centre de Recherche en Cancérologie de Toulouse, 2 Avenue H. Curien, F-31037, France
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Centre Hospitalier Universitaire de Toulouse, 1 avenue Irène Joliot-Curie, Toulouse F-31059, France
| | - Nicolas Cenac
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Gilles Dietrich
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| | - Eric Espinosa
- Université Toulouse III - Paul Sabatier, 118 route de Narbone, Toulouse F-31062, France
- Institut National de la Santé et de la Recherche Médicale, U1220, Institut de Recherche en Santé Digestive, Institut National de la Recherche Agronomique, Institut National Polytechnique de Toulouse-École Nationale Vétérinaire de Toulouse, CHU Purpan place du Dr Baylac CS 60039, Toulouse F-31024, France
| |
Collapse
|
17
|
Shen Y, Shi R, Lu S, Wang Y, Zhou Z, Wu C, You Q, Fan H, Wu J. Role of Peptidyl Arginine Deiminase 4-Dependent Macrophage Extracellular Trap Formation in Type 1 Diabetes Pathogenesis. Diabetes 2024; 73:1862-1874. [PMID: 39137121 DOI: 10.2337/db23-1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
Excessive formation of macrophage extracellular trap (MET) has been implicated in several autoimmune disease pathogeneses; however, its impact on type 1 diabetes (T1D) and related mechanisms remains enigmatic. We demonstrated the pivotal role of peptidyl arginine deiminase 4 (PAD4) in driving profuse MET formation and macrophage M1 polarization in intestinal inflammation in NOD mice. Genetic knockout of PAD4 or adoptive transfer of METs altered the proportion of proinflammatory T cells in the intestine, subsequently influencing their migration to the pancreas. Combining RNA sequencing and CUT&Tag analysis, we found activated PAD4 transcriptionally regulated CXCL10 expression. This study comprehensively investigated how excessive PAD4-mediated MET formation in the colon increases the aggravation of intestinal inflammation and proinflammatory T-cell migration and finally is involved in T1D progression, suggesting that inhibition of MET formation may be a potential therapeutic target in T1D. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yiming Shen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ruiya Shi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - ShiPing Lu
- Center for Translational Research in Infection and Inflammation, Tulane University, New Orleans, LA
| | - Yan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ziqi Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chenhua Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qi You
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hongye Fan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jie Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
18
|
Zhou K, Lu J. Progress in cytokine research for ARDS: A comprehensive review. Open Med (Wars) 2024; 19:20241076. [PMID: 39479463 PMCID: PMC11524396 DOI: 10.1515/med-2024-1076] [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: 05/07/2024] [Revised: 09/16/2024] [Accepted: 10/06/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction Acute respiratory distress syndrome (ARDS) is a critical form of acute respiratory failure characterized by diffuse alveolar damage, refractory hypoxemia, and non-cardiogenic pulmonary edema, resulting in high mortality. Dysregulated inflammation, driven by cytokines, is central to ARDS pathogenesis, progression, and prognosis. Objective This review synthesizes current knowledge on the role of cytokines in ARDS and evaluates their potential as therapeutic targets, offering new insights for clinical management. Methods A comprehensive analysis of recent studies was conducted to explore the roles of pro-inflammatory cytokines (e.g., IL-1β, IL-6, IL-8) and anti-inflammatory cytokines (e.g., IL-10, IL-22) in ARDS pathogenesis and to assess current and emerging therapies targeting these cytokines. Results Pro-inflammatory cytokines are crucial in initiating inflammatory responses and lung injury in early ARDS, while anti-inflammatory cytokines help regulate and resolve inflammation. Targeted therapies, such as IL-1 and IL-6 inhibitors, show potential in managing ARDS, particularly in COVID-19, but their clinical efficacy is still debated. Combination therapy strategies may enhance outcomes, but further large-scale, multicenter randomized controlled trials are required to establish their safety and efficacy. Conclusion Understanding cytokine regulation in ARDS could lead to innovative therapeutic approaches. Future research should focus on cytokine roles across ARDS subtypes and stages and develop biomarker-driven, individualized treatments.
Collapse
Affiliation(s)
- Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxuedong Road, Nanning, Guangxi 530007, China
| |
Collapse
|
19
|
Morrocchi E, Pascucci GR, Cotugno N, Pighi C, Dominguez-Rodriguez S, Petrara MR, Tagarro A, Kuhn L, Cotton MF, Otwombe K, Lain MG, Vaz P, Barnabas SL, Spyer MJ, Lopez E, Fernández-Luis S, Nhampossa T, Maiga AI, Dolo O, De Rossi A, Rojo P, Giaquinto C, Lichterfeld M, Violari A, Smit T, Behuhuma O, Klein N, De Armas L, Pahwa S, Rossi P, Palma P. Early inflammation as a footprint of increased mortality risk in infants living with HIV from three African countries. Sci Rep 2024; 14:25792. [PMID: 39468166 PMCID: PMC11519903 DOI: 10.1038/s41598-024-74066-4] [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: 05/21/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
In this work our aim was to identify early biomarkers in plasma samples associated with mortality in children with perinatal HIV treated early in life, to potentially inform early intervention targeting this vulnerable group. 20/215 children (9.3%) with perinatal HIV, enrolled within 3 months of age died prematurely within the first year of the study, despite early ART initiation. Using a propensity score, we selected 40 alive study participants having similar clinical and virological records compared to the deceased group. 13 HIV unexposed (HU) healthy children were additionally used as controls. Baseline plasma samples were analyzed using a targeted proteomic approach, and to assess pathogen-associated and damage-associated molecular patterns (PAMPs, DAMPs) levels. Data from deceased participants were compared to both control groups, with multivariate logistic regression models used to evaluate the association between mortality and plasma proteins. We developed a machine learning model to predict mortality risk, finding that IL-6 and CXCL11 not only were higher in deceased children than Matched-children with HIV (p < 0.001 and p = 0.0034) but also predictive of mortality (accuracy of 77%); levels of PAMPs were higher in deceased children (p = 0.0016). Thus, measuring early inflammatory biomarkers, particularly IL-6, could help mortality risk prediction and potentially guide targeted interventions.
Collapse
Affiliation(s)
- Elena Morrocchi
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Giuseppe R Pascucci
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Nicola Cotugno
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine, Molecular Medicine, and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
| | - Chiara Pighi
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sara Dominguez-Rodriguez
- Instituto de Investigación Sanitaria Hospital 12 Octubre, Madrid, Spain
- Pediatrics Research Group, Universidad Europea de Madrid, Madrid, Spain
| | | | - Alfredo Tagarro
- Instituto de Investigación Sanitaria Hospital 12 Octubre, Madrid, Spain
- Pediatrics Department, Hospital Universitario Infanta Sofía, Infanta Sofia University Hospital and Henares University Hospital Foundation for Biomedical Research and Innovation (FIIB HUIS HHEN), San Sebastián de los Reyes, Madrid, Spain
- Pediatrics Research Group, Universidad Europea de Madrid, Madrid, Spain
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Mark F Cotton
- Family Centre for Research With Ubuntu, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Kennedy Otwombe
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maria G Lain
- Fundação Ariel Glaser contra o SIDA Pediátrico, Maputo, Mozambique
| | - Paula Vaz
- Fundação Ariel Glaser contra o SIDA Pediátrico, Maputo, Mozambique
| | - Shaun L Barnabas
- Family Centre for Research With Ubuntu, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Moira J Spyer
- University College London, Great Ormond Street Institute for Child Health, Infection, Immunity and Inflammation, London, UK
| | - Elisa Lopez
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Sheila Fernández-Luis
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Tacilta Nhampossa
- Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Almoustapha I Maiga
- University Clinical Research Center (UCRC), University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Oumar Dolo
- University Clinical Research Center (UCRC), University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Anita De Rossi
- Section of Oncology and Immunology, DiSCOG, University of Padova, Padova, Italy
- Istituto Oncologico Veneto (IOV-IRCCS), Padua, Italy
| | - Pablo Rojo
- Universidad Computense de Madrid, Istituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Carlo Giaquinto
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128, Padua, Italy
- Penta - Child Health Research, Corso Stati Uniti, 4 - 35127, Padua, Italy
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Avy Violari
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Theresa Smit
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Osee Behuhuma
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Nigel Klein
- Africa Health Research Institute (AHRI), Durban, South Africa
- Great Ormond Street Institute for Child Health (GOS ICH), University College London (UCL), London, UK
| | - Lesley De Armas
- Department of Microbiology and Immunology, Miami Center for AIDS Research, Miller School of Medicine, University of Miami, Miami, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, Miami Center for AIDS Research, Miller School of Medicine, University of Miami, Miami, USA
| | - Paolo Rossi
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine, Molecular Medicine, and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
- Department of Systems Medicine, Molecular Medicine, and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
20
|
Mastrogiovanni M, Donnadieu E, Pathak R, Di Bartolo V. Subverting Attachment to Prevent Attacking: Alteration of Effector Immune Cell Migration and Adhesion as a Key Mechanism of Tumor Immune Evasion. BIOLOGY 2024; 13:860. [PMID: 39596815 PMCID: PMC11591779 DOI: 10.3390/biology13110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
Cell adhesion regulates specific migratory patterns, location, communication with other cells, physical interactions with the extracellular matrix, and the establishment of effector programs. Proper immune control of cancer strongly depends on all these events occurring in a highly accurate spatiotemporal sequence. In response to cancer-associated inflammatory signals, effector immune cells navigating the bloodstream shift from their patrolling exploratory migration mode to establish adhesive interactions with vascular endothelial cells. This interaction enables them to extravasate through the blood vessel walls and access the cancer site. Further adhesive interactions within the tumor microenvironment (TME) are crucial for coordinating their distribution in situ and for mounting an effective anti-tumor immune response. In this review, we examine how alterations of adhesion cues in the tumor context favor tumor escape by affecting effector immune cell infiltration and trafficking within the TME. We discuss the mechanisms by which tumors directly modulate immune cell adhesion and migration patterns to affect anti-tumor immunity and favor tumor evasion. We also explore indirect immune escape mechanisms that involve modifications of TME characteristics, such as vascularization, immunogenicity, and structural topography. Finally, we highlight the significance of these aspects in designing more effective drug treatments and cellular immunotherapies.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emmanuel Donnadieu
- Equipe Labellisée Ligue Contre le Cancer, CNRS, INSERM, Institut Cochin, Université Paris Cité, F-75014 Paris, France;
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Vincenzo Di Bartolo
- Immunoregulation Unit, Institut Pasteur, Université Paris Cité, F-75015 Paris, France;
| |
Collapse
|
21
|
Nielsen AJ, Albert GK, Sanchez A, Chen J, Liu J, Davalos AS, Geng D, Bradeen X, Hintzsche JD, Robinson W, McCarter M, Amato C, Tobin R, Couts K, Wilky BA, Davila E. DNA-PK inhibition enhances neoantigen diversity and increases T cell responses to immunoresistant tumors. J Clin Invest 2024; 134:e180278. [PMID: 39436696 PMCID: PMC11645140 DOI: 10.1172/jci180278] [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: 02/13/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Effective antitumor T cell activity relies on the expression and MHC presentation of tumor neoantigens. Tumor cells can evade T cell detection by silencing the transcription of antigens or by altering MHC machinery, resulting in inadequate neoantigen-specific T cell activation. We identified the DNA-protein kinase inhibitor (DNA-PKi) NU7441 as a promising immunomodulator that reduced immunosuppressive proteins, while increasing MHC-I expression in a panel of human melanoma cell lines. In tumor-bearing mice, combination therapy using NU7441 and the immune adjuvants stimulator of IFN genes (STING) ligand and the CD40 agonist NU-SL40 substantially increased and diversified the neoantigen landscape, antigen-presenting machinery, and, consequently, substantially increased both the number and repertoire of neoantigen-reactive, tumor-infiltrating lymphocytes (TILs). DNA-PK inhibition or KO promoted transcription and protein expression of various neoantigens in human and mouse melanomas and induced sensitivity to immune checkpoint blockade (ICB) in resistant tumors. In patients, protein kinase, DNA-activated catalytic subunit (PRKDC) transcript levels were inversely correlated with MHC-I expression and CD8+ TILs but positively correlated with increased neoantigen loads and improved responses to ICB. These studies suggest that inhibition of DNA-PK activity can restore tumor immunogenicity by increasing neoantigen expression and presentation and broadening the neoantigen-reactive T cell population.
Collapse
Affiliation(s)
- Allison J. Nielsen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Gabriella K. Albert
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Amelia Sanchez
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jiangli Chen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
| | - Jing Liu
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Andres S. Davalos
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Degui Geng
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
| | - Xander Bradeen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - William Robinson
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Martin McCarter
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Carol Amato
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Richard Tobin
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kasey Couts
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Breelyn A. Wilky
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Eduardo Davila
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| |
Collapse
|
22
|
Anastasiadou DP, Couturier N, Goel S, Argyris DG, Vodopyanov S, Rivera-Sanchez L, Gonzalez E, Kreger J, Griffen A, Kazakov A, Burt J, Recoder N, Duran CL, Harney AS, Quesnel A, Filippou PS, Lenis VP, Shukla S, Entenberg D, Zintiridou A, Chen X, Eddy RJ, Oktay MH, Condeelis JS, Karagiannis NS, Briceno A, Guzik H, Alon R, DesMarais V, Ioannou G, Gnjatic S, Raynolds DM, Macedo R, Reshef R, Gil-Henn H, MacLean AL, Torres ER, LaFave LM, Lauvau G, Karagiannis GS. Intratumoral CXCL12 Gradients Contextualize Tumor Cell Invasion, Migration and Immune Suppression in Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618571. [PMID: 39464015 PMCID: PMC11507869 DOI: 10.1101/2024.10.15.618571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Although the CXCL12/CXCR4 pathway has been prior investigated for its prometastatic and immuno- suppressive roles in the tumor microenvironment, evidence on the spatiotemporal regulation of these hallmarks has been lacking. Here, we demonstrate that CXCL12 forms a gradient specifically around cancer cell intravasation doorways, also known as Tumor Microenvironment of Metastasis (TMEM) doorways, thus facilitating the chemotactic translocation of prometastatic tumor cells expressing CXCR4 toward the perivascular TMEM doorways for subsequent entry into peripheral circulation. Fur- thermore, we demonstrate that the CXCL12-rich micro-environment around TMEM doorways may cre- ate immunosuppressive niches, whereby CD8 + T cells, despite being attracted to these regions, often exhibit reduced effector functions, limiting their efficacy. While the CXCL12/CXCR4 pathway can mini- mally influence the overall composition of immune cell populations, it biases the distribution of CD8 + T cells away from TMEM doorways, justifying its prior-established role as immunosuppressive factor for CD8 + T cells. Our research suggests that the complex interactions between CXCL12 and the various tumor and immune cell types contributes not only to the completion of the initial steps of the metastatic cascade, but also offers an immunological "sanctuary" to prometastatic tumor cells homed around TMEM doorways. Overall, our study enhances our current understanding on the mechanisms, via which CXCL12 orchestrates tumor cell behavior and immune dynamics, potentially guiding future thera- peutic strategies to combat breast cancer metastasis and improve anti-tumor immunity.
Collapse
|
23
|
Xu J, Yu Y, Zhang Y, Dai H, Yang Q, Wang B, Ma Q, Chen Y, Xu F, Shi X, Liu Z, Wang C. Oral administration of garlic-derived nanoparticles improves cancer immunotherapy by inducing intestinal IFNγ-producing γδ T cells. NATURE NANOTECHNOLOGY 2024; 19:1569-1578. [PMID: 39054386 DOI: 10.1038/s41565-024-01722-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/17/2024] [Indexed: 07/27/2024]
Abstract
Gamma-delta (γδ) T cell-based cancer immunotherapies represent a promising avenue for cancer treatment. However, their development is challenged by the limited expansion and differentiation of the cells ex vivo. Here we induced the endogenous expansion and activation of γδ T cells through oral administration of garlic-derived nanoparticles (GNPs). We found that GNPs could significantly promote the proliferation and activation of endogenous γδ T cells in the intestine, leading to generation of large amount of interferon-γ (IFNγ). Moreover GNP-treated mice showed increased levels of chemokine CXCR3 in intestinal γδ T cells, which can drive their migration from the gut to the tumour environment. The translocation of γδ T cells and IFNγ from the intestine to extraintestinal subcutaneous tumours remodels the tumour immune microenvironment and synergizes with anti-PD-L1, inducing robust antitumour immunity. Our study delineates mechanistic insight into the complex gut-tumour interactome and provides an alternative approach for γδ T cell-based immunotherapy.
Collapse
MESH Headings
- Animals
- Interferon-gamma/metabolism
- Nanoparticles/chemistry
- Garlic/chemistry
- Mice
- Administration, Oral
- Immunotherapy/methods
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Mice, Inbred C57BL
- Receptors, CXCR3/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Cell Line, Tumor
- Female
- B7-H1 Antigen/metabolism
- Intestines/immunology
- Humans
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Neoplasms/therapy
- Neoplasms/immunology
Collapse
Affiliation(s)
- Jialu Xu
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Yue Yu
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Yue Zhang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Huaxing Dai
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Qianyu Yang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Beilei Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Qingle Ma
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Yitong Chen
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Fang Xu
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Xiaolin Shi
- Medical College of Soochow University, Suzhou, China
| | - Zhuang Liu
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China.
| | - Chao Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, China.
| |
Collapse
|
24
|
Wojas-Krawczyk K, Krawczyk P, Błach J, Kucharczyk T, Grenda A, Krzyżanowska N, Szklener K, Horaczyńska-Wojtaś A, Wójcik-Superczyńska M, Chmielewska I, Milanowski J. Immunological insights: assessing immune parameters in medical professionals exposed to SARS-CoV-2. BMC Infect Dis 2024; 24:865. [PMID: 39187767 PMCID: PMC11348584 DOI: 10.1186/s12879-024-09772-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND The immunological background responsible for the severe course of COVID-19 and the immune factors that protect against SARS-CoV-2 infection are still unclear. The aim of this study was to investigate immune system status in persons with high exposure to SARS-CoV-2 infection. METHODS Seventy-one persons employed in the observation and infectious diseases unit were qualified for the study between November 2020 and October 2021. Symptomatic COVID-19 was diagnosed in 35 persons. Anti-SARS-CoV-2 antibodies were also found in 8 persons. Peripheral blood mononuclear cells subpopulations were analyzed by flow cytometry, and the concentrations of cytokines and anti-SARS-CoV-2 antibodies were determined by ELISA. RESULTS The percentages of cytotoxic T lymphocytes (CTLs), CD28+ and T helper (Th) cells with invariant T-cell receptors were significantly higher in persons with symptomatic COVID-19 than in those who did not develop COVID-19' symptoms. Conversely, symptomatic COVID-19 persons had significantly lower percentages of: a) CTLs in the late stage of activation (CD8+/CD95+), b) NK cells, c) regulatory-like Th cells (CD4+/CTLA-4+), and d) Th17-like cells (CD4+/CD161+) compared to asymptomatic COVID-19' persons. Additionally, persons with anti-SARS-CoV-2 antibodies had a significantly higher lymphocyte count and IL-6 concentration than persons without these antibodies. CONCLUSION Numerous lymphocyte populations are permanently altered by SARS-CoV-2 infection. High percentages of both populations: NK cells-as a part of the non-specific response, and T helper cells' as those regulating the immune response, could protect against the acute COVID-19 symptoms development. Understanding the immune background of COVID-19 may improve the prevention of this disease by identifying people at risk of a severe course of infection. TRIAL REGISTRATION This is a retrospective observational study without a trial registration number.
Collapse
Affiliation(s)
- Kamila Wojas-Krawczyk
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland.
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Justyna Błach
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
- Department of Clinical Immunology Medical University of Lublin, Lublin, Poland
| | - Tomasz Kucharczyk
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Anna Grenda
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Natalia Krzyżanowska
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy Medical University of Lublin, Lublin, Poland
| | - Anna Horaczyńska-Wojtaś
- Department of Pediatric Otolaryngology, Phoniatrics and Audiology, University Children's Hospital, Lublin, Poland
| | - Magdalena Wójcik-Superczyńska
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| |
Collapse
|
25
|
Yang Y, Chen X, Pan J, Ning H, Zhang Y, Bo Y, Ren X, Li J, Qin S, Wang D, Chen MM, Zhang Z. Pan-cancer single-cell dissection reveals phenotypically distinct B cell subtypes. Cell 2024; 187:4790-4811.e22. [PMID: 39047727 DOI: 10.1016/j.cell.2024.06.038] [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: 06/09/2023] [Revised: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Characterizing the compositional and phenotypic characteristics of tumor-infiltrating B cells (TIBs) is important for advancing our understanding of their role in cancer development. Here, we establish a comprehensive resource of human B cells by integrating single-cell RNA sequencing data of B cells from 649 patients across 19 major cancer types. We demonstrate substantial heterogeneity in their total abundance and subtype composition and observe immunoglobulin G (IgG)-skewness of antibody-secreting cell isotypes. Moreover, we identify stress-response memory B cells and tumor-associated atypical B cells (TAABs), two tumor-enriched subpopulations with prognostic potential, shared in a pan-cancer manner. In particular, TAABs, characterized by a high clonal expansion level and proliferative capacity as well as by close interactions with activated CD4 T cells in tumors, are predictive of immunotherapy response. Our integrative resource depicts distinct clinically relevant TIB subsets, laying a foundation for further exploration of functional commonality and diversity of B cells in cancer.
Collapse
Affiliation(s)
- Yu Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xueyan Chen
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jieying Pan
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Huiheng Ning
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jiesheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Shishang Qin
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Min-Min Chen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
26
|
Wu TN, Hung JT, Hung TH, Wang YH, Wu JC, Yu AL. Effective suppression of tumor growth and hepatic metastasis of neuroblastoma by NKT-stimulatory phenyl glycolipid. Biomed Pharmacother 2024; 177:117040. [PMID: 38959605 DOI: 10.1016/j.biopha.2024.117040] [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: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Invariant natural killer T cell (iNKT) cells produce large amounts of cytokines in response to α-Galactosylceramide (α-GalCer) stimulation. An analog containing two phenyl rings on the acyl chain, C34, was previously found to be more Th1-biased than α-GalCer and triggered greater anticancer activities against breast cancer, melanoma and lung cancer in mice. Since liver is enriched in iNKT cells, we investigated anticancer efficacy of C34 on neuroblastoma with hepatic metastasis. C34 induced Th1-biased cytokine secretions in the liver, significantly suppressed neuroblastoma growth/metastasis and prolonged mouse survival. The anti-tumor efficacy might be attributed to greater expansions of hepatic NKT, NK, CD4+ T, and CD8+ T cells as well as reduction of the number of SSCloGr1intCD11b+ subset of myeloid-derived suppressor cells (MDSCs) in the liver of tumor-bearing mice, as compared to DMSO control group. C34 also upregulated expression of CD1d and CD11c, especially in the SSCloGr1intCD11b+ subset of MDSCs, which might be killed by C34-activated NKT cells, attributing to their reduced number. In addition, C34 also induced expansion of CD4+ T, CD8+ T, and NK cells, which might eliminate neuroblastoma cells. These immune-modulating effects of C34 might act in concert in the local milieu of liver to suppress the tumor growth. Further analysis of database of neuroblastoma revealed that patients with high CD11c expression in the monocytic MDSCs in the tumor had longer survival, suggesting the potential clinical application of C34 for treatment of neuroblastoma.
Collapse
Affiliation(s)
- Tai-Na Wu
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; Institute of Biotechnology, National Taiwan University, Taipei 115, Taiwan.
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan.
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Hui Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Jen-Chine Wu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, University of California in San Diego, La Jolla, CA, USA
| |
Collapse
|
27
|
Wei Y, Zhu X, Lin S, Yang W, Wang T, Nie X, Shi Z, Liu Z, Zhang R, Li D. Zinc gluconate improves atopic dermatitis by modulating CXCL10 release of keratinocytes via PPARα activation. Biomed Pharmacother 2024; 177:117129. [PMID: 39018874 DOI: 10.1016/j.biopha.2024.117129] [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: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition with complex causes involving immune factors. The presence of essential trace elements that support immune system function can influence the development of this condition. This study investigated how serum trace elements impact the pathogenesis of atopic dermatitis. Upon analyzing serum microelements in AD patients and control subjects, it was observed that patients with AD had notably lower zinc levels. Genomic analysis of AD skin revealed distinct gene expression patterns, specifically the increased expression of CXCL10 in the epidermis. The heightened levels of CXCL10 in AD skin lesions were found to correlate with reduced serum zinc levels. Treatment with zinc gluconate showed reduced chemotactic response and CXCL10 release, suggesting its potential to regulate CXCL10 expression of keratinocytes in AD. The mechanism behind this involved the downregulation of STAT phosphorylation through activating PPARα. In the AD-like dermatitis mouse model, zinc gluconate therapy decreased serum IgE levels, alleviated skin lesion severity, reduced skin thickness, and lowered CXCL10 expression, demonstrating its efficacy in managing AD-like skin conditions. These findings indicate that zinc gluconate can reduce inflammation in keratinocytes by activating PPARα, inhibiting STAT signaling, and decreasing CXCL10 release, thus highlighting its potential as a therapeutic target for AD.
Collapse
Affiliation(s)
- Yujia Wei
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| | - Xiaomei Zhu
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| | - Shan Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tingmei Wang
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| | - Xiaoqi Nie
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| | - Zeqi Shi
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| | - Zhong Liu
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| | - Ri Zhang
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| | - Dong Li
- Department of Dermatology, Huazhong University of Science and Technology Tongji Medical College Tongji Hospital, Wuhan, China.
| |
Collapse
|
28
|
Xuan Z, Chen X, Zhou W, Shen Y, Sun Z, Zhang H, Yao Z. Exploring causal correlations between circulating cytokines and atopic dermatitis: a bidirectional two-sample Mendelian randomization study. Front Immunol 2024; 15:1367958. [PMID: 39055710 PMCID: PMC11269137 DOI: 10.3389/fimmu.2024.1367958] [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: 01/09/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Objectives Numerous observational studies have reported associations between circulating cytokines and atopic dermatitis (AD); however, the causal relationships between them remain unclear. To explore the causal correlations and direction of causal effects between AD and levels of 91 circulating cytokines. Methods Two-sample Mendelian randomization (MR) analyses were conducted to examine the causal relationships between 91 circulating cytokines and AD using summary statistics from genome-wide association studies (GWAS). Reverse MR analyses were performed to investigate reverse causation. Pleiotropy and heterogeneity tests were conducted to assess the robustness of the findings. Additional transcriptome database and clinical peripheral blood mononuclear cells (PBMCs) samples were utilized to validate the results of MR analyses. Results Levels of interleukin (IL)-13, IL-18 Receptor 1, Tumor necrosis factor ligand superfamily member 14 (TNFSF14), TNF-related activation-induced cytokine (TRANCE), C-X-C motif chemokine (CXCL)11, IL-33, TNF-beta and CD5 were suggestively associated with the risk of AD (odds ratio, OR: 1.202, 95% CI: 1.018-1.422, p = 0.030; OR: 1.029, 95% CI: 1.029-1.157, p = 0.004; OR: 1.159, 95% CI: 1.018-1.320, p = 0.026; OR: 1.111, 95% CI: 1.016-1.214, p = 0.020; OR: 0.878, 95% CI: 0.783-0.984, p = 0.025; OR: 0.809, 95% CI: 0.661-0.991, p = 0.041; OR: 0.945, 95% CI: 0.896-0.997, p = 0.038; OR: 0.764, 95% CI: 0.652-0.895, p = 8.26e-04). In addition, levels of cytokines including Axin-1, CXCL5, CXCL10, Oncostatin-M (OSM), Sulfotransferase 1A1 (SULT1A1) and TNFSF14 were suggested to be consequences of AD (Beta: -0.080, p = 0.016; Beta: -0.062, p = 0.036; Beta: -0.066, p = 0.049; Beta: -0.073, p = 0.013; Beta: -0.089, p = 0.008; Beta: -0.079, p = 0.031). IL-13, IL-18R1, TNFSF14, and TRANCE were upregulated in both lesional skin biopsies and PBMCs from AD patients. Conclusion The study indicates that several cytokines, including IL-13, IL-18R1, TNFSF14, TRANCE, CXCL11, IL-33, TNF-beta, and CD5, are upstream of AD development, whereas a few circulating cytokines are potentially downstream in the development of AD.
Collapse
Affiliation(s)
- Zhenquan Xuan
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuanyi Chen
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weinan Zhou
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yihang Shen
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhe Sun
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hui Zhang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Yao
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Chatelain C, Berland L, Grard M, Jouand N, Fresquet J, Nader J, Hirigoyen U, Petithomme T, Combredet C, Pons-Tostivint E, Fradin D, Treps L, Blanquart C, Boisgerault N, Tangy F, Fonteneau JF. Interplay between oncolytic measles virus, macrophages and cancer cells induces a proinflammatory tumor microenvironment. Oncoimmunology 2024; 13:2377830. [PMID: 39005546 PMCID: PMC11244337 DOI: 10.1080/2162402x.2024.2377830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Attenuated measles virus (MV) exerts its oncolytic activity in malignant pleural mesothelioma (MPM) cells that lack type-I interferon (IFN-I) production or responsiveness. However, other cells in the tumor microenvironment (TME), such as myeloid cells, possess functional antiviral pathways. In this study, we aimed to characterize the interplay between MV and the myeloid cells in human MPM. We cocultured MPM cell lines with monocytes or macrophages and infected them with MV. We analyzed the transcriptome of each cell type and studied their secretion and phenotypes by high-dimensional flow cytometry. We also measured transgene expression using an MV encoding GFP (MV-GFP). We show that MPM cells drive the differentiation of monocytes into M2-like macrophages. These macrophages inhibit GFP expression in tumor cells harboring a defect in IFN-I production and a functional signaling downstream of the IFN-I receptor, while having minimal effects on GFP expression in tumor cells with defect of responsiveness to IFN-I. Interestingly, inhibition of the IFN-I signaling by ruxolitinib restores GFP expression in tumor cells. Upon MV infection, cocultured macrophages express antiviral pro-inflammatory genes and induce the expression of IFN-stimulated genes in tumor cells. MV also increases the expression of HLA and costimulatory molecules on macrophages and their phagocytic activity. Finally, MV induces the secretion of inflammatory cytokines, especially IFN-I, and PD-L1 expression in tumor cells and macrophages. These results show that macrophages reduce viral proteins expression in some MPM cell lines through their IFN-I production and generate a pro-inflammatory interplay that may stimulate the patient's anti-tumor immune response.
Collapse
Affiliation(s)
- Camille Chatelain
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Laurine Berland
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Marion Grard
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Nicolas Jouand
- LabEx IGO, Nantes Université, Nantes, France
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Judith Fresquet
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Joëlle Nader
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Ugo Hirigoyen
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Tacien Petithomme
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Chantal Combredet
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Elvire Pons-Tostivint
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
- Centre Hospitalier Universitaire Nantes, Medical Oncology, Nantes University, Nantes, France
| | - Delphine Fradin
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Lucas Treps
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Christophe Blanquart
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Nicolas Boisgerault
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Frédéric Tangy
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
- Oncovita, Paris, France
| | - Jean-François Fonteneau
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| |
Collapse
|
30
|
Zhou H, Wang W, Xu H, Liang Y, Ding J, Lv M, Ren B, Peng H, Fu YX, Zhu M. Metabolic reprograming mediated by tumor cell-intrinsic type I IFN signaling is required for CD47-SIRPα blockade efficacy. Nat Commun 2024; 15:5759. [PMID: 38982116 PMCID: PMC11233683 DOI: 10.1038/s41467-024-50136-z] [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: 06/10/2023] [Accepted: 07/02/2024] [Indexed: 07/11/2024] Open
Abstract
Type I interferons have been well recognized for their roles in various types of immune cells during tumor immunotherapy. However, their direct effects on tumor cells are less understood. Oxidative phosphorylation is typically latent in tumor cells. Whether oxidative phosphorylation can be targeted for immunotherapy remains unclear. Here, we find that tumor cell responsiveness to type I, but not type II interferons, is essential for CD47-SIRPα blockade immunotherapy in female mice. Mechanistically, type I interferons directly reprogram tumor cell metabolism by activating oxidative phosphorylation for ATP production in an ISG15-dependent manner. ATP extracellular release is also promoted by type I interferons due to enhanced secretory autophagy. Functionally, tumor cells with genetic deficiency in oxidative phosphorylation or autophagy are resistant to CD47-SIRPα blockade. ATP released upon CD47-SIRPα blockade is required for antitumor T cell response induction via P2X7 receptor-mediated dendritic cell activation. Based on this mechanism, combinations with inhibitors of ATP-degrading ectoenzymes, CD39 and CD73, are designed and show synergistic antitumor effects with CD47-SIRPα blockade. Together, these data reveal an important role of type I interferons on tumor cell metabolic reprograming for tumor immunotherapy and provide rational strategies harnessing this mechanism for enhanced efficacy of CD47-SIRPα blockade.
Collapse
Affiliation(s)
- Hang Zhou
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Wenjun Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Hairong Xu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yong Liang
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Jiyu Ding
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Mengjie Lv
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Boyang Ren
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hua Peng
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.
| | - Mingzhao Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
31
|
Hunter C, Larimer B. Chemokine receptor PET imaging: Bridging molecular insights with clinical applications. Nucl Med Biol 2024; 134-135:108912. [PMID: 38691942 PMCID: PMC11180593 DOI: 10.1016/j.nucmedbio.2024.108912] [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: 11/10/2023] [Revised: 03/07/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Chemokine receptors are important components of cellular signaling and play a critical role in directing leukocytes during inflammatory reactions. Their importance extends to numerous pathological processes, including tumor differentiation, angiogenesis, metastasis, and associations with multiple inflammatory disorders. The necessity to monitor the in vivo interactions of cellular chemokine receptors has been driven the recent development of novel positron emission tomography (PET) imaging agents. This imaging modality provides non-invasive localization and quantitation of these receptors that cannot be provided through blood or tissue-based assays. Herein, we provide a review of PET imaging of the chemokine receptors that have been imaged to date, namely CXCR3, CXCR4, CCR2, CCR5, and CMKLR1. The quantification of these receptors can aid in understanding various diseases, including cancer, atherosclerosis, idiopathic pulmonary fibrosis, and acute respiratory distress syndrome. The development of specific radiotracers targeting these receptors will be discussed, including promising results for disease diagnosis and management. However, challenges persist in fully translating these imaging advancements into practical therapeutic applications. Given the success of CXCR4 PET imaging to date, future research should focus on clinical translation of these approaches to understand their role in the management of a wide variety of diseases.
Collapse
Affiliation(s)
- Chanelle Hunter
- Graduate Biomedical Sciences Cancer Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Benjamin Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, AL 35294, USA.
| |
Collapse
|
32
|
Yan Z, Wen JX, Niu Y, Jiang TW, Huang JH, Chen H, Chen Q, Wang YF, Yan L, Hu ZD, Zheng WQ. Diagnostic accuracy and cellular origin of pleural fluid CXCR3 ligands for tuberculous pleural effusion. Cytokine 2024; 179:156618. [PMID: 38663252 DOI: 10.1016/j.cyto.2024.156618] [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: 02/18/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Pleural biomarkers represent potential diagnostic tools for tuberculous pleural effusion (TPE) due to their advantages of low cost, short turnaround time, and less invasiveness. This study evaluated the diagnostic accuracy of two CXCR3 ligands, C-X-C motif chemokine ligand 9 (CXCL9) and CXCL11, for TPE. In addition, we investigated the cellular origins and biological roles of CXCL9 and CXCL11 in the development of TPE. METHODS This double-blind study prospectively enrolled patients with undiagnosed pleural effusion from two centers (Hohhot and Changshu) in China. Pleural fluid on admission was obtained and levels of CXCL9 and CXCL11 were measured by an enzyme-linked immunosorbent assay (ELISA). The receiver operating characteristic (ROC) curve and the decision curve analysis (DCA) were used to evaluate their diagnostic accuracy and net benefit, respectively. THP-1 cell-derived macrophages were treated with Bacillus Calmette-Guérin (BCG), and quantitative real-time PCR (qRT-PCR) and ELISA were used to determine the mRNA and protein levels of CXCL9 and CXCL11. The chemoattractant activities of CXCL9 and CXCL11 for T helper (Th) cells were analyzed by a transwell assay. RESULTS One hundred and fifty-three (20 TPEs and 133 non-TPEs) patients were enrolled in the Hohhot Center, and 58 (13 TPEs and 45 non-TPEs) were enrolled in the Changshu Center. In both centers, we observed increased CXCL9 and CXCL11 in TPE patients. The areas under the ROC curves (AUCs) of pleural CXCL9 and CXCL11 in the Hohhot Center were 0.70 (95 % CI: 0.55-0.85) and 0.68 (95 % CI: 0.52-0.84), respectively. In the Changshu Center, the AUCs of CXCL9 and CXCL11 were 0.96 (95 % CI: 0.92-1.00) and 0.97 (95 % CI: 0.94-1.00), respectively. The AUCs of CXCL9 and CXCL11 decreased with the advancement of age. The decision curves of CXCL9 and CXCL11 showed net benefits in both centers. CXCL9 and CXCL11 were upregulated in BCG-treated macrophages. Pleural fluid from TPE and conditioned medium from BCG-treated macrophages were chemotactic for Th cells. Anti-CXCL9 or CXCL11 neutralizing antibodies could partly block the chemotactic activity. CONCLUSIONS Pleural CXCL9 and CXCL11 are potential diagnostic markers for TPE, but their diagnostic accuracy is compromised in elderly patients. CXCL9 and CXCL11 can promote the migration of peripheral Th cells, thus representing a therapeutic target for the treatment of TPE.
Collapse
Affiliation(s)
- Zhi Yan
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China; School of Medical Laboratory & Department of Cell Biology, Tianjin Medical University, Tianjin 300203, China
| | - Jian-Xun Wen
- Department of Medical Experiment Center, the Basic Medical Sciences College of Inner Mongolia Medical University, Hohhot 010050, China
| | - Yan Niu
- Department of Medical Experiment Center, the Basic Medical Sciences College of Inner Mongolia Medical University, Hohhot 010050, China
| | - Ting-Wang Jiang
- Department of Key Laboratory, the Affiliated Changshu Hospital of Nantong University, Changshu 215500, China
| | - Jin-Hong Huang
- Department of Pulmonary and Critical Care Medicine, the Affiliated Changshu Hospital of Nantong University, Changshu 215500, China
| | - Hong Chen
- Department of Pulmonary and Critical Care Medicine, the Affiliated Changshu Hospital of Nantong University, Changshu 215500, China
| | - Qi Chen
- The Third Clinical Medical College of Ningxia Medical University, Yinchuan 750004, China
| | - Ya-Fei Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China; Key Laboratory for Biomarkers, Inner Mongolia Medical University, Hohhot 010050, China
| | - Li Yan
- Key Laboratory for Biomarkers, Inner Mongolia Medical University, Hohhot 010050, China; Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Zhi-De Hu
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China; Key Laboratory for Biomarkers, Inner Mongolia Medical University, Hohhot 010050, China
| | - Wen-Qi Zheng
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China; Key Laboratory for Biomarkers, Inner Mongolia Medical University, Hohhot 010050, China.
| |
Collapse
|
33
|
Yang W, Cao J, Di S, Chen W, Cheng H, Ren H, Xie Y, Chen L, Yu M, Chen Y, Cui X. Immunogenic Material Vaccine for Cancer Immunotherapy by Structure-Dependent Immune Cell Trafficking and Modulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402580. [PMID: 38630978 DOI: 10.1002/adma.202402580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/15/2024] [Indexed: 04/19/2024]
Abstract
Inherently immunogenic materials offer enormous prospects in enhancing vaccine efficacy. However, the understanding and improving material adjuvanticity remain elusive. Herein how the structural presentation of immunopotentiators in a material governs the dynamic dialogue between innate and adaptive immunity for enhanced cancer vaccination is reported. The immunopotentiator manganese into six differing structures that resemble the architectures of two types of pathogens (spherical viruses or rod-like bacteria) is precisely manipulated. The results reveal that innate immune cells accurately sense and respond to the architectures, of which two outperformed material candidates (151 nm hollow spheres and hollow microrods with an aspect ratio of 4.5) show higher competence in creating local proinflammatory environment with promoted innate immune cell influx and stimulation on dendritic cells (DCs). In combination with viral peptides, model proteins, or cell lysate antigens, the outperformed microrod material remarkably primes antigen-specific CD8 cytolytic T cells. In prophylactic and therapeutic regimens, the microrod adjuvanted vaccines display optimal aptitude in tumor suppression in four aggressive murine tumor models, by promoting the infiltration of heterogeneous cytolytic effector cells while decreasing suppressive immunoregulatory populations in tumors. This study demonstrates that a rationally selected architecture of immunogenic materials potentially advances the clinical reality of cancer vaccination.
Collapse
Affiliation(s)
- Wei Yang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, P. R. China
| | - Jianwei Cao
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, P. R. China
| | - Sichen Di
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, P. R. China
| | - Wenjin Chen
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, P. R. China
| | - Hui Cheng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, P. R. China
| |
Collapse
|
34
|
Kim JW, Ahn MH, Jung JY, Suh CH, Han JH, Kim HA. Role of chemokines CXCL9, CXCL10, CXCL11, and CXCR3 in the serum and minor salivary gland tissues of patients with Sjögren's syndrome. Clin Exp Med 2024; 24:133. [PMID: 38900301 PMCID: PMC11189950 DOI: 10.1007/s10238-024-01401-4] [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: 05/02/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
This study aimed to investigate the serum and expression levels of C-X-C motif chemokine ligand 9 (CXCL9), CXCL10, CXCL11, and CXC receptor 3 (CXCR3) in minor salivary glands (MSGs) of patients with primary Sjögren's syndrome (pSS), and to explore their correlations with clinical parameters. Serum samples from 49 patients diagnosed with pSS, 33 patients with rheumatoid arthritis (RA), and 30 healthy controls (HCs) were collected for measurements of CXCL9, CXCL10, CXCL11, and CXCR3. Additionally, CXCL levels in the MSG tissues were measured in 41 patients who underwent MSG biopsy. Correlations between CXCL and CXCL/CXCR levels in serum/MSG tissues and clinical factors/salivary scintigraphy parameters were analyzed. Serum CXCL11 and CXCR3 showed statistically significant differences among patients with pSS and RA and HCs (serum CXCL11, pSS:RA:HC = 235.6 ± 500.1 pg/mL:90.0 ± 200.3 pg/mL:45.9 ± 53.6 pg/mL; p = 0.041, serum CXCR3, pSS:RA:HC = 3.27 ± 1.32 ng/mL:3.29 ± 1.17 ng/mL:2.00 ± 1.12 ng/mL; p < 0.001). Serum CXCL10 showed a statistically significant difference between pSS (64.5 ± 54.2 pg/mL) and HCs (18.6 ± 18.1 pg/mL, p < 0.001), while serum CXCL9 did not exhibit a significant difference among the groups. Correlation analysis of clinical factors revealed that serum CXCL10 and CXCL11 levels positively correlated with erythrocyte sedimentation rate (r = 0.524, p < 0.001 and r = 0.707, p < 0.001, respectively), total protein (r = 0.375, p = 0.008 and r = 0.535, p < 0.001, respectively), globulin (r = 0.539, p < 0.001 and r = 0.639, p < 0.001, respectively), and European Alliance of Associations for Rheumatology SS Disease Activity Index (r = 0.305, p = 0.033 and r = 0.321, p = 0.025). Additionally, serum CXCL10 negatively correlated with the Schirmer test score (r = - 0.354, p = 0.05), while serum CXCL11 positively correlated with the biopsy focus score (r = 0.612, p = 0.02). In the MSG tissue, the percentage of infiltrating CXCL9-positive cells was highest (75.5%), followed by CXCL10 (29.1%) and CXCL11 (27.9%). In the correlation analysis, CXCL11-expressing cells were inversely related to the mean washout percentage on salivary gland scintigraphy (r = - 0.448, p = 0.007). Our study highlights distinct serum and tissue chemokine patterns in pSS, emphasizing CXCL9's potential for early diagnosis. This suggests that CXCL10 and CXCL11 are indicators of disease progression, warranting further investigation into their roles in autoimmune disorders beyond pSS.
Collapse
Affiliation(s)
- Ji-Won Kim
- Department of Rheumatology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Mi-Hyun Ahn
- Department of Rheumatology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Ju-Yang Jung
- Department of Rheumatology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea
| | - Jae Ho Han
- Department of Pathology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea.
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 16499, Republic of Korea.
| |
Collapse
|
35
|
柴 晓, 孙 子, 李 海, 朱 靓, 刘 小, 刘 延, 裴 斐, 常 青. [Clinicopathological characteristics of the CD8 + T lymphocytes infiltration and its mechanism in distinct molecular subtype of medulloblastoma]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2024; 56:512-518. [PMID: 38864138 PMCID: PMC11167556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To investigate the characteristics of the CD8+ T cells infiltration from the 4 subtypes in medulloblastoma (MB), to analyze the relationship between CD8+ T cells infiltration and prognosis, to study the function of C-X-C motif chemokine ligand 11 (CXCL11) and its receptor in CD8+ T cells infiltration into tumors and to explore the potential mechanism, and to provide the necessary clinicopathological basis for exploring the immunotherapy of MB. METHODS In the study, 48 clinical MB samples (12 cases in each of 4 subtypes) were selected from the multiple medical center from 2012 to 2019. The transcriptomics analysis for the tumor of 48 clinical samples was conducted on the NanoString PanCancer IO360TM Panel (NanoString Technologies). Immunohistochemistry (IHC) staining of formalin-fixed, paraffin-embedded sections from MB was carried out using CD8 primary antibody to analyze diffe-rential quantities of CD8+ T cells in the MB four subtypes. Through bioinformatics analysis, the relationship between CD8+T cells infiltration and prognosis of the patients and the expression differences of various chemokines in the different subtypes of MB were investigated. The expression of CXCR3 receptor on the surface of CD8+T cells in MB was verified by double immunofluorescence staining, and the underlying molecular mechanism of CD8+T cells infiltration into the tumor was explored. RESULTS The characteristic index of CD8+T cells in the WNT subtype of MB was relatively high, suggesting that the number of CD8+T cells in the WNT subtype was significantly higher than that in the other three subtypes, which was confirmed by CD8 immunohistochemical staining and Gene Expression Omnibus (GEO) database analysis by using R2 online data analysis platform. And the increase of CD8+T cells infiltration was positively correlated with the patient survival. The expression level of CXCL11 in the WNT subtype MB was significantly higher than that of the other three subtypes. Immunofluorescence staining showed the presence of CXCL11 receptor, CXCR3, on the surface of CD8+T cells, suggesting that the CD8+T cells might be attracted to the MB microenvironment by CXCL11 through CXCR3. CONCLUSION The CD8+T cells infiltrate more in the WNT subtype MB than other subtypes. The mechanism may be related to the activation of CXCL11-CXCR3 chemokine system, and the patients with more infiltration of CD8+T cells in tumor have better prognosis. This finding may provide the necessary clinicopathological basis for the regulatory mechanism of CD8+T cells infiltration in MB, and give a new potential therapeutic target for the future immunotherapy of MB.
Collapse
Affiliation(s)
- 晓东 柴
- 北京大学第三医院病理科, 北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
- 北京大学基础医学院病理学系, 北京 100191Department of Pathology, Peking University School of Basic Medical Sciences, Beijing 100191, China
| | - 子文 孙
- 北京大学第三医院病理科, 北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
| | - 海爽 李
- 北京大学第三医院病理科, 北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
| | - 靓怡 朱
- 北京大学第三医院病理科, 北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
| | - 小旦 刘
- 北京大学第三医院病理科, 北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
- 北京大学基础医学院病理学系, 北京 100191Department of Pathology, Peking University School of Basic Medical Sciences, Beijing 100191, China
| | - 延涛 刘
- 北京大学第三医院病理科, 北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
| | - 斐 裴
- 北京大学第三医院病理科, 北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
- 北京大学基础医学院病理学系, 北京 100191Department of Pathology, Peking University School of Basic Medical Sciences, Beijing 100191, China
| | - 青 常
- 北京市神经外科研究所神经病理中心, 北京 100070Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Beijing 100070, China
| |
Collapse
|
36
|
Reschke R, Enk AH, Hassel JC. Chemokines and Cytokines in Immunotherapy of Melanoma and Other Tumors: From Biomarkers to Therapeutic Targets. Int J Mol Sci 2024; 25:6532. [PMID: 38928238 PMCID: PMC11203481 DOI: 10.3390/ijms25126532] [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: 05/09/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Chemokines and cytokines represent an emerging field of immunotherapy research. They are responsible for the crosstalk and chemoattraction of immune cells and tumor cells. For instance, CXCL9/10/11 chemoattract effector CD8+ T cells to the tumor microenvironment, making an argument for their promising role as biomarkers for a favorable outcome. The cytokine Interleukin-15 (IL-15) can promote the chemokine expression of CXCR3 ligands but also XCL1, contributing to an important DC-T cell interaction. Recruited cytotoxic T cells can be clonally expanded by IL-2. Delivering or inducing these chemokines and cytokines can result in tumor shrinkage and might synergize with immune checkpoint inhibition. In addition, blocking specific chemokine and cytokine receptors such as CCR2, CCR4 or Il-6R can reduce the recruitment of tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs) or regulatory T cells (Tregs). Efforts to target these chemokines and cytokines have the potential to personalize cancer immunotherapy further and address patients that are not yet responsive because of immune cell exclusion. Targeting cytokines such as IL-6 and IL-15 is currently being evaluated in clinical trials in combination with immune checkpoint-blocking antibodies for the treatment of metastatic melanoma. The improved overall survival of melanoma patients might outweigh potential risks such as autoimmunity. However, off-target toxicity needs to be elucidated.
Collapse
Affiliation(s)
- Robin Reschke
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Alexander H. Enk
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Jessica C. Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Liang R, Li P, Yang N, Xiao X, Gong J, Zhang X, Bai Y, Chen Y, Xie Z, Liao Q. Parabacteroides distasonis-Derived Outer Membrane Vesicles Enhance Antitumor Immunity Against Colon Tumors by Modulating CXCL10 and CD8 + T Cells. Drug Des Devel Ther 2024; 18:1833-1853. [PMID: 38828018 PMCID: PMC11144014 DOI: 10.2147/dddt.s457338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Purpose Given the potent immunostimulatory effects of bacterial outer membrane vesicles (OMVs) and the significant anti-colon tumor properties of Parabacteroides distasonis (Pd), this study aimed to elucidate the role and potential mechanisms of Pd-derived OMVs (Pd-OMVs) against colon cancer. Methods This study isolated and purified Pd-OMVs from Pd cultures and assessed their characteristics. The effects of Pd-OMVs on CT26 cell uptake, proliferation, and invasion were investigated in vitro. In vivo, a CT26 colon tumor model was used to investigate the anti-colon tumor effects and underlying mechanisms of Pd-OMVs. Finally, we evaluated the biosafety of Pd-OMVs. Results Purified Pd-OMVs had a uniform cup-shaped structure with an average size of 165.5 nm and a zeta potential of approximately -9.56 mV, and their proteins were associated with pathways related to immunity and apoptosis. In vitro experiments demonstrated that CT26 cells internalized the Pd-OMVs, resulting in a significant decrease in their proliferation and invasion abilities. Further in vivo studies confirmed the accumulation of Pd-OMVs in tumor tissues, which significantly inhibited the growth of colon tumors. Mechanistically, Pd-OMVs increased the expression of CXCL10, promoting infiltration of CD8+ T cells into tumor tissues and expression of pro-inflammatory factors TNF-α, IL-1β, and IL-6. Notably, Pd-OMVs demonstrated a high level of biosafety. Conclusion This paper elucidates that Pd-OMVs can exert significant anti-colon tumor effects by upregulating the expression of the chemokine CXCL10, thereby increasing the infiltration of CD8+ T cells into tumors and enhancing antitumor immune responses. This suggests that Pd-OMVs may be developed as a novel nanoscale potent immunostimulant with great potential for application in tumor immunotherapy. As well as developed as a novel nano-delivery carrier for combination with other antitumor drugs.
Collapse
Affiliation(s)
- Rongyao Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Pei Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Na Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Xiaoyi Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Jing Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Xingyuan Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yunuan Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yanlong Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People’s Republic of China
| |
Collapse
|
38
|
Wu K, Zhang G, Shen C, Zhu L, Yu C, Sartorius K, Ding W, Jiang Y, Lu Y. Role of T cells in liver metastasis. Cell Death Dis 2024; 15:341. [PMID: 38755133 PMCID: PMC11099083 DOI: 10.1038/s41419-024-06726-2] [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: 12/20/2023] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The liver is a major metastatic site (organ) for gastrointestinal cancers (such as colorectal, gastric, and pancreatic cancers) as well as non-gastrointestinal cancers (such as lung, breast, and melanoma cancers). Due to the innate anatomical position of the liver, the apoptosis of T cells in the liver, the unique metabolic regulation of hepatocytes and other potential mechanisms, the liver tends to form an immunosuppressive microenvironment and subsequently form a pre-metastatic niche (PMN), which can promote metastasis and colonization by various tumor cells(TCs). As a result, the critical role of immunoresponse in liver based metastasis has become increasingly appreciated. T cells, a centrally important member of adaptive immune response, play a significant role in liver based metastases and clarifying the different roles of the various T cells subsets is important to guide future clinical treatment. In this review, we first introduce the predisposing factors and related mechanisms of liver metastasis (LM) before introducing the PMN and its transition to LM. Finally, we detail the role of different subsets of T cells in LM and advances in the management of LM in order to identify potential therapeutic targets for patients with LM.
Collapse
Affiliation(s)
- Kejia Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Guozhu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Emergency Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Changbing Shen
- Department of Hepatobiliary and Pancreatic Surgery, Taizhou Second People's Hospital Affiliated with Yangzhou University, Taizhou, China
| | - Li Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Emergency Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Chongyuan Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Kurt Sartorius
- School of Laboratory Medicine and Molecular Sciences, University of Kwazulu-Natal, Durban, South Africa
- Africa Hepatopancreatobiliary Cancer Consortium, Mayo Clinic, Jacksonville, FL, USA
| | - Wei Ding
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China.
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
| | - Yong Jiang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Yunjie Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Africa Hepatopancreatobiliary Cancer Consortium, Mayo Clinic, Jacksonville, FL, USA.
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China.
| |
Collapse
|
39
|
Gupta T, Antanaviciute A, Hyun-Jung Lee C, Ottakandathil Babu R, Aulicino A, Christoforidou Z, Siejka-Zielinska P, O'Brien-Ball C, Chen H, Fawkner-Corbett D, Geros AS, Bridges E, McGregor C, Cianci N, Fryer E, Alham NK, Jagielowicz M, Santos AM, Fellermeyer M, Davis SJ, Parikh K, Cheung V, Al-Hillawi L, Sasson S, Slevin S, Brain O, Fernandes RA, Koohy H, Simmons A. Tracking in situ checkpoint inhibitor-bound target T cells in patients with checkpoint-induced colitis. Cancer Cell 2024; 42:797-814.e15. [PMID: 38744246 DOI: 10.1016/j.ccell.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/09/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
The success of checkpoint inhibitors (CPIs) for cancer has been tempered by immune-related adverse effects including colitis. CPI-induced colitis is hallmarked by expansion of resident mucosal IFNγ cytotoxic CD8+ T cells, but how these arise is unclear. Here, we track CPI-bound T cells in intestinal tissue using multimodal single-cell and subcellular spatial transcriptomics (ST). Target occupancy was increased in inflamed tissue, with drug-bound T cells located in distinct microdomains distinguished by specific intercellular signaling and transcriptional gradients. CPI-bound cells were largely CD4+ T cells, including enrichment in CPI-bound peripheral helper, follicular helper, and regulatory T cells. IFNγ CD8+ T cells emerged from both tissue-resident memory (TRM) and peripheral populations, displayed more restricted target occupancy profiles, and co-localized with damaged epithelial microdomains lacking effective regulatory cues. Our multimodal analysis identifies causal pathways and constitutes a resource to inform novel preventive strategies.
Collapse
Affiliation(s)
- Tarun Gupta
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Agne Antanaviciute
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.
| | - Chloe Hyun-Jung Lee
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Rosana Ottakandathil Babu
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Anna Aulicino
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Zoe Christoforidou
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Paulina Siejka-Zielinska
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Caitlin O'Brien-Ball
- Chinese Academy of Medical Sciences (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7BN, UK
| | - Hannah Chen
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - David Fawkner-Corbett
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Academic Paediatric Surgery Unit (APSU), Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Ana Sousa Geros
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Esther Bridges
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Colleen McGregor
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Nicole Cianci
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Eve Fryer
- Pathology, Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Nasullah Khalid Alham
- Nuffield Department of Surgical Sciences and Oxford NIHR Biomedical Research Centre (BRC), University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Marta Jagielowicz
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Ana Mafalda Santos
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Martin Fellermeyer
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Simon J Davis
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Kaushal Parikh
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Vincent Cheung
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Lulia Al-Hillawi
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Sarah Sasson
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Stephanie Slevin
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Oliver Brain
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Ricardo A Fernandes
- Chinese Academy of Medical Sciences (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7BN, UK
| | - Hashem Koohy
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.
| | - Alison Simmons
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| |
Collapse
|
40
|
Cai N, Cheng K, Ma Y, Liu S, Tao R, Li Y, Li D, Guo B, Jia W, Liang H, Zhao J, Xia L, Ding ZY, Chen J, Zhang W. Targeting MMP9 in CTNNB1 mutant hepatocellular carcinoma restores CD8 + T cell-mediated antitumour immunity and improves anti-PD-1 efficacy. Gut 2024; 73:985-999. [PMID: 38123979 PMCID: PMC11103337 DOI: 10.1136/gutjnl-2023-331342] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE The gain of function (GOF) CTNNB1 mutations (CTNNB1 GOF ) in hepatocellular carcinoma (HCC) cause significant immune escape and resistance to anti-PD-1. Here, we aimed to investigate the mechanism of CTNNB1 GOF HCC-mediated immune escape and raise a new therapeutic strategy to enhance anti-PD-1 efficacy in HCC. DESIGN RNA sequencing was performed to identify the key downstream genes of CTNNB1 GOF associated with immune escape. An in vitro coculture system, murine subcutaneous or orthotopic models, spontaneously tumourigenic models in conditional gene-knock-out mice and flow cytometry were used to explore the biological function of matrix metallopeptidase 9 (MMP9) in tumour progression and immune escape. Single-cell RNA sequencing and proteomics were used to gain insight into the underlying mechanisms of MMP9. RESULTS MMP9 was significantly upregulated in CTNNB1 GOF HCC. MMP9 suppressed infiltration and cytotoxicity of CD8+ T cells, which was critical for CTNNB1 GOF to drive the suppressive tumour immune microenvironment (TIME) and anti-PD-1 resistance. Mechanistically, CTNNB1 GOF downregulated sirtuin 2 (SIRT2), resulting in promotion of β-catenin/lysine demethylase 4D (KDM4D) complex formation that fostered the transcriptional activation of MMP9. The secretion of MMP9 from HCC mediated slingshot protein phosphatase 1 (SSH1) shedding from CD8+ T cells, leading to the inhibition of C-X-C motif chemokine receptor 3 (CXCR3)-mediated intracellular of G protein-coupled receptors signalling. Additionally, MMP9 blockade remodelled the TIME and potentiated the sensitivity of anti-PD-1 therapy in HCC. CONCLUSIONS CTNNB1 GOF induces a suppressive TIME by activating secretion of MMP9. Targeting MMP9 reshapes TIME and potentiates anti-PD-1 efficacy in CTNNB1 GOF HCC.
Collapse
Affiliation(s)
- Ning Cai
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Kun Cheng
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yue Ma
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, People's Republic of China
| | - Sha Liu
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ran Tao
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yani Li
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Danfeng Li
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Bin Guo
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Wenlong Jia
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Huifang Liang
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jianping Zhao
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ze-Yang Ding
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jinhong Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, People's Republic of China
| | - Wanguang Zhang
- Hepatic Surgery Center, Clinical Medicine Research Center of Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
41
|
Yin T, Mou S, Zhang H, Dong Y, Yan B, Huang W, Liu Y, Mei H. CXCL10 could be a prognostic and immunological biomarker in bladder cancer. Discov Oncol 2024; 15:148. [PMID: 38720149 PMCID: PMC11078901 DOI: 10.1007/s12672-024-00982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION As proteins that promote immune cell differentiation, chemokines have attracted great interest regarding their role in anti-tumor immune responses within the cancer environment. However, the exact role of CXCL10, a chemokine, in bladder cancer (BLCA) is still not fully elucidated. METHOD In the present study, we employed bioinformatics approaches to examine the expression pattern, prognostic value, and immune infiltration of CXCL10 in BLCA. Furthermore, we focused on examining the impact of CXCL10 on immune therapy in BLCA. Additionally, we validated the expression of CXCL10 in various BLCA cell lines using PCR techniques. RESULTS We observed an upregulation of CXCL10 in BLCA tissues as well as in different cell lines. Additionally, upregulation of CXCL10 indicates a better prognosis for BLCA patients. ESTIMATE and CIBERSORT algorithms suggest that CXCL10 is closely associated with the immune microenvironment of BLCA. Through multiple immune therapy cohorts, we also identified that CXCL10 has shown promising predictive value for assessing the efficacy of immune therapy in in BLCA. CONCLUSION Our study indicates that CXCL10 has the potential to serve as a favorable prognostic factor and is strongly associated with immune infiltration in BLCA.
Collapse
Affiliation(s)
- Tao Yin
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Shenzhen University Medical College, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Shuanzhu Mou
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Shenzhen University Medical College, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Haiyu Zhang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Ying Dong
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Shenzhen University Medical College, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Bing Yan
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Shenzhen University Medical College, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Weisheng Huang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yuhan Liu
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China.
- Shenzhen University Medical College, Shenzhen, China.
- Shenzhen Second People's Hospital, Clinical Medicine College of Anhui Medical University, Shenzhen, China.
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
| |
Collapse
|
42
|
Paul S, Mukherjee T, Das K. Coagulation Protease-Driven Cancer Immune Evasion: Potential Targets for Cancer Immunotherapy. Cancers (Basel) 2024; 16:1568. [PMID: 38672649 PMCID: PMC11048528 DOI: 10.3390/cancers16081568] [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: 03/13/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Blood coagulation and cancer are intrinsically connected, hypercoagulation-associated thrombotic complications are commonly observed in certain types of cancer, often leading to decreased survival in cancer patients. Apart from the common role in coagulation, coagulation proteases often trigger intracellular signaling in various cancers via the activation of a G protein-coupled receptor superfamily protease: protease-activated receptors (PARs). Although the role of PARs is well-established in the development and progression of certain types of cancer, their impact on cancer immune response is only just emerging. The present review highlights how coagulation protease-driven PAR signaling plays a key role in modulating innate and adaptive immune responses. This is followed by a detailed discussion on the contribution of coagulation protease-induced signaling in cancer immune evasion, thereby supporting the growth and development of certain tumors. A special section of the review demonstrates the role of coagulation proteases, thrombin, factor VIIa, and factor Xa in cancer immune evasion. Targeting coagulation protease-induced signaling might be a potential therapeutic strategy to boost the immune surveillance mechanism of a host fighting against cancer, thereby augmenting the clinical consequences of targeted immunotherapeutic regimens.
Collapse
Affiliation(s)
- Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, West Bengal, India;
| | - Tanmoy Mukherjee
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani 741251, West Bengal, India
| |
Collapse
|
43
|
Lim RJ, Salehi-Rad R, Tran LM, Oh MS, Dumitras C, Crosson WP, Li R, Patel TS, Man S, Yean CE, Abascal J, Huang Z, Ong SL, Krysan K, Dubinett SM, Liu B. CXCL9/10-engineered dendritic cells promote T cell activation and enhance immune checkpoint blockade for lung cancer. Cell Rep Med 2024; 5:101479. [PMID: 38518770 PMCID: PMC11031384 DOI: 10.1016/j.xcrm.2024.101479] [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: 08/11/2023] [Revised: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
Immune checkpoint blockade (ICB) with PD-1/PD-L1 inhibition has revolutionized the treatment of non-small cell lung cancer (NSCLC). Durable responses, however, are observed only in a subpopulation of patients. Defective antigen presentation and an immunosuppressive tumor microenvironment (TME) can lead to deficient T cell recruitment and ICB resistance. We evaluate intratumoral (IT) vaccination with CXCL9- and CXCL10-engineered dendritic cells (CXCL9/10-DC) as a strategy to overcome resistance. IT CXCL9/10-DC leads to enhanced T cell infiltration and activation in the TME and tumor inhibition in murine NSCLC models. The antitumor efficacy of IT CXCL9/10-DC is dependent on CD4+ and CD8+ T cells, as well as CXCR3-dependent T cell trafficking from the lymph node. IT CXCL9/10-DC, in combination with ICB, overcomes resistance and establishes systemic tumor-specific immunity in murine models. These studies provide a mechanistic understanding of CXCL9/10-DC-mediated host immune activation and support clinical translation of IT CXCL9/10-DC to augment ICB efficacy in NSCLC.
Collapse
Affiliation(s)
- Raymond J Lim
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ramin Salehi-Rad
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Linh M Tran
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Michael S Oh
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Camelia Dumitras
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - William P Crosson
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rui Li
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tejas S Patel
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samantha Man
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cara E Yean
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jensen Abascal
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - ZiLing Huang
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephanie L Ong
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kostyantyn Krysan
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Bin Liu
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Miao SN, Chai MQ, Liu XY, Wei CY, Zhang CC, Sun NN, Fei QZ, Peng LL, Qiu H. Exercise accelerates recruitment of CD8 + T cell to promotes anti-tumor immunity in lung cancer via epinephrine. BMC Cancer 2024; 24:474. [PMID: 38622609 PMCID: PMC11021002 DOI: 10.1186/s12885-024-12224-7] [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: 01/06/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND AND PURPOSE In recent years, there has been extensive research on the role of exercise as an adjunctive therapy for cancer. However, the potential mechanisms underlying the anti-tumor therapy of exercise in lung cancer remain to be fully elucidated. As such, our study aims to confirm whether exercise-induced elevation of epinephrine can accelerate CD8+ T cell recruitment through modulation of chemokines and thus ultimately inhibit tumor progression. METHOD C57BL/6 mice were subcutaneously inoculated with Lewis lung cancer cells (LLCs) to establish a subcutaneous tumor model. The tumor mice were randomly divided into different groups to performed a moderate-intensity exercise program on a treadmill for 5 consecutive days a week, 45 min a day. The blood samples and tumor tissues were collected after exercise for IHC, RT-qPCR, ELISA and Western blot. In addition, another group of mice received daily epinephrine treatment for two weeks (0.05 mg/mL, 200 µL i.p.) (EPI, n = 8) to replicate the effects of exercise on tumors in vivo. Lewis lung cancer cells were treated with different concentrations of epinephrine (0, 5, 10, 20 µM) to detect the effect of epinephrine on chemokine levels via ELISA and RT-qPCR. RESULTS This study reveals that both pre- and post-cancer exercise effectively impede the tumor progression. Exercise led to an increase in EPI levels and the infiltration of CD8+ T cell into the lung tumor. Exercise-induced elevation of EPI is involved in the regulation of Ccl5 and Cxcl10 levels further leading to enhanced CD8+ T cell infiltration and ultimately inhibiting tumor progression. CONCLUSION Exercise training enhance the anti-tumor immunity of lung cancer individuals. These findings will provide valuable insights for the future application of exercise therapy in clinical practice.
Collapse
Affiliation(s)
- Sai-Nan Miao
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Meng-Qi Chai
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Xiang-Yu Liu
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Cheng-Yu Wei
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Cun-Cun Zhang
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Ning-Ning Sun
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Qing-Ze Fei
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Lin-Lin Peng
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Huan Qiu
- School of Nursing, Anhui Medical University, 230032, Hefei, China.
| |
Collapse
|
45
|
Philip DT, Goins NM, Catanzaro NJ, Misumi I, Whitmire JK, Atkins HM, Lazear HM. Interferon lambda restricts herpes simplex virus skin disease by suppressing neutrophil-mediated pathology. mBio 2024; 15:e0262323. [PMID: 38426749 PMCID: PMC11005406 DOI: 10.1128/mbio.02623-23] [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: 09/25/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Type III interferons (IFN-λ) are antiviral and immunomodulatory cytokines that have been best characterized in respiratory and gastrointestinal infections, but the effects of IFN-λ against skin infections have not been extensively investigated. We sought to define the skin-specific effects of IFN-λ against the highly prevalent human pathogen, herpes simplex virus (HSV). We infected mice lacking the IFN-λ receptor (Ifnlr1-/-), both the IFN-λ and the IFN-α/β receptors (Ifnar1-/-Ifnlr1-/-), or IFN-λ cytokines (Ifnl2/3-/-) and found that IFN-λ restricts the severity of HSV-1 and HSV-2 skin lesions without affecting viral loads. We used RNAseq to define IFN-λ- and IFN-β-induced transcriptional responses in primary mouse keratinocytes. Using conditional knockout mice, we found that IFN-λ signaling in both keratinocytes and neutrophils was necessary to control HSV-1 skin lesion severity and that IFN-λ signaling in keratinocytes suppressed CXCL9-mediated neutrophil recruitment to the skin. Furthermore, depleting neutrophils or blocking CXCL9 protected against severe HSV-1 skin lesions in Ifnlr1-/- mice. Altogether, our results suggest that IFN-λ plays an immunomodulatory role in the skin that restricts neutrophil-mediated pathology during HSV infection and suggests potential applications for IFN-λ in treating viral skin infections.IMPORTANCEType III interferons (IFN-λ) have been shown to have antiviral and immunomodulatory effects at epithelial barriers such as the respiratory and gastrointestinal tracts, but their effects on the skin have not been extensively investigated. We used mice lacking IFN-λ signaling to investigate the skin-specific effects of IFN-λ against the herpes simplex virus (HSV), which targets epithelial tissues to cause cold sores and genital herpes. We found that IFN-λ limited the severity of HSV skin lesions without affecting viral load and that this protective effect required IFN-λ signaling in both keratinocytes and neutrophils. We found that IFN-λ signaling in keratinocytes suppressed neutrophil recruitment to the skin and that depleting neutrophils protected against severe HSV skin lesions in the absence of IFN-λ. Altogether, our results suggest that IFN-λ plays an immunomodulatory role in the skin that restricts neutrophil-mediated pathology during HSV infection and suggests potential applications for IFN-λ in treating viral skin infections.
Collapse
Affiliation(s)
- Drake T. Philip
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel M. Goins
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nicholas J. Catanzaro
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ichiro Misumi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jason K. Whitmire
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hannah M. Atkins
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Helen M. Lazear
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
46
|
Gudd CLC, Mitchell E, Atkinson SR, Mawhin MA, Turajlic S, Larkin J, Thursz MR, Goldin RD, Powell N, Antoniades CG, Woollard KJ, Possamai LA, Triantafyllou E. Therapeutic inhibition of monocyte recruitment prevents checkpoint inhibitor-induced hepatitis. J Immunother Cancer 2024; 12:e008078. [PMID: 38580334 PMCID: PMC11002390 DOI: 10.1136/jitc-2023-008078] [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] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Checkpoint inhibitor-induced hepatitis (CPI-hepatitis) is an emerging problem with the widening use of CPIs in cancer immunotherapy. Here, we developed a mouse model to characterize the mechanism of CPI-hepatitis and to therapeutically target key pathways driving this pathology. METHODS C57BL/6 wild-type (WT) mice were dosed with toll-like receptor (TLR)9 agonist (TLR9-L) for hepatic priming combined with anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) plus anti-programmed cell death 1 (PD-1) ("CPI") or phosphate buffered saline (PBS) control for up to 7 days. Flow cytometry, histology/immunofluorescence and messenger RNA sequencing were used to characterize liver myeloid/lymphoid subsets and inflammation. Hepatocyte damage was assessed by plasma alanine transaminase (ALT) and cytokeratin-18 (CK-18) measurements. In vivo investigations of CPI-hepatitis were carried out in Rag2-/- and Ccr2rfp/rfp transgenic mice, as well as following anti-CD4, anti-CD8 or cenicriviroc (CVC; CCR2/CCR5 antagonist) treatment. RESULTS Co-administration of combination CPIs with TLR9-L induced liver pathology closely resembling human disease, with increased infiltration and clustering of granzyme B+perforin+CD8+ T cells and CCR2+ monocytes, 7 days post treatment. This was accompanied by apoptotic hepatocytes surrounding these clusters and elevated ALT and CK-18 plasma levels. Liver RNA sequencing identified key signaling pathways (JAK-STAT, NF-ΚB) and cytokine/chemokine networks (Ifnγ, Cxcl9, Ccl2/Ccr2) as drivers of CPI-hepatitis. Using this model, we show that CD8+ T cells mediate hepatocyte damage in experimental CPI-hepatitis. However, their liver recruitment, clustering, and cytotoxic activity is dependent on the presence of CCR2+ monocytes. The absence of hepatic monocyte recruitment in Ccr2rfp/rfp mice and CCR2 inhibition by CVC treatment in WT mice was able to prevent the development and reverse established experimental CPI-hepatitis. CONCLUSION This newly established mouse model provides a platform for in vivo mechanistic studies of CPI-hepatitis. Using this model, we demonstrate the central role of liver infiltrating CCR2+ monocyte interaction with tissue-destructive CD8+ T cells in the pathogenesis of CPI-hepatitis and highlight CCR2 inhibition as a novel therapeutic target.
Collapse
Affiliation(s)
- Cathrin L C Gudd
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Eoin Mitchell
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stephen R Atkinson
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Marie-Anne Mawhin
- Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Samra Turajlic
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Units, The Royal Marsden NHS Foundation Trust, London, UK
- Melanoma and Kidney Cancer Team, The Institute of Cancer Research, London, UK
| | - James Larkin
- Renal and Skin Units, The Royal Marsden NHS Foundation Trust, London, UK
- Melanoma and Kidney Cancer Team, The Institute of Cancer Research, London, UK
| | - Mark R Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Robert D Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Nick Powell
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Kevin J Woollard
- Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Lucia A Possamai
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | |
Collapse
|
47
|
Chen J, Amoozgar Z, Liu X, Aoki S, Liu Z, Shin SM, Matsui A, Hernandez A, Pu Z, Halvorsen S, Lei PJ, Datta M, Zhu L, Ruan Z, Shi L, Staiculescu D, Inoue K, Munn LL, Fukumura D, Huang P, Sassi S, Bardeesy N, Ho WJ, Jain RK, Duda DG. Reprogramming the Intrahepatic Cholangiocarcinoma Immune Microenvironment by Chemotherapy and CTLA-4 Blockade Enhances Anti-PD-1 Therapy. Cancer Immunol Res 2024; 12:400-412. [PMID: 38260999 PMCID: PMC10985468 DOI: 10.1158/2326-6066.cir-23-0486] [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: 06/11/2023] [Revised: 11/05/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) has limited therapeutic options and a dismal prognosis. Adding blockade of the anti-programmed cell death protein (PD)-1 pathway to gemcitabine/cisplatin chemotherapy has recently shown efficacy in biliary tract cancers but with low response rates. Here, we studied the effects of anti-cytotoxic T lymphocyte antigen (CTLA)-4 when combined with anti-PD-1 and gemcitabine/cisplatin in orthotopic murine models of ICC. This combination therapy led to substantial survival benefits and reduction of morbidity in two aggressive ICC models that were resistant to immunotherapy alone. Gemcitabine/cisplatin treatment increased tumor-infiltrating lymphocytes and normalized the ICC vessels and, when combined with dual CTLA-4/PD-1 blockade, increased the number of activated CD8+Cxcr3+IFNγ+ T cells. CD8+ T cells were necessary for the therapeutic benefit because the efficacy was compromised when CD8+ T cells were depleted. Expression of Cxcr3 on CD8+ T cells is necessary and sufficient because CD8+ T cells from Cxcr3+/+ but not Cxcr3-/- mice rescued efficacy in T cell‒deficient mice. Finally, rational scheduling of anti-CTLA-4 "priming" with chemotherapy followed by anti-PD-1 therapy achieved equivalent efficacy with reduced overall drug exposure. These data suggest that this combination approach should be clinically tested to overcome resistance to current therapies in ICC patients.
Collapse
Affiliation(s)
- Jiang Chen
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Immuno-oncology Research and Development, Sanofi, Cambridge, Massachusetts
| | - Xin Liu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuichi Aoki
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Tohoku Graduate School of Medicine, Sendai, Japan
| | - Zelong Liu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sarah M. Shin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Aya Matsui
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Kanazawa University Institute of Medical, Pharmaceutical and Health Sciences Faculty of Medicine, Kanazawa, Japan
| | - Alexei Hernandez
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Zhangya Pu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Xiangya Hospital, Central South University, Changsha, China
| | - Stefan Halvorsen
- Center of Computational and Integrative Biology (CCIB), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pin-Ji Lei
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meenal Datta
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Aerospace and Mechanical Engineering, College of Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Lingling Zhu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- West China Hospital of Sichuan University, Chengdu, China
| | - Zhiping Ruan
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Jiaotong University, Xi'an, China
| | - Lei Shi
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Daniel Staiculescu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Koetsu Inoue
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Tohoku Graduate School of Medicine, Sendai, Japan
| | - Lance L. Munn
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dai Fukumura
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peigen Huang
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Slim Sassi
- Center of Computational and Integrative Biology (CCIB), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Orthopedics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nabeel Bardeesy
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dan G. Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
48
|
Yu J, Gadwa J, Ross RB, Knitz M, Darragh LB, Abdelazeem KNM, Beynor J, Neupert B, Nguyen A, Nguyen D, Olimpo N, Corbo S, Van Court B, D'Alessandro A, Saviola A, Karam SD. IL7 in combination with radiotherapy stimulates a memory T-cell response to improve outcomes in HNSCC models. Cancer Immunol Immunother 2024; 73:90. [PMID: 38554147 PMCID: PMC10981637 DOI: 10.1007/s00262-024-03664-y] [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: 01/08/2024] [Accepted: 02/27/2024] [Indexed: 04/01/2024]
Abstract
Clinically approved head and neck squamous cell carcinoma (HNSCC) immunotherapies manipulate the immune checkpoint blockade (ICB) axis but have had limited success outside of recurrent/metastatic disease. Interleukin-7 (IL7) has been shown to be essential for effector T-cell survival, activation, and proliferation. Here, we show that IL7 in combination with radiotherapy (RT) is effective in activating CD8 + T-cells for reducing tumor growth. Our studies were conducted using both human papillomavirus related and unrelated orthotopic HNSCC murine models. Immune populations from the tumor, draining lymph nodes, and blood were compared between treatment groups and controls using flow cytometry, proteomics, immunofluorescence staining, and RNA sequencing. Treatment with RT and IL7 (RT + IL7) resulted in significant tumor growth reduction, high CD8 T-cell tumor infiltration, and increased proliferation of T-cell progenitors in the bone marrow. IL7 also expanded a memory-like subpopulation of CD8 T-cells. These results indicate that IL7 in combination with RT can serve as an effective immunotherapy strategy outside of the conventional ICB axis to drive the antitumor activity of CD8 T-cells.
Collapse
Affiliation(s)
- Justin Yu
- Department of Otolaryngology - Head and Neck Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jacob Gadwa
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Richard B Ross
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Michael Knitz
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Laurel B Darragh
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Khalid N M Abdelazeem
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Radiation Biology Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Jessica Beynor
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Brooke Neupert
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Alexander Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Diemmy Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Nicholas Olimpo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sophia Corbo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Anthony Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
49
|
Hung HC, Mao TL, Fan MH, Huang GZ, Minhalina AP, Chen CL, Liu CL. Enhancement of Tumorigenicity, Spheroid Niche, and Drug Resistance of Pancreatic Cancer Cells in Three-Dimensional Culture System. J Cancer 2024; 15:2292-2305. [PMID: 38495500 PMCID: PMC10937281 DOI: 10.7150/jca.87494] [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: 06/26/2023] [Accepted: 01/27/2024] [Indexed: 03/19/2024] Open
Abstract
The three-dimensional (3D) cell culture technique has been applied comprehensively as a variable platform for medical research, biochemical signal pathway analysis, and evaluation of anti-tumor treatment response due to an excellent recapitulation of a tumor microenvironment (TME) in the in vitro cultured cancer cells. Pancreatic cancer (PaC) is one of the toughest malignancies with a complex TME and refractory treatment response. To comprehensively study the TME of PaC, there is an eager need to develop a 3D culture model to decompose the cellular components and their cross interactions. Herein, we establish a 3D PaC culture system with cancer stem cell (CSC) and scalability properties. To validate our model, we tested the individual PaC cell and the combined effects with cancer-associated fibroblasts (CAFs) on cancer tumorigenicity, the cellular interaction through the CXCR3/CXCL10 axis, and cellular responses reflection of anti-cancer treatments. With the help of our 3D technology, a simulated malignant spheroid with important stromal populations and TME physiochemical properties may be successfully recreated. It can be used in a wide range of preclinical research and helpful in advancing basic and translational cancer biology.
Collapse
Affiliation(s)
- Hao-Chien Hung
- Department of General Surgery, Chang-Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Tsui-Lien Mao
- Department of Pathology, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Ming-Huei Fan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Guan-Zhi Huang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ainani Priza Minhalina
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Chao-Lien Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
50
|
Lyu M, Xu G, Zhou J, Reboud J, Wang Y, Lai H, Chen Y, Zhou Y, Zhu G, Cooper JM, Ying B. Single-Cell Sequencing Reveals Functional Alterations in Tuberculosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305592. [PMID: 38192178 PMCID: PMC10953544 DOI: 10.1002/advs.202305592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/21/2023] [Indexed: 01/10/2024]
Abstract
Despite its importance, the functional heterogeneity surrounding the dynamics of interactions between mycobacterium tuberculosis and human immune cells in determining host immune strength and tuberculosis (TB) outcomes, remains far from understood. This work now describes the development of a new technological platform to elucidate the immune function differences in individuals with TB, integrating single-cell RNA sequencing and cell surface antibody sequencing to provide both genomic and phenotypic information from the same samples. Single-cell analysis of 23 990 peripheral blood mononuclear cells from a new cohort of primary TB patients and healthy controls enables to not only show four distinct immune phenotypes (TB, myeloid, and natural killer (NK) cells), but also determine the dynamic changes in cell population abundance, gene expression, developmental trajectory, transcriptomic regulation, and cell-cell signaling. In doing so, TB-related changes in immune cell functions demonstrate that the immune response is mediated through host T cells, myeloid cells, and NK cells, with TB patients showing decreased naive, cytotoxicity, and memory functions of T cells, rather than their immunoregulatory function. The platform also has the potential to identify new targets for immunotherapeutic treatment strategies to restore T cells from dysfunctional or exhausted states.
Collapse
Affiliation(s)
- Mengyuan Lyu
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Gaolian Xu
- School of Biomedical Engineering/Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jian Zhou
- Department of Thoracic SurgeryWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Julien Reboud
- Division of Biomedical EngineeringUniversity of GlasgowGlasgowG12 8LTUnited Kingdom
| | - Yili Wang
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Hongli Lai
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Yi Chen
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Yanbing Zhou
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Guiying Zhu
- School of Biomedical Engineering/Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jonathan M. Cooper
- Division of Biomedical EngineeringUniversity of GlasgowGlasgowG12 8LTUnited Kingdom
| | - Binwu Ying
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| |
Collapse
|