1
|
Li Z, Zhao Y, Huang P, Wu Z, Ouyang D, Nyarko AO, Ai L, Zhang Z, Chang S. Pan-cancer analysis of the immunological and oncogenic roles of ATAD2 with verification in papillary thyroid carcinoma. Sci Rep 2024; 14:22263. [PMID: 39333272 PMCID: PMC11436736 DOI: 10.1038/s41598-024-73274-2] [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: 04/22/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
ATAD2 (ATPase Family AAA Domain-Containing 2) is highly expressed across varies tumor types, yet its common roles in tumor progression and immune interaction remain unclear. We analyzed the expression and alteration profiles of ATAD2, along with its diagnostic and prognostic role in pan-cancer, utilizing TCGA, GTEx, CPTAC, HPA, and cBioPortal databases. Furthermore, we examined the relationship between ATAD2 and immune infiltration utilizing single-cell sequencing data and TCGA database. Additionally, the expression and oncogenic functions of ATAD2 were verified in papillary thyroid carcinoma (PTC) through MTT, wound-healing, transwell, and flow cytometry assays. Our results revealed significant overexpression of ATAD2 in most cancers, strongly associated with poor prognosis. Amplification was the most frequent alteration type of ATAD2, with its mutation correlating with improved overall survival. ATAD2 was positively correlated with multiple inhibitory immune checkpoints and closely associated with the immunosuppressive microenvironment, particularly in PTC. In vitro experiments demonstrated that ATAD2 promoted the proliferation, migration, and invasion of PTC cells by activating the PI3K-AKT pathway and modulating the G1/S cell cycle checkpoint. Collectively, ATAD2 holds promise as a biomarker for pan-cancer diagnosis and prognosis, as well as a predictor of immunotherapeutic responsiveness and a therapeutic target to enhance the efficacy of existing anti-tumor immune therapies.
Collapse
Affiliation(s)
- Zhecheng Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yaxin Zhao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Peng Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhaoyi Wu
- Department of Thyroid and Breast Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Human Normal University, Changsha, 410008, China
| | - Dengjie Ouyang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Adolphus Osei Nyarko
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Lei Ai
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhejia Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
2
|
Barbagallo D, Ponti D, Bassani B, Bruno A, Pulze L, Akkihal SA, George-William JN, Gundamaraju R, Campomenosi P. MiR-223-3p in Cancer Development and Cancer Drug Resistance: Same Coin, Different Faces. Int J Mol Sci 2024; 25:8191. [PMID: 39125761 PMCID: PMC11311375 DOI: 10.3390/ijms25158191] [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: 06/06/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
MicroRNAs (miRNAs) are mighty post-transcriptional regulators in cell physiology and pathophysiology. In this review, we focus on the role of miR-223-3p (henceforth miR-223) in various cancer types. MiR-223 has established roles in hematopoiesis, inflammation, and most cancers, where it can act as either an oncogenic or oncosuppressive miRNA, depending on specific molecular landscapes. MiR-223 has also been linked to either the sensitivity or resistance of cancer cells to treatments in a context-dependent way. Through this detailed review, we highlight that for some cancers (i.e., breast, non-small cell lung carcinoma, and glioblastoma), the oncosuppressive role of miR-223 is consistently reported in the literature, while for others (i.e., colorectal, ovarian, and pancreatic cancers, and acute lymphocytic leukemia), an oncogenic role prevails. In prostate cancer and other hematological malignancies, although an oncosuppressive role is frequently described, there is less of a consensus. Intriguingly, NLRP3 and FBXW7 are consistently identified as miR-223 targets when the miRNA acts as an oncosuppressor or an oncogene, respectively, in different cancers. Our review also describes that miR-223 was increased in biological fluids or their extracellular vesicles in most of the cancers analyzed, as compared to healthy or lower-risk conditions, confirming the potential application of this miRNA as a diagnostic and prognostic biomarker in the clinic.
Collapse
Affiliation(s)
- Davide Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics “Giovanni Sichel”, University of Catania, Via Santa Sofia 89, 95123 Catania, Italy
- Interdisciplinary Research Centre on the Diagnosis and Therapy of Brain Tumors, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Donatella Ponti
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Via Fantoli 16/15, 20138 Milano, Italy; (B.B.); (A.B.)
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Via Fantoli 16/15, 20138 Milano, Italy; (B.B.); (A.B.)
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| | - Laura Pulze
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| | - Shreya A. Akkihal
- Independent Researcher, 35004 SE Swenson St, Snoqualmie, WA 98065, USA;
| | - Jonahunnatha N. George-William
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi, 93, 20054 Segrate, Italy;
| | - Rohit Gundamaraju
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA;
- ER Stress and Mucosal Immunology Team, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia
| | - Paola Campomenosi
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| |
Collapse
|
3
|
Zhu X, Qiu Z, Yang M, Yang J, Kong L, Li L, Huang Y, Xie L. Causal associations of BAFF-R on IgD+ CD24- B cell immune cell trait with hepatocellular carcinoma and the mediating role of phenylacetylglutamate levels: a Mendelian randomization study. J Cancer 2024; 15:4591-4603. [PMID: 39006080 PMCID: PMC11242345 DOI: 10.7150/jca.96059] [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: 03/07/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
We conducted a bi-directional two-sample Mendelian randomization (MR) analysis to investigate the causal associations between immune cell traits and hepatocellular carcinoma (HCC) and identified the mediating factor of metabolites. The exposure factors were immune cell traits, the mediators were metabolites, and the outcome variable was HCC. Inverse-variance weighted method (IVW) was the main method. Weighted median, MR-Egger regression, weighted mode, simple mode, and MR pleiotropy residual sum and outlier (MRPRESSO) methods were used as complementary methods. The results were tested by using the Bayesian weighted Mendelian randomization (BWMR) approach in our MR study. Subsequently, the potential mediating effect was investigated by conducting a two-step mediation analysis. We identified 26 traits with suggestive correlations between immune cell traits and HCC, with 4 immune cell traits among them having causal correlations with HCC. There were no causal correlations between HCC and immune cell traits in the reverse MR analysis. In the mediation analysis, we found a positive causal association between B cell-activating factor receptors (BAFF-R) on IgD+ CD24- B cell and HCC [IVW: odd ratio (OR), 0.845; 95% CI, 0.759-0.942; p = 0.002]. Phenylacetylglutamate (PAG) levels mediated 7.353% of the causal pathway from BAFF-R on IgD+ CD24- B cell and HCC. In conclusion, BAFF-R on IgD+ CD24- B cell lowers risk of HCC, with PAG levels playing a mediating role.
Collapse
Affiliation(s)
- Xuan Zhu
- Department of Medical Laboratory, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guanxi, China
| | - Zongchao Qiu
- Fujian Yuanshizhuling Community Health Service center, Jiangnan district, Nanning 530031, Guanxi, China
| | - Maochun Yang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guanxi, China
| | - Jiali Yang
- Department of Occupational and Environmental Health, Beihai Center for Diseases Prevention and Control (Beihai Institute of Health Supervisio), Beihai, 536000, Guanxi, China
| | - Lingxi Kong
- Department of Medical Laboratory, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guanxi, China
| | - Limin Li
- Department of Medical Laboratory, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guanxi, China
| | - Yingting Huang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guanxi, China
| | - Li Xie
- Department of Medical Laboratory, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guanxi, China
| |
Collapse
|
4
|
Hansen FJ, David P, Weber GF. The Multifaceted Functionality of Plasmacytoid Dendritic Cells in Gastrointestinal Cancers: A Potential Therapeutic Target? Cancers (Basel) 2024; 16:2216. [PMID: 38927922 PMCID: PMC11201847 DOI: 10.3390/cancers16122216] [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: 05/08/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Gastrointestinal (GI) tumors pose a significant global health burden, necessitating the exploration of novel therapeutic approaches. Plasmacytoid dendritic cells (pDCs) play a crucial role in tumor immunity, exhibiting both anti-tumor and pro-tumor effects. This review aims to summarize the role of pDCs in different types of GI tumors and assess their potential as therapeutic targets. In gastric cancer, hepatocellular carcinoma, and intrahepatic cholangiocarcinoma, increased infiltration of pDCs was associated with a worse outcome, whereas in esophageal cancer, pancreatic cancer, and colorectal cancer, pDC infiltration improved the outcome. Initial animal studies of gastric cancer and hepatocellular carcinoma showed that pDCs could be a successful therapeutic target. In conclusion, pDCs play a multifaceted role in GI tumors, influencing both anti-tumor immunity and tumor progression. Further research is needed to optimize their clinical application and explore combinatorial approaches.
Collapse
Affiliation(s)
| | - Paul David
- Department of General and Visceral Surgery, Medical Faculty of Friedrich-Alexander-University Erlangen, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Georg F. Weber
- Department of General and Visceral Surgery, Medical Faculty of Friedrich-Alexander-University Erlangen, University Hospital Erlangen, 91054 Erlangen, Germany;
| |
Collapse
|
5
|
Lu Z, Wang Z, Li G. High expression of CCNB2 is an independent predictive poor prognostic biomarker and correlates with immune infiltrates in breast carcinoma. Heliyon 2024; 10:e31586. [PMID: 38831807 PMCID: PMC11145498 DOI: 10.1016/j.heliyon.2024.e31586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
Background Cyclin B2 (CCNB2) is associated with cell cycle progression, acting as a cell cycle checkpoint in progression of G2/M transition. In many cancer patients, it has been observed that overexpression of CCNB2 enhances tumor invasiveness and leads to adverse prognosis. However, the association of CCNB2 with the tumor microenvironment remains unclear. Therefore, it is necessary to clarify the associations of CCNB2 with the immune status and prognosis of breast carcinoma (BRCA). Methods Gene expression and clinical data for BRCA were obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases, followed by association analyses of CCNB2 expression with prognosis, immune cell infiltration, and immune checkpoints. This study further performed drug sensitivity analysis and constructed a prognostic nomogram for CCNB2. Results 3619 differentially expressed genes were identified in BRCA, including CCNB2 that emerged as a key gene in the network. High CCNB2 expression correlated with poor prognosis. Functional analysis demonstrated enrichment of CCNB2 co-expressed genes with the cell cycle, cancer progression, cell energy, and immune pathways. Microsatellite instability and tumor mutation burden analyses indicated CCNB2 as a candidate immunotherapy target. Tumor-infiltrating myeloid-derived suppressor cells, regulatory T cells, and T helper 2 cells were associated with CCNB2-related tumor progression and metastasis. CCNB2 expression positively correlated with immune checkpoints, indicating that high CCNB2 expression might facilitate tumor immune escape. Tumors with high CCNB2 expression showed sensitivity to phosphoinositide 3-kinase-protein kinase B-mammalian target of rapamycin and cyclin-dependent kinase (CDK) 4/6 inhibitors, and the nomogram had good prognostic predictive ability for patients with BRCA. Conclusions CCNB2 may play a crucial role in tumorigenesis and serve as an independent prognostic biomarker associated with tumor microenvironment, tumor immune infiltration and immunotherapy in BRCA.
Collapse
Affiliation(s)
- Zonghong Lu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhihong Wang
- Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, GI Cancer Research Institute, Wuhan, Hubei, 430030, China
| | - Guodong Li
- Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, GI Cancer Research Institute, Wuhan, Hubei, 430030, China
| |
Collapse
|
6
|
Xie Z, Qin Y, Chen X, Yang S, Yang J, Gui L, Liu P, He X, Zhou S, Zhang C, Tang L, Shi Y. Deciphering the Prognostic Significance of MYD88 and CD79B Mutations in Diffuse Large B-Cell Lymphoma: Insights into Treatment Outcomes. Target Oncol 2024; 19:383-400. [PMID: 38643457 DOI: 10.1007/s11523-024-01057-w] [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] [Accepted: 03/15/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND The clinical and genetic characteristics, as well as treatment outcomes, of diffuse large B-cell lymphoma (DLBCL) patients with different MYD88 and CD79B mutation status merit further investigation. OBJECTIVE This study aims to investigate the distinctions in clinical manifestations, genetic characteristics, and treatment outcomes among MYD88-CD79Bco-mut, MYD88/CD79Bsingle-mut, and MYD88-CD79Bco-wt DLBCL patients. PATIENTS AND METHODS Clinical and genetic characteristics, along with treatment outcomes among 2696 DLBCL patients bearing MYD88-CD79Bco-mut, MYD88/CD79Bsingle-mut, and MYD88-CD79Bco-wt treated with R-CHOP/R-CHOP-like regimens from the Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College and six external cohorts were analyzed. Potential molecular mechanisms were investigated through Gene Set Enrichment Analysis and xCell methodology. RESULTS In the MCD subtype, patients with MYD88-CD79Bco-mut showed comparable progression-free survival (PFS) and overall survival (OS) compared to MYD88/CD79Bsingle-mut or MYD88-CD79Bco-wt. However, in the non-MCD subtype, patients with MYD88-CD79Bco-mut exhibited significantly inferior OS than MYD88/CD79Bsingle-mut or MYD88-CD79Bco-wt, while there was no significant OS difference between MYD88/CD79Bsingle-mut and MYD88-CD79Bco-wt (median OS: 68.8 [95% CI 22-NA] vs NA [95% CI 112-NA] vs 177.7 [95% CI 159-NA] months; MYD88-CD79Bco-mut vs MYD88/CD79Bsingle-mut: p = 0.02; MYD88-CD79Bco-mut vs MYD88-CD79Bco-wt: p = 0.03; MYD88/CD79Bsingle-mut vs MYD88-CD79Bco-wt: p = 0.33). Regarding patients with MYD88-CD79Bco-mut, there was no significant difference in PFS and OS between the MCD and non-MCD subtypes. Within the MYD88-CD79Bco-mut group, patients with PIM1mut had better PFS than PIM1wt (median PFS: 8.34 [95% CI 5.56-NA] vs 43.8 [95% CI 26.4-NA] months; p = 0.02). Possible mechanisms contributing to the superior PFS of PIM1mut patients may include activated lymphocyte-mediated immunity and interferon response, a higher proportion of natural killer T cells and plasmacytoid dendritic cells, as well as suppressed angiogenesis and epithelial-mesenchymal transition, along with lower fibroblast and stromal score. CONCLUSIONS In the MCD subtype, patients with MYD88-CD79Bco-mut showed comparable PFS and OS compared to MYD88/CD79Bsingle-mut or MYD88-CD79Bco-wt, while in the non-MCD subtype, they exhibited significantly inferior OS. There was no significant disparity in PFS and OS of MYD88-CD79Bco-mut between the MCD and non-MCD subtypes. The presence of PIM1mut within the MYD88-CD79Bco-mut group correlated with better PFS, which may result from an intricate interplay of immune processes and tumor microenvironment alterations.
Collapse
Affiliation(s)
- Zucheng Xie
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Yan Qin
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xinrui Chen
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Sheng Yang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Jianliang Yang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Lin Gui
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Peng Liu
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Xiaohui He
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Shengyu Zhou
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Changgong Zhang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
7
|
Motallebzadeh Khanmiri J, Alizadeh M, Esmaeili S, Gholami Z, Safarzadeh A, Khani-Eshratabadi M, Baghbanzadeh A, Alizadeh N, Baradaran B. Dendritic cell vaccination strategy for the treatment of acute myeloid leukemia: a systematic review. Cytotherapy 2024; 26:427-435. [PMID: 38483358 DOI: 10.1016/j.jcyt.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND AIMS Acute myeloid leukemia (AML) is classified as a hematologic malignancy characterized by the proliferation of immature blood cells within the bone marrow (BM), resulting in an aberrant and unregulated cellular growth. The primary therapeutic modalities for AML include chemotherapy and hematopoietic stem cell transplantation. However, it is important to note that these treatments are accompanied by important adverse effects and mortality rates. Therefore, the need for more effective treatment options seems necessary, and dendritic cell (DC) vaccine therapy can be one of these options. In this study, we aim to investigate the effectiveness of DC vaccination therapy for the management of AML. METHODS PubMed, Scopus, ProQuest, Web of Science, and Google Scholar databases were searched for this systematic review. The articles were evaluated based on the inclusion criteria of this study and initially compared in terms of titles or abstracts. Finally, the articles related to the topic of this review were obtained in full text. The complete remission and partial remission, survival, correlative immune assays, and health-related metrics were used to evaluate this cellular immunotherapy effectiveness. The quality of the studies was assessed independently using the Cochrane risk-of-bias tools. The compiled data were input into a standard Excel spreadsheet. Each domain was evaluated as having either a "low risk," "high risk," or "unclear risk" of bias. RESULTS Among the 3986 studies that were determined, a total of 11 correlated trials were selected for inclusion in this systematic review. DC vaccine therapy was effective in inducing complete and partial remission, and stabilization of the disease. Additionally, it was discovered that the treatment strengthened the immune system as seen by increased levels of CD4+ and CD8+ T cells, Th1 cytokines, WT1-specific T cells, and activated NK cells. CONCLUSION We conducted a systematic review that supports the use of DC vaccine therapy as an effective treatment for AML. The therapy demonstrated potentials in achieving remission, enhancing the immune system function, and increasing overall survival. However, more studies are required to improve the methods of preparing and delivering the DC vaccine, and to confirm its long-term safety and effectiveness.
Collapse
Affiliation(s)
- Jamal Motallebzadeh Khanmiri
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Esmaeili
- Student Research Committee, Shahed University, Tehran, Iran
| | - Zeinab Gholami
- Faculty of Medicine, University of Medical Sciences, Tabriz, Iran
| | - Ali Safarzadeh
- Department of Biology, University of Padova, Padova, Italy
| | | | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Palomares F, Pina A, Dakhaoui H, Leiva-Castro C, Munera-Rodriguez AM, Cejudo-Guillen M, Granados B, Alba G, Santa-Maria C, Sobrino F, Lopez-Enriquez S. Dendritic Cells as a Therapeutic Strategy in Acute Myeloid Leukemia: Vaccines. Vaccines (Basel) 2024; 12:165. [PMID: 38400148 PMCID: PMC10891551 DOI: 10.3390/vaccines12020165] [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: 12/16/2023] [Revised: 01/11/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Dendritic cells (DCs) serve as professional antigen-presenting cells (APC) bridging innate and adaptive immunity, playing an essential role in triggering specific cellular and humoral responses against tumor and infectious antigens. Consequently, various DC-based antitumor therapeutic strategies have been developed, particularly vaccines, and have been intensively investigated specifically in the context of acute myeloid leukemia (AML). This hematological malignancy mainly affects the elderly population (those aged over 65), which usually presents a high rate of therapeutic failure and an unfavorable prognosis. In this review, we examine the current state of development and progress of vaccines in AML. The findings evidence the possible administration of DC-based vaccines as an adjuvant treatment in AML following initial therapy. Furthermore, the therapy demonstrates promising outcomes in preventing or delaying tumor relapse and exhibits synergistic effects when combined with other treatments during relapses or disease progression. On the other hand, the remarkable success observed with RNA vaccines for COVID-19, delivered in lipid nanoparticles, has revealed the efficacy and effectiveness of these types of vectors, prompting further exploration and their potential application in AML, as well as other neoplasms, loading them with tumor RNA.
Collapse
Affiliation(s)
- Francisca Palomares
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
- Institute of Biomedicine of Seville (IBiS) HUVR/CSIC/University of Seville, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
| | - Alejandra Pina
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Hala Dakhaoui
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Camila Leiva-Castro
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Ana M. Munera-Rodriguez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Marta Cejudo-Guillen
- Institute of Biomedicine of Seville (IBiS) HUVR/CSIC/University of Seville, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
- Department of Pharmacology, Pediatry, and Radiology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Beatriz Granados
- Distrito Sanitario de Atención Primaria Málaga, Sistema Sanitario Público de Andalucía, 29004 Malaga, Spain;
| | - Gonzalo Alba
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Consuelo Santa-Maria
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Francisco Sobrino
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Soledad Lopez-Enriquez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
- Institute of Biomedicine of Seville (IBiS) HUVR/CSIC/University of Seville, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
| |
Collapse
|
9
|
Monti M, Ferrari G, Grosso V, Missale F, Bugatti M, Cancila V, Zini S, Segala A, La Via L, Consoli F, Orlandi M, Valerio A, Tripodo C, Rossato M, Vermi W. Impaired activation of plasmacytoid dendritic cells via toll-like receptor 7/9 and STING is mediated by melanoma-derived immunosuppressive cytokines and metabolic drift. Front Immunol 2024; 14:1227648. [PMID: 38239354 PMCID: PMC10795195 DOI: 10.3389/fimmu.2023.1227648] [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: 05/23/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Plasmacytoid dendritic cells (pDCs) infiltrate a large set of human cancers. Interferon alpha (IFN-α) produced by pDCs induces growth arrest and apoptosis in tumor cells and modulates innate and adaptive immune cells involved in anti-cancer immunity. Moreover, effector molecules exert tumor cell killing. However, the activation state and clinical relevance of pDCs infiltration in cancer is still largely controversial. In Primary Cutaneous Melanoma (PCM), pDCs density decreases over disease progression and collapses in metastatic melanoma (MM). Moreover, the residual circulating pDC compartment is defective in IFN-α production. Methods The activation of tumor-associated pDCs was evaluated by in silico and microscopic analysis. The expression of human myxovirus resistant protein 1 (MxA), as surrogate of IFN-α production, and proximity ligation assay (PLA) to test dsDNA-cGAS activation were performed on human melanoma biopsies. Moreover, IFN-α and CXCL10 production by in vitro stimulated (i.e. with R848, CpG-A, ADU-S100) pDCs exposed to melanoma cell lines supernatants (SN-mel) was tested by intracellular flow cytometry and ELISA. We also performed a bulk RNA-sequencing on SN-mel-exposed pDCs, resting or stimulated with R848. Glycolytic rate assay was performed on SN-mel-exposed pDCs using the Seahorse XFe24 Extracellular Flux Analyzer. Results Based on a set of microscopic, functional and in silico analyses, we demonstrated that the melanoma milieu directly impairs IFN-α and CXCL10 production by pDCs via TLR-7/9 and cGAS-STING signaling pathways. Melanoma-derived immunosuppressive cytokines and a metabolic drift represent relevant mechanisms enforcing pDC-mediated melanoma escape. Discussion These findings propose a new window of intervention for novel immunotherapy approaches to amplify the antitumor innate immune response in cutaneous melanoma (CM).
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giorgia Ferrari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valentina Grosso
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Nederlands Kanker Instituut, Amsterdam, Netherlands
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Stefania Zini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Agnese Segala
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Consoli
- Oncology Unit, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili di Brescia, Brescia, Italy
| | - Matteo Orlandi
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
10
|
Łazarczyk A, Streb J, Glajcar A, Streb-Smoleń A, Hałubiec P, Wcisło K, Laskowicz Ł, Hodorowicz-Zaniewska D, Szpor J. Dendritic Cell Subpopulations Are Associated with Prognostic Characteristics of Breast Cancer after Neoadjuvant Chemotherapy-An Observational Study. Int J Mol Sci 2023; 24:15817. [PMID: 37958800 PMCID: PMC10648319 DOI: 10.3390/ijms242115817] [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: 10/03/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BC) is the most prevalent malignancy in women and researchers have strived to develop optimal strategies for its diagnosis and management. Neoadjuvant chemotherapy (NAC), which reduces tumor size, risk of metastasis and patient mortality, often also allows for a de-escalation of breast and axillary surgery. Nonetheless, complete pathological response (pCR) is achieved in no more than 40% of patients who underwent NAC. Dendritic cells (DCs) are professional antigen-presenting cells present in the tumor microenvironment. The multitude of their subtypes was shown to be associated with the pathological and clinical characteristics of BC, but it was not evaluated in BC tissue after NAC. We found that highe r densities of CD123+ plasmacytoid DCs (pDCs) were present in tumors that did not show pCR and had a higher residual cancer burden (RCB) score and class. They were of higher stage and grade and more frequently HER2-negative. The density of CD123+ pCDs was an independent predictor of pCR in the studied group. DC-LAMP+ mature DCs (mDCs) were also related to characteristics of clinical relevance (i.e., pCR, RCB, and nuclear grade), although no clear trends were identified. We conclude that CD123+ pDCs are candidates for a novel biomarker of BC response to NAC.
Collapse
Affiliation(s)
- Agnieszka Łazarczyk
- Department of Pathomorphology, Jagiellonian University Medical College, 31-501 Cracow, Poland (J.S.)
| | - Joanna Streb
- Department of Oncology, Jagiellonian University Medical College, 31-501 Cracow, Poland
- University Centre of Breast Disease, University Hospital, 31-501 Cracow, Poland
| | - Anna Glajcar
- Department of Pathomorphology, University Hospital, 30-688 Cracow, Poland
| | - Anna Streb-Smoleń
- Department of Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 31-115 Cracow, Poland
| | - Przemysław Hałubiec
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 31-530 Cracow, Poland
| | - Kacper Wcisło
- Department of Pathomorphology, Jagiellonian University Medical College, 31-501 Cracow, Poland (J.S.)
- Department of Pathomorphology, University Hospital, 30-688 Cracow, Poland
| | - Łukasz Laskowicz
- Clinical Department of Gynecology and Gynecological Oncology, University Hospital, 30-688 Cracow, Poland
| | - Diana Hodorowicz-Zaniewska
- General, Oncological and Gastrointestinal Surgery, Jagiellonian University Medical College, 31-501 Cracow, Poland;
- Department of General Surgery, University Hospital, 31-501 Cracow, Poland
| | - Joanna Szpor
- Department of Pathomorphology, Jagiellonian University Medical College, 31-501 Cracow, Poland (J.S.)
- University Centre of Breast Disease, University Hospital, 31-501 Cracow, Poland
- Department of Pathomorphology, University Hospital, 30-688 Cracow, Poland
| |
Collapse
|
11
|
Gorodilova AV, Kitaeva KV, Filin IY, Mayasin YP, Kharisova CB, Issa SS, Solovyeva VV, Rizvanov AA. The Potential of Dendritic Cell Subsets in the Development of Personalized Immunotherapy for Cancer Treatment. Curr Issues Mol Biol 2023; 45:8053-8070. [PMID: 37886952 PMCID: PMC10605421 DOI: 10.3390/cimb45100509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023] Open
Abstract
Since the discovery of dendritic cells (DCs) in 1973 by Ralph Steinman, a tremendous amount of knowledge regarding these innate immunity cells has been accumulating. Their role in regulating both innate and adaptive immune processes is gradually being uncovered. DCs are proficient antigen-presenting cells capable of activating naive T-lymphocytes to initiate and generate effective anti-tumor responses. Although DC-based immunotherapy has not yielded significant results, the substantial number of ongoing clinical trials underscores the relevance of DC vaccines, particularly as adjunctive therapy or in combination with other treatment options. This review presents an overview of current knowledge regarding human DCs, their classification, and the functions of distinct DC populations. The stepwise process of developing therapeutic DC vaccines to treat oncological diseases is discussed, along with speculation on the potential of combined therapy approaches and the role of DC vaccines in modern immunotherapy.
Collapse
Affiliation(s)
- Anna Valerevna Gorodilova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Kristina Viktorovna Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Ivan Yurevich Filin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Yuri Pavlovich Mayasin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Chulpan Bulatovna Kharisova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Shaza S. Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Valeriya Vladimirovna Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Albert Anatolyevich Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| |
Collapse
|
12
|
Luo S, Chen J, Xu F, Chen H, Li Y, Li W. Dendritic Cell-Derived Exosomes in Cancer Immunotherapy. Pharmaceutics 2023; 15:2070. [PMID: 37631284 PMCID: PMC10457773 DOI: 10.3390/pharmaceutics15082070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Exosomes are nanoscale vesicles released by diverse types of cells for complex intercellular communication. Numerous studies have shown that exosomes can regulate the body's immune response to tumor cells and interfere with the tumor microenvironment (TME). In clinical trials on dendritic cell (DC)-based antitumor vaccines, no satisfactory results have been achieved. However, recent studies suggested that DC-derived exosomes (DEXs) may be superior to DC-based antitumor vaccines in avoiding tumor cell-mediated immunosuppression. DEXs contain multiple DC-derived surface markers that capture tumor-associated antigens (TAAs) and promote immune cell-dependent tumor rejection. These findings indicate the necessity of the further development and improvement of DEX-based cell-free vaccines to complement chemotherapy, radiotherapy, and other immunotherapies. In this review, we highlighted the recent progress of DEXs in cancer immunotherapy, particularly by concentrating on landmark studies and the biological characterization of DEXs, and we summarized their important role in the tumor immune microenvironment (TIME) and clinical application in targeted cancer immunotherapy. This review could enhance comprehension of advances in cancer immunotherapy and contribute to the elucidation of how DEXs regulate the TIME, thereby providing a reference for utilizing DEX-based vaccines in clinical practice.
Collapse
Affiliation(s)
- Shumin Luo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Jing Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Fang Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Huan Chen
- Integrated Chinese and Western Medicine Center, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China;
| | - Yiru Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| | - Weihua Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China; (S.L.); (J.C.); (F.X.); (Y.L.)
| |
Collapse
|
13
|
He T, Muhetaer M, Wu J, Wan J, Hu Y, Zhang T, Wang Y, Wang Q, Cai H, Lu Z. Immune Cell Infiltration Analysis Based on Bioinformatics Reveals Novel Biomarkers of Coronary Artery Disease. J Inflamm Res 2023; 16:3169-3184. [PMID: 37525634 PMCID: PMC10387251 DOI: 10.2147/jir.s416329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/08/2023] [Indexed: 08/02/2023] Open
Abstract
Background Coronary artery disease (CAD) is a multifactorial immune disease, but research into the specific immune mechanism is still needed. The present study aimed to identify novel immune-related markers of CAD. Methods Three CAD-related datasets (GSE12288, GSE98583, GSE113079) were downloaded from the Gene Expression Integrated Database. Gene ontology annotation, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and weighted gene co-expression network analysis were performed on the common significantly differentially expressed genes (DEGs) of these three data sets, and the most relevant module genes for CAD obtained. The immune cell infiltration of module genes was evaluated with the CIBERSORT algorithm, and characteristic genes accompanied by their diagnostic effectiveness were screened by the machine-learning algorithm least absolute shrinkage and selection operator (LASSO) regression analysis. The expression levels of characteristic genes were evaluated in the peripheral blood mononuclear cells of CAD patients and healthy controls for verification. Results A total of 204 upregulated and 339 downregulated DEGs were identified, which were mainly enriched in the following pathways: "Apoptosis", "Th17 cell differentiation", "Th1 and Th2 cell differentiation", "Glycerolipid metabolism", and "Fat digestion and absorption". Five characteristic genes, LMAN1L, DOK4, CHFR, CEL and CCDC28A, were identified by LASSO analysis, and the results of the immune cell infiltration analysis indicated that the proportion of immune infiltrating cells (activated CD8 T cells and CD56 DIM natural killer cells) in the CAD group was lower than that in the control group. The expressions of CHFR, CEL and CCDC28A in the peripheral blood of the healthy controls and CAD patients were significantly different. Conclusion We identified CHFR, CEL and CCDC28A as potential biomarkers related to immune infiltration in CAD based on public data sets and clinical samples. This finding will contribute to providing a potential target for early noninvasive diagnosis and immunotherapy of CAD.
Collapse
Affiliation(s)
- Tianwen He
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Muheremu Muhetaer
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Jiahe Wu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Jingjing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Yushuang Hu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Tong Zhang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Yunxiang Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Qiongxin Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Huanhuan Cai
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
14
|
Yan M, Gu Y, Sun H, Ge Q. Neutrophil extracellular traps in tumor progression and immunotherapy. Front Immunol 2023; 14:1135086. [PMID: 36993957 PMCID: PMC10040667 DOI: 10.3389/fimmu.2023.1135086] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
Tumor immunity is a growing field of research that involves immune cells within the tumor microenvironment. Neutrophil extracellular traps (NETs) are neutrophil-derived extracellular web-like chromatin structures that are composed of histones and granule proteins. Initially discovered as the predominant host defense against pathogens, NETs have attracted increasing attention due to they have also been tightly associated with tumor. Excessive NET formation has been linked to increased tumor growth, metastasis, and drug resistance. Moreover, through direct and/or indirect effects on immune cells, an abnormal increase in NETs benefits immune exclusion and inhibits T-cell mediated antitumor immune responses. In this review, we summarize the recent but rapid progress in understanding the pivotal roles of NETs in tumor and anti-tumor immunity, highlighting the most relevant challenges in the field. We believe that NETs may be a promising therapeutic target for tumor immunotherapy.
Collapse
Affiliation(s)
- Meina Yan
- Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
- *Correspondence: Meina Yan, ;
| | - Yifeng Gu
- Department of Laboratory Medicine, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu, China
| | - Hongxia Sun
- Department of Gynecology and Obstetrics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Qinghong Ge
- Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
15
|
Hannani D, Leplus E, Laurin D, Caulier B, Aspord C, Madelon N, Bourova-Flin E, Brambilla C, Brambilla E, Toffart AC, Laulagnier K, Chaperot L, Plumas J. A New Plasmacytoid Dendritic Cell-Based Vaccine in Combination with Anti-PD-1 Expands the Tumor-Specific CD8+ T Cells of Lung Cancer Patients. Int J Mol Sci 2023; 24:ijms24031897. [PMID: 36768214 PMCID: PMC9915756 DOI: 10.3390/ijms24031897] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
The purpose of immune checkpoint inhibitor (ICI)-based therapies is to help the patient's immune system to combat tumors by restoring the immune response mediated by CD8+ cytotoxic T cells. Despite impressive clinical responses, most patients do not respond to ICIs. Therapeutic vaccines with autologous professional antigen-presenting cells, including dendritic cells, do not show yet significant clinical benefit. To improve these approaches, we have developed a new therapeutic vaccine based on an allogeneic plasmacytoid dendritic cell line (PDC*line), which efficiently activates the CD8+ T-cell response in the context of melanoma. The goal of the study is to demonstrate the potential of this platform to activate circulating tumor-specific CD8+ T cells in patients with lung cancer, specifically non-small-cell lung cancer (NSCLC). PDC*line cells loaded with peptides derived from tumor antigens are used to stimulate the peripheral blood mononuclear cells of NSCLC patients. Very interestingly, we demonstrate an efficient activation of specific T cells for at least two tumor antigens in 69% of patients irrespective of tumor antigen mRNA overexpression and NSCLC subtype. We also show, for the first time, that the antitumor CD8+ T-cell expansion is considerably improved by clinical-grade anti-PD-1 antibodies. Using PDC*line cells as an antigen presentation platform, we show that circulating antitumor CD8+ T cells from lung cancer patients can be activated, and we demonstrate the synergistic effect of anti-PD-1 on this expansion. These results are encouraging for the development of a PDC*line-based vaccine in NSCLC patients, especially in combination with ICIs.
Collapse
Affiliation(s)
| | | | - David Laurin
- Recherche et Développement, EFS, 38000 Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
| | - Benjamin Caulier
- Recherche et Développement, EFS, 38000 Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
| | - Caroline Aspord
- Recherche et Développement, EFS, 38000 Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
| | - Natacha Madelon
- Recherche et Développement, EFS, 38000 Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
| | - Ekaterina Bourova-Flin
- Groupe EpiMed, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
| | - Christian Brambilla
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
- Centre Hospitalo-Universitaire Grenoble-Alpes, Université Grenoble-Alpes, 38000 Grenoble, France
| | - Elisabeth Brambilla
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
- Centre Hospitalo-Universitaire Grenoble-Alpes, Université Grenoble-Alpes, 38000 Grenoble, France
| | - Anne-Claire Toffart
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
- Centre Hospitalo-Universitaire Grenoble-Alpes, Université Grenoble-Alpes, 38000 Grenoble, France
| | | | - Laurence Chaperot
- Recherche et Développement, EFS, 38000 Grenoble, France
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, 38000 Grenoble, France
| | - Joël Plumas
- PDC*line Pharma, 38000 Grenoble, France
- Recherche et Développement, EFS, 38000 Grenoble, France
- Correspondence:
| |
Collapse
|
16
|
Sánchez-León ML, Jiménez-Cortegana C, Cabrera G, Vermeulen EM, de la Cruz-Merino L, Sánchez-Margalet V. The effects of dendritic cell-based vaccines in the tumor microenvironment: Impact on myeloid-derived suppressor cells. Front Immunol 2022; 13:1050484. [PMID: 36458011 PMCID: PMC9706090 DOI: 10.3389/fimmu.2022.1050484] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/27/2022] [Indexed: 09/27/2023] Open
Abstract
Dendritic cells (DCs) are a heterogenous population of professional antigen presenting cells whose main role is diminished in a variety of malignancies, including cancer, leading to ineffective immune responses. Those mechanisms are inhibited due to the immunosuppressive conditions found in the tumor microenvironment (TME), where myeloid-derived suppressor cells (MDSCs), a heterogeneous population of immature myeloid cells known to play a key role in tumor immunoevasion by inhibiting T-cell responses, are extremely accumulated. In addition, it has been demonstrated that MDSCs not only suppress DC functions, but also their maturation and development within the myeloid linage. Considering that an increased number of DCs as well as the improvement in their functions boost antitumor immunity, DC-based vaccines were developed two decades ago, and promising results have been obtained throughout these years. Therefore, the remodeling of the TME promoted by DC vaccination has also been explored. Here, we aim to review the effectiveness of different DCs-based vaccines in murine models and cancer patients, either alone or synergistically combined with other treatments, being especially focused on their effect on the MDSC population.
Collapse
Affiliation(s)
- María Luisa Sánchez-León
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, University of Seville, Seville, Spain
- Medical Oncology Service, Virgen Macarena University Hospital, Seville, Spain
| | - Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, University of Seville, Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, Seville, Spain
| | - Gabriel Cabrera
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe capital, Argentina
| | - Elba Mónica Vermeulen
- Laboratorio de Células Presentadoras de Antígeno y Respuesta Inflamatoria, Instituto de Medicina Experimental (IMEX) - CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | - Victor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, University of Seville, Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, Seville, Spain
| |
Collapse
|