1
|
Shi X, Zhao H, Yu J, Cai P, Zhou S, Yang N, Li D. Changes in PD-1 expression on T lymphocyte subsets and related immune indicators before and after definitive chemoradiotherapy for esophageal squamous cell carcinoma. Ann Med 2025; 57:2445190. [PMID: 39713872 DOI: 10.1080/07853890.2024.2445190] [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: 05/25/2024] [Revised: 07/29/2024] [Accepted: 11/30/2024] [Indexed: 12/24/2024] Open
Abstract
OBJECTIVE This study aimed to observe the dynamic changes in the expression of T lymphocytes, natural killer (NK) cells, and PD-1 in patients with first-diagnosed esophageal squamous cell carcinoma (ESCC) before and after chemoradiotherapy (CRT) and evaluate the impact of PD-1 expression in peripheral blood on the short-term outcome of patients with ESCC. PATIENTS AND METHODS Seventy-three patients with ESCC who were treated with definitive CRT were enrolled. Before and after CRT, flow cytometry was used to detect thePD-1 expression in the peripheral blood and related immune indicators. Peripheral blood from 10 healthy individuals was used as control. RESULTS The percentages of CD3+ (p = 0.018), CD4+ (p < 0.001), and CD8+ T cells (p < 0.001); NK cells (p = 0.009); and the CD4+/CD8+ ratio (p < 0.001), as well as PD-1+CD3+ (p < 0.001), PD-1+CD4+ (p < 0.001), and PD-1+CD8+ (p < 0.001) T cells, before CRT significantly differed from those in the post-CRT group. The percentages of PD-1+CD8+ T cells differed significantly between the radiotherapy alone and CRT groups (p < 0.05). PD-1 expression in CD3+, CD4+, and CD8+ T cells significantly decreased in patients achieving overall response rate (all p < 0.05). Compared with those in the incomplete response group, PD-1+CD8+ T cells significantly decreased in the CR group (p < 0.05). CONCLUSION CRT aggravated immunosuppression and increased PD-1 expression in T lymphocyte subsets in patients with ESCC, possibly related to the radiation field. PD-1 expression in T lymphocyte subsets can predict short-term outcomes in patients and provide a theoretical basis for the sequential application of PD-1 immunosuppressants after radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xueling Shi
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Hongyu Zhao
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Jiaqi Yu
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Peng Cai
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Shixiang Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Ning Yang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Duojie Li
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
2
|
Luo R, Wang Z, Xu F, Xie K. Dexmedetomidine improve lung inflammation by regulating autophagy and apoptosis of CD4+ T cell via AMPK/mTOR signaling. Mol Immunol 2025; 183:1-11. [PMID: 40311186 DOI: 10.1016/j.molimm.2025.04.011] [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: 03/06/2025] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
OBJECTIVES To investigate the protective effect and potential mechanism of dexmedetomidine (Dex) in acute lung injury (ALI). MATERIALS AND METHODS C57BL/6 mice and EL-4 cells were used for in vivo and in vitro studies, respectively. Cecal ligation and puncture (CLP) method was used to prepare an acute lung injury model. After dexmedetomidine intervention, tissue and cell samples were collected to evaluate and measure the severity of lung damage, the proportion of Treg cells, the expression of autophagy-related protein levels and AMPK/mTOR pathways. RESULTS Dex reduced lung damage, and IL-17a, MPO positive cells in the lung, decreased the levels of pro-inflammatory cytokines, and restrain autophagy and apoptosis via the activation of the AMPK/mTOR pathway and increase of the proportion of Tregs. CONCLUSIONS Dex can inhibit the levels of autophagy and apoptosis, increase the proportion of Treg cells, and reduce CLP induced acute lung injury through regulating AKMP/MTOR pathway.
Collapse
Affiliation(s)
- Renjie Luo
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, China
| | - Zhao Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Ke Xie
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Lim BJW, Liu M, Wang L, Kong SLY, Yin T, Yan C, Xiang K, Cao C, Wu H, Mihai A, Tay FPL, Wang E, Jiang Q, Ma Z, Tan L, Chia RN, Qin D, Pan CC, Wang XF, Li QJ. Neoadjuvant anti-4-1BB confers protection against spontaneous metastasis through low-affinity intratumor CD8 + T cells in triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635356. [PMID: 39975187 PMCID: PMC11838326 DOI: 10.1101/2025.01.29.635356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neoadjuvant immunotherapy seeks to harness the primary tumor as a source of relevant tumor antigens to enhance systemic anti-tumor immunity through improved immunological surveillance. Despite having revolutionized the treatment of patients with high-risk early-stage triple-negative breast cancer (TNBC), a significant portion of patients remain unresponsive and succumb to metastatic recurrence post-treatment. Here, we found that optimally scheduled neoadjuvant administration of anti-4-1BB monotherapy was able to counteract metastases and prolong survival following surgical resection. Phenotypic and transcriptional profiling revealed enhanced 4-1BB expression on tumor-infiltrating intermediate (T int ), relative to progenitor (T prog ) and terminally exhausted (T term ) T cells. Furthermore, T int was enriched in low-affinity T cells. Treatment with anti-4-1BB drove clonal expansion of T int , with reduced expression of tissue-retention marker CD103 in T prog . This was accompanied by increased TCR clonotype sharing between paired tumors and pre-metastatic lungs. Further interrogation of sorted intratumor T cells confirmed enhanced T cell egress into circulation following anti-4-1BB treatment. In addition, gene signature extracted from anti-4-1BB treated T int was consistently associated with improved clinical outcomes in BRCA patients. Combinatorial neoadjuvant anti-4-1BB and ablation of tumor-derived CXCL16 resulted in enhanced therapeutic effect. These findings illustrate the intratumor changes underpinning the efficacy of neoadjuvant anti-4-1BB, highlighting the reciprocity between local tissue-retention and distant immunologic fortification, suggesting treatment can reverse the siphoning of intratumor T cells to primary tumor, enabling redistribution to distant tissues and subsequent protection against metastases.
Collapse
|
4
|
Shen M, Sun Z, Wang C, Zhang S, Jia B, Huang B, Xu L, Zhu Z, Bu Q, Li C, Zhu R, Wei L, Wei K. Duck circovirus regulates the expression of duck CLDN2 protein by activating the MAPK-ERK pathway to affect its adhesion and infection. J Virol 2024; 98:e0049724. [PMID: 39345142 PMCID: PMC11495148 DOI: 10.1128/jvi.00497-24] [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: 03/15/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
Duck circovirus (DuCV) is widely recognized as a prominent virus in China's duck farming industry, known for its ability to cause persistent infections and significant immunosuppression, which can lead to an increased susceptibility to secondary infections, posing a significant threat to the duck industry. Moreover, clinical evidence also indicates the potential vertical transmission of the virus through duck embryos to subsequent generations of ducklings. However, the limited availability of suitable cell lines for in vitro cultivation of DuCV has hindered further investigation into the molecular mechanisms underlying its infection and pathogenicity. In this study, we observed that oral DuCV infection in female breeding ducks can lead to oviduct, ovarian, and follicular infections. Subsequently, the infection can be transmitted to the fertilized eggs, resulting in the emergence of virus-carrying ducklings upon hatching. In contrast, the reproductive organs of male breeding ducks were unaffected by the virus, thus confirming that vertical transmission of DuCV primarily occurs through infection in female breeding ducks. By analyzing transcriptome sequencing data from the oviduct, we focused on claudin-2, a gene encoding the tight junction protein CLDN2 located on the cell membrane, which showed significantly increased expression in DuCV-infected oviducts of female breeding ducks. Notably, CLDN2 was confirmed to interact with the unique structural protein of DuCV, namely capsid protein (Cap), through a series of experimental approaches including co-immunoprecipitation (co-IP), GST pull-down, immunofluorescence, and adhesion-blocking assays. Furthermore, we demonstrated that the Cap protein binds to the extracellular loop structural domains EL1 and EL2 of CLDN2. Subsequently, by constructing a series of truncated bodies of the CLDN2 promoter region, we identified the transcription factor SP5 for CLDN2. Moreover, we found that DuCV infection triggers the activation of the MAPK-ERK signaling pathway in DEF cells and ducks, leading to an upregulation of SP5 and CLDN2 expression. This process ultimately leads to the transportation of mature CLDN2 to the cell surface, thereby facilitating increased virus adherence to the target organs. In conclusion, we discovered that DuCV utilizes host CLDN2 proteins to enhance adhesion and infection in oviducts and other target organs. Furthermore, we elucidated the signaling pathways involved in the interaction between DuCV Cap proteins and CLDN2, which provides valuable insights into the molecular mechanism underlying DuCV's infection and vertical transmission. IMPORTANCE Although duck circovirus (DuCV) poses a widespread infection and a serious hazard to the duck industry, the molecular mechanisms underlying DuCV infection and transmission remain elusive. We initially demonstrated vertical transmission of DuCV through female breeding ducks by simulating natural infection. Furthermore, a differentially expressed membrane protein CLDN2 was identified on the DuCV-infected oviduct of female ducks, and its extracellular loop structural domains EL1 and EL2 were identified as the interaction sites of DuCV Cap proteins. Moreover, the binding of DuCV Cap to CLDN2 triggered the intracellular MAPK-ERK pathway and activated the downstream transcription factor SP5. Importantly, we demonstrated that intracellular Cap also interacts with SP5, leading to upregulation of CLDN2 transcription and facilitating enhanced adherence of DuCV to target tissue, thereby promoting viral infection and transmission. Our study sheds light on the molecular mechanisms underlying vertical transmission of DuCV, highlighting CLDN2 as a promising target for drug development against DuCV infection.
Collapse
Affiliation(s)
- Mingyue Shen
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Zhenhong Sun
- College of Basic Medical Sciences, Shandong First Medical University, Tai'an, China
| | - Cheng Wang
- Shandong Academy of Chinese Medicine, Ji'nan, China
| | - Shuyu Zhang
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Baoyu Jia
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Bohan Huang
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Li Xu
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Zhiyu Zhu
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Qingyun Bu
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Chen Li
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Ruiliang Zhu
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Liangmeng Wei
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Kai Wei
- Shandong Provincial Key Laboratory of Zoonoses, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
5
|
Giansanti M, Theinert T, Boeing SK, Haas D, Schlegel PG, Vacca P, Nazio F, Caruana I. Exploiting autophagy balance in T and NK cells as a new strategy to implement adoptive cell therapies. Mol Cancer 2023; 22:201. [PMID: 38071322 PMCID: PMC10709869 DOI: 10.1186/s12943-023-01893-w] [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: 09/14/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Autophagy is an essential cellular homeostasis pathway initiated by multiple stimuli ranging from nutrient deprivation to viral infection, playing a key role in human health and disease. At present, a growing number of evidence suggests a role of autophagy as a primitive innate immune form of defense for eukaryotic cells, interacting with components of innate immune signaling pathways and regulating thymic selection, antigen presentation, cytokine production and T/NK cell homeostasis. In cancer, autophagy is intimately involved in the immunological control of tumor progression and response to therapy. However, very little is known about the role and impact of autophagy in T and NK cells, the main players in the active fight against infections and tumors. Important questions are emerging: what role does autophagy play on T/NK cells? Could its modulation lead to any advantages? Could specific targeting of autophagy on tumor cells (blocking) and T/NK cells (activation) be a new intervention strategy? In this review, we debate preclinical studies that have identified autophagy as a key regulator of immune responses by modulating the functions of different immune cells and discuss the redundancy or diversity among the subpopulations of both T and NK cells in physiologic context and in cancer.
Collapse
Affiliation(s)
- Manuela Giansanti
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Tobias Theinert
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Sarah Katharina Boeing
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Dorothee Haas
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paola Vacca
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Francesca Nazio
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy.
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Ignazio Caruana
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
6
|
Gatti A, Zizzo G, De Paschale M, Tamburello A, Castelnovo L, Faggioli PM, Clerici P, Brando B, Mazzone A. Assessing SARS-CoV-2-specific T-cell reactivity in late convalescents and vaccinees: Comparison and combination of QuantiFERON and activation-induced marker assays, and relation with antibody status. PLoS One 2023; 18:e0285728. [PMID: 37220145 DOI: 10.1371/journal.pone.0285728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 05/25/2023] Open
Abstract
OBJECTIVES Monitoring of SARS-CoV-2 spread and vaccination strategies have relied on antibody (Ab) status as a correlate of protection. We used QuantiFERON™ (QFN) and Activation-Induced Marker (AIM) assays to measure memory T-cell reactivity in unvaccinated individuals with prior documented symptomatic infection (late convalescents) and fully vaccinated asymptomatic donors (vaccinees). METHODS Twenty-two convalescents and 13 vaccinees were enrolled. Serum anti-SARS-CoV-2 S1 and N Abs were measured using chemiluminescent immunoassays. QFN was performed following instructions and interferon-gamma (IFN-γ) measured by ELISA. AIM was performed on aliquots of antigen-stimulated samples from QFN tubes. SARS-CoV-2-specific memory CD4+CD25+CD134+, CD4+CD69+CD137+ and CD8+CD69+CD137+ T-cell frequencies were measured by flow cytometry. RESULTS In convalescents, substantial agreement was observed between QFN and AIM assays. IFN-γ concentrations and AIM+ (CD69+CD137+) CD4+ T-cell frequencies correlated with each other, with Ab levels and AIM+ CD8+ T-cell frequencies, whereas AIM+ (CD25+CD134+) CD4+ T-cell frequencies correlated with age. AIM+ CD4+ T-cell frequencies increased with time since infection, whereas AIM+ CD8+ T-cell expansion was greater after recent reinfection. QFN-reactivity and anti-S1 titers were lower, whereas anti-N titers were higher, and no statistical difference in AIM-reactivity and Ab positivity emerged compared to vaccinees. CONCLUSIONS Albeit on a limited sample size, we confirm that coordinated, cellular and humoral responses are detectable in convalescents up to 2 years after prior infection. Combining QFN with AIM may enhance detection of naturally acquired memory responses and help stratify virus-exposed individuals in T helper 1-type (TH1)-reactive (QFNpos AIMpos Abshigh), non-TH1-reactive (QFNneg AIMpos Abshigh/low), and pauci-reactive (QFNneg AIMneg Abslow).
Collapse
Affiliation(s)
- Arianna Gatti
- Laboratory of Haematology, Transfusion Center, Legnano Hospital, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Gaetano Zizzo
- Department of Internal Medicine, Legnano and Cuggiono Hospitals, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Massimo De Paschale
- Unit of Microbiology, Legnano Hospital, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Antonio Tamburello
- Department of Internal Medicine, Legnano and Cuggiono Hospitals, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Laura Castelnovo
- Department of Internal Medicine, Legnano and Cuggiono Hospitals, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Paola Maria Faggioli
- Department of Internal Medicine, Legnano and Cuggiono Hospitals, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Pierangelo Clerici
- Unit of Microbiology, Legnano Hospital, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Bruno Brando
- Laboratory of Haematology, Transfusion Center, Legnano Hospital, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Antonino Mazzone
- Department of Internal Medicine, Legnano and Cuggiono Hospitals, ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| |
Collapse
|
7
|
Jin Z, Sun X, Wang Y, Zhou C, Yang H, Zhou S. Regulation of autophagy fires up the cold tumor microenvironment to improve cancer immunotherapy. Front Immunol 2022; 13:1018903. [PMID: 36300110 PMCID: PMC9589261 DOI: 10.3389/fimmu.2022.1018903] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapies, such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells, have revolutionized the treatment of patients with advanced and metastatic tumors resistant to traditional therapies. However, the immunosuppressed tumor microenvironment (TME) results in a weak response to immunotherapy. Therefore, to realize the full potential of immunotherapy and obstacle barriers, it is essential to explore how to convert cold TME to hot TME. Autophagy is a crucial cellular process that preserves cellular stability in the cellular components of the TME, contributing to the characterization of the immunosuppressive TME. Targeted autophagy ignites immunosuppressive TME by influencing antigen release, antigen presentation, antigen recognition, and immune cell trafficking, thereby enhancing the effectiveness of cancer immunotherapy and overcoming resistance to immunotherapy. In this review, we summarize the characteristics and components of TME, explore the mechanisms and functions of autophagy in the characterization and regulation of TME, and discuss autophagy-based therapies as adjuvant enhancers of immunotherapy to improve the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Zhicheng Jin
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Xuefeng Sun
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Yaoyao Wang
- Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, China
| | - Chao Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
- *Correspondence: Suna Zhou, ; HaihuaYang,
| | - Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
- Department of Radiation Oncology, Xi’an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, China
- *Correspondence: Suna Zhou, ; HaihuaYang,
| |
Collapse
|
8
|
Hasanvand A. COVID-19 and the role of cytokines in this disease. Inflammopharmacology 2022; 30:789-798. [PMID: 35505267 PMCID: PMC9064717 DOI: 10.1007/s10787-022-00992-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022]
Abstract
Studies have shown that SARS-CoV-2 has the ability to activate and mature proinflammatory cytokines in the body. Cytokine markers are a group of polypeptide signalling molecules that can induce and regulate many cellular biological processes by stimulating cell receptors at the surface. SARS-CoV-2 has been shown to be associated with activation of innate immunity, and an increase in neutrophils, mononuclear phagocytes, and natural killer cells has been observed, as well as a decrease in T cells including CD4+ and CD8. It is noteworthy that during the SARS-CoV-2 infection, an increase in the secretion or production of IL-6 and IL-8 is seen in COVID-19 patients along with a decrease in CD4+ and CD8+ and T cells in general. SARS-CoV-2 has been shown to significantly increase Th2, Th1/Th17 cells and antibody production in the body of patients with COVID-19. Specific immune profiles of SARS-CoV-2 infection can lead to secondary infections and dysfunction of various organs in the body. It has been shown that Interleukins (such as IL-1, IL-4, IL-6, IL-7, IL-10, IL-12, IL-17, and IL-18), IFN-γ, TNF-α,TGF-β and NF-κB play major roles in the body's inflammatory response to SARS-CoV-2 infection. The most important goal of this review is to study the role of inflammatory cytokines in COVID-19.
Collapse
Affiliation(s)
- Amin Hasanvand
- Department of Physiology and Pharmacology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
9
|
McCann K, von Witzleben A, Thomas J, Wang C, Wood O, Singh D, Boukas K, Bendjama K, Silvestre N, Nielsen FC, Thomas G, Sanchez-Elsner T, Greenbaum J, Schoenberger S, Peters B, Vijayanand P, Savelyeva N, Ottensmeier C. Targeting the tumor mutanome for personalized vaccination in a TMB low non-small cell lung cancer. J Immunother Cancer 2022; 10:e003821. [PMID: 35361728 PMCID: PMC8971766 DOI: 10.1136/jitc-2021-003821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Cancer is characterized by an accumulation of somatic mutations, of which a significant subset can generate cancer-specific neoepitopes that are recognized by autologous T cells. Such neoepitopes are emerging as important targets for cancer immunotherapy, including personalized cancer vaccination strategies. METHODS We used whole-exome and RNA sequencing analysis to identify potential neoantigens for a patient with non-small cell lung cancer. Thereafter, we assessed the autologous T-cell reactivity to the candidate neoantigens using a long peptide approach in a cultured interferon gamma ELISpot and tracked the neoantigen-specific T-cells in the tumor by T-cell receptor (TCR) sequencing. In parallel, identified gene variants were incorporated into a Modified Vaccinia Ankara-based vaccine, which was evaluated in the human leucocyte antigen A*0201 transgenic mouse model (HHD). RESULTS Sequencing revealed a tumor with a low mutational burden: 2219 sequence variants were identified from the primary tumor, of which 23 were expressed in the transcriptome, involving 18 gene products. We could demonstrate spontaneous T-cell responses to 5/18 (28%) mutated gene variants, and further analysis of the TCR repertoire of neoantigen-specific CD4+ and CD8+ T cells revealed TCR clonotypes that were expanded in both blood and tumor tissue. Following vaccination of HHD mice, de novo T-cell responses were generated to 4/18 (22%) mutated gene variants; T cells reactive against two variants were also evident in the autologous setting. Subsequently, we determined the major histocompatibility complex restriction of the T-cell responses and used in silico prediction tools to determine the likely neoepitopes. CONCLUSIONS Our study demonstrates the feasibility of efficiently identifying tumor-specific neoantigens that can be targeted by vaccination in tumors with a low mutational burden, promising successful clinical exploitation, with trials currently underway.
Collapse
Affiliation(s)
- Katy McCann
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Adrian von Witzleben
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
- Department of Otorhinolaryngology, Head & Neck Surgery, University of Ulm, Ulm, Germany
| | - Jaya Thomas
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Chuan Wang
- Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Oliver Wood
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Divya Singh
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Konstantinos Boukas
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Kaidre Bendjama
- Research and Development Department, Transgene, Illkirch-Graffenstaden, France
| | - Nathalie Silvestre
- Research and Development Department, Transgene, Illkirch-Graffenstaden, France
| | | | - Gareth Thomas
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Tilman Sanchez-Elsner
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Jason Greenbaum
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Stephen Schoenberger
- Laboratory of Cellular Immunology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Natalia Savelyeva
- Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Christian Ottensmeier
- Cancer Research UK Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton Faculty of Medicine, Southampton, UK
- Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- La Jolla Institute for Immunology, La Jolla, California, USA
| |
Collapse
|
10
|
Nunes-Cabaço H, Ramalho-dos-Santos A, Pires AR, Martins LR, Barata JT, Sousa AE. Human CD4 T Cells From Thymus and Cord Blood Are Convertible Into CD8 T Cells by IL-4. Front Immunol 2022; 13:834033. [PMID: 35222424 PMCID: PMC8880616 DOI: 10.3389/fimmu.2022.834033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Commitment to the CD4+ or CD8+ T cell lineages is linked to the acquisition of a functional program broadly defined by helper and cytotoxic properties, respectively. The mechanisms underlying these processes in the human thymus remain largely unclear. Moreover, recent thymic emigrants are thought to have some degree of plasticity, which may be important for the shaping of the immune system and adjustment to specific peripheral needs. We show here that IL-4 induces proliferation-independent de novo synthesis of CD8αβ in human CD4 single-positive (SP) thymocytes, generating a stable CD8SP population that features a diverse TCRαβ repertoire, CD4 expression shut-down and ThPOK downregulation. IL-4 also promotes an innate-like program in both CD4SP and CD8SP thymocytes, characterized by Eomes upregulation in the absence of T-bet, in line with its recognized role in the generation of thymic innate-like CD8+ T cells. The clinical relevance of these findings is further supported by the profile of IL-4 production and IL-4 receptor expression that we identified in the human thymus. Importantly, human cord blood CD4+ T cells preserve the ability to generate Eomes+ CD8+ T cells in the presence of IL-4, with implications in neonatal immunity. Our results support a role for IL-4 in the dynamic regulation of human thymocyte plasticity and identify novel strategies to modulate immune responses.
Collapse
Affiliation(s)
- Helena Nunes-Cabaço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | | | | | | | - Ana E. Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
Honan AM, Vazquez EN, Chen Z. Lymph Node Stromal Cell-Intrinsic MHC Class II Expression Promotes MHC Class I-Restricted CD8 T Cell Lineage Conversion to Regulatory CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:1530-1544. [PMID: 34408011 DOI: 10.4049/jimmunol.2100396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/07/2021] [Indexed: 12/29/2022]
Abstract
MHC class I (MHC-I)-restricted CD4+ T cells have long been discovered in the natural repertoire of healthy humans as well as patients with autoimmune diseases or cancer, but the exact origin of these cells remains to be fully characterized. In mouse models, mature peripheral CD8+ T cells have the potential to convert to CD4+ T cells in the mesenteric lymph nodes. This conversion can produce a unique population of MHC-I-restricted CD4+ T cells including Foxp3+ regulatory T cells termed MHC-I-restricted CD4+Foxp3+ T (CI-Treg) cells. In this study we examined the cellular and molecular elements that promote CD8-to-CD4 lineage conversion and the development of CI-Treg cells in mice. Using adoptive transfer and bone marrow chimera experiments, we found that the differentiation of CI-Treg cells was driven by lymph node stromal cell (LNSC)-intrinsic MHC-II expression as opposed to transcytosis of MHC-II from bone marrow-derived APCs. The lineage conversion was accompanied by Runx3 versus ThPOK transcriptional switch. This finding of a new role for LNSCs in vivo led us to develop an efficient tissue culture method using LNSCs to generate and expand CI-Treg cells in vitro. CI-Treg cells expanded in vitro with LNSCs effectively suppressed inflammatory tissue damage caused by pathogenic CD4+ T cells in mouse models of colitis. This study identified a novel role of MHC-II expressed by LNSCs in immune regulation and the potential utilization of LNSCs to generate novel subsets of immune regulatory cells for therapeutic applications.
Collapse
Affiliation(s)
- Amanda M Honan
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and
| | - Emily N Vazquez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
12
|
Chen C, Liu Y, Cui B. Effect of radiotherapy on T cell and PD-1 / PD-L1 blocking therapy in tumor microenvironment. Hum Vaccin Immunother 2021; 17:1555-1567. [PMID: 33428533 DOI: 10.1080/21645515.2020.1840254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer is a worldwide problem that threatens human health. Radiotherapy plays an important role in a variety of cancer treatment methods. The administration of radiotherapy can alter the differentiation pathways and functions of T cells, which in turn improves the immune response of T cells. Radiotherapy can also induce up-regulation of PD-L1 expression, which means that it has great potential for enhancing the therapeutic effect of anti-PD-1/PD-L1 inhibitors and reducing the risk of drug resistance toward them. At present, the combination of radiotherapy and anti-PD-1/PD-L1 inhibitors has shown significant therapeutic effects in clinical tumor research. This review focuses on the mechanism of radiotherapy on T cells reported in recent years, as well as related research progress in the application of PD-1/PD-L1 blockers. It will provide a theoretical basis for the rational clinical application of radiotherapy combined with PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Chen Chen
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Yanlong Liu
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Binbin Cui
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| |
Collapse
|
13
|
Xu P, Luo H, Kong Y, Lai WF, Cui L, Zhu X. Cancer neoantigen: Boosting immunotherapy. Biomed Pharmacother 2020; 131:110640. [PMID: 32836075 DOI: 10.1016/j.biopha.2020.110640] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/13/2020] [Accepted: 08/16/2020] [Indexed: 12/21/2022] Open
Abstract
Tumor neoantigen has a high degree of immunogenicity. As one of the emerging methods of tumor immunotherapy, the vaccine developed against it has served to clinical trials of various solid tumors, especially in the treatment of melanoma. Currently, a variety of immunotherapy methods have been applied to the treatment of the tumor. However, other therapeutic methods have the disadvantages of low specificity and prominent side effects. Treatments require tumor antigen with higher immunogenicity as the target of immune attack. This review will recommend the identification of neoantigen, the influencing factors of neoantigen, and the application of personalized vaccines for neoantigen in metastatic tumors such as malignant melanoma.
Collapse
Affiliation(s)
- Peijia Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, 524023, China
| | - Haiqing Luo
- Cancer Center, Affiliated Hospital, Guangdong Medical University, Zhanjiang, 524023, China
| | - Ying Kong
- Department of Clinical Laboratory, Hubei No. 3 People's Hospital of Jianghan University, Wuhan, 430033, China
| | - Wing-Fu Lai
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China; School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China.
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, China.
| | - Xiao Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, 524023, China.
| |
Collapse
|
14
|
Yang K, Chi H. Autophagy modulates CD4 + T-cell lineage recommitment upon pathogen infection. Cell Mol Immunol 2020; 17:682-683. [PMID: 32047257 PMCID: PMC7331744 DOI: 10.1038/s41423-020-0368-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 11/09/2022] Open
Affiliation(s)
- Kai Yang
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|