{"title":"Chronological Effects of Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer","authors":"Xinyue Guo, Lanqun Qin, Xinmeng Wang, Qian Geng, Dongqing Li, Yingying Lu, Hua Jiang","doi":"10.1111/imm.13897","DOIUrl":"10.1111/imm.13897","url":null,"abstract":"<div>\u0000 \u0000 <p>Circadian rhythm is a physiological process that oscillates in a 24 h cycle. It has a complex connection with the function of the human immune system and even with the development of tumours. Previous studies demonstrated the time-dependent effects of chemotherapy and radiotherapy; however, there are few studies on the timing effects of immunotherapy. Here, we explored the differences in the efficacy of immune checkpoint inhibitors (ICIs) administered at different circadian rhythm times in non-small cell lung cancer (NSCLC). C57BL/6N lung Lewis cancer mice models were constructed. Then, mice were intraperitoneally injected with saline or anti-PD-1 antibody at 7 AM or 7 PM, The expression of PD-L1 was detected by flow cytometry, and the expressions of clock gene BMAL1 and PER2 were detected by polymerase chain reaction (PCR) after treatment. A retrospective analysis was conducted on patients with NSCLC who received ICIs in our department from June 2020 to December 2022. Animal experiments showed that mice treated with ICIs in the morning showed slower tumour growth and smaller tumour volumes than those in the afternoon, accompanied by increased expression of BMAL1 and PER2 and suppression of PD-L1 expression. Retrospective analysis showed that patients who received ICIs in the afternoon (after 12:00) had significantly longer progression-free survival than those in the morning (before 12:00) (median was 16.5 months versus 9.8 months, respectively, <i>p =</i> 0.031, hazard ratio = 1.87). These findings suggest that immunotherapy may have time dependence, offering a novel therapeutic strategy.</p>\u0000 </div>","PeriodicalId":13508,"journal":{"name":"Immunology","volume":"174 4","pages":"402-410"},"PeriodicalIF":4.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948215","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunologyPub Date : 2025-01-08DOI: 10.1111/imm.13896
Takamasa Kido, Hiroyuki Yanagisawa, Machi Suka
{"title":"Zinc Deficiency Reduces Intestinal Secretory Immunoglobulin A and Induces Inflammatory Responses via the Gut-Liver Axis","authors":"Takamasa Kido, Hiroyuki Yanagisawa, Machi Suka","doi":"10.1111/imm.13896","DOIUrl":"10.1111/imm.13896","url":null,"abstract":"<div>\u0000 \u0000 <p>Nutritional zinc (Zn) deficiency could impair immune function and affect bowel conditions. However, the mechanism by which Zn deficiency affects the immune function of gut-associated lymphoid tissue (GALT) remains unclear. We investigated how Zn deficiency affects the function of GALT and level of secretory IgA (sIgA), a key component of the intestinal immune barrier, its underlying mechanisms, and whether Zn deficiency induces bacterial translocation to the liver. As previous research has indicated that interleukin (IL)-4 administration or Zn supplementation has a beneficial effect on the spleen of Zn-deficient rats, we investigated whether these supplements reverse the GALT immune system. Five-week-old male rats were fed a standard diet, Zn-deficient diet supplemented with saline or IL-4 for 6 weeks, or Zn-deficient diet followed by a standard diet for 4 weeks. Zn deficiency suppressed sIgA secretion in the intestinal tract by affecting GALT function and induced inflammatory responses through bacterial translocation to the liver via the portal vein. Furthermore, IL-4 administration and Zn supplementation in rats with Zn deficiency elicited comparable beneficial effects on GALT function, suggesting that the administration of either IL-4 or Zn could prevent inflammatory response via bacterial translocation to the liver.</p>\u0000 </div>","PeriodicalId":13508,"journal":{"name":"Immunology","volume":"174 3","pages":"363-373"},"PeriodicalIF":4.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948219","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Effective Mucosal Adjuvantation of the Intranasal Enterovirus A71 Vaccine With Zymosan","authors":"Chiao-Li Chin, Yu-Li Lin, Pei-Yun Cheng, Ping Lee, Bor-Luen Chiang","doi":"10.1111/imm.13895","DOIUrl":"10.1111/imm.13895","url":null,"abstract":"<div>\u0000 \u0000 <p>Enterovirus A71 (EV-A71) has caused hand, foot, and mouth disease with an increased prevalence of neurological complications and acute mortality, threatening young children around the globe. By provoking mucosal immunity, intranasal vaccination has been suggested to prevent EV-A71 infection. However, antigens delivered via the nasal route usually fail to induce a protective memory response. Zymosan has been identified to activate multiple pattern recognition receptors to orchestrate innate and adaptive immunity. Herein, we aimed to investigate the capacity of zymosan to strengthen the vaccine response induced by an intranasal EV-A71 vaccine. First, we confirmed its remarkable capacity to ignite innate signaling by upregulating cytokine production in primary DCs in vitro. Second, we verified its capacity to promote the vaccine immunogenicity in vivo after triple vaccination with EV-A71, especially with the notable induction of virus-specific IgA at multiple mucosae and the IL-17-producing splenic population after antigen reencounter. Lastly, we validated its capacity to improve vaccine efficacy in vivo after dual vaccination by furnishing neonatal protection against lethal infection. Our findings show that zymosan, at a preferable dosage, could augment the benefits of the intranasal vaccination to tackle EV-A71 infection. This research provides a feasible strategy for preventing EV-A71 infection with severe complications and contributes to the development of nasal spray vaccination.</p>\u0000 </div>","PeriodicalId":13508,"journal":{"name":"Immunology","volume":"174 3","pages":"349-362"},"PeriodicalIF":4.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948216","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ImmunologyPub Date : 2025-01-07DOI: 10.1111/imm.13883
Jeppe Madura Larsen, Emma Alberte Lundsgaard, Niels Banhos Danneskiold-Samsøe, Si Brask Sonne, Neil Marcus Rigby, Alan Robert Mackie, Karsten Kristiansen, Katrine Lindholm Bøgh
{"title":"Prophylactic Peanut Allergen Ara h 6 Sublingual Immunotherapy Drives Expansion of FoxP3\u0000 +Helios− Regulatory T Cells in the Absence of Allergen-Specific IgA","authors":"Jeppe Madura Larsen, Emma Alberte Lundsgaard, Niels Banhos Danneskiold-Samsøe, Si Brask Sonne, Neil Marcus Rigby, Alan Robert Mackie, Karsten Kristiansen, Katrine Lindholm Bøgh","doi":"10.1111/imm.13883","DOIUrl":"10.1111/imm.13883","url":null,"abstract":"<p>Insights into the underlying immunological mechanisms of prophylactic sublingual immunotherapy (SLIT) may support the development of new strategies for improved prevention and treatment of food allergy. Here, we investigated the humoral, regulatory and sublingual tissue immune response to prophylactic SLIT administration of a single purified peanut allergen in Brown Norway (BN) rats. BN rats received daily sublingual administration of peanut allergen Ara h 6 for three weeks. Suppression of sensitisation was evaluated by subsequent intraperitoneal administration of Ara h 6. Ara h 6-specific IgE, IgA, IgG1 and IgG2a-c levels were measured in serum. The frequency of regulatory T (Treg) cells was analysed using flow cytometry. The sublingual tissue response to Ara h 6 was analysed by transcriptional profiling using mRNA-sequencing. Ara h 6 SLIT protected rats from subsequent sensitisation without inducing a detectable humoral immune response (Ara h 6-specific IgE, IgA, IgG1 and IgG2a-c) in serum. SLIT furthermore promoted the relative expansion of induced Helios<sup>−</sup> Treg cells within the conventional CD4<sup>+</sup>CD25<sup>+</sup>FoxP3<sup>+</sup> Treg population in sublingual draining lymph nodes and blood. In conclusion, prophylactic Ara h 6 SLIT drives the relative expansion of induced Helios<sup>−</sup> Treg cells in the absence of Ara h 6-specific IgA highlighting a potential novel IgA-independent Treg-related immune response at the sublingual mucosal site.</p>","PeriodicalId":13508,"journal":{"name":"Immunology","volume":"174 3","pages":"340-348"},"PeriodicalIF":4.9,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imm.13883","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142948217","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}