Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1553757
Lu Chen, Guangcong Ruan, Xuefei Zhao, Ailin Yi, Zhifeng Xiao, Yuting Tian, Yi Cheng, Dongfeng Chen, Yanling Wei
{"title":"<i>Pseudomonas aeruginosa</i> enhances anti-PD-1 efficacy in colorectal cancer by activating cytotoxic CD8<sup>+</sup> T cells.","authors":"Lu Chen, Guangcong Ruan, Xuefei Zhao, Ailin Yi, Zhifeng Xiao, Yuting Tian, Yi Cheng, Dongfeng Chen, Yanling Wei","doi":"10.3389/fimmu.2025.1553757","DOIUrl":"10.3389/fimmu.2025.1553757","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint therapy for colorectal cancer (CRC) has been found to be unsatisfactory for clinical treatment. Fecal microbiota transplantation (FMT) has been shown to remodel the intestinal flora, which may improve the therapeutic effect of αPD-1. Further exploration of key genera that can sensitize cells to αPD-1 for CRC treatment and preliminary exploration of immunological mechanisms may provide effective guidance for the clinical treatment of CRC.</p><p><strong>Methods: </strong>In this study, 16S rRNA gene sequencing was analyzed in the fecal flora of both responders and no-responders to αPD-1 treatment, and the therapeutic effect was experimentally verified.</p><p><strong>Results: </strong><i>Pseudomonas aeruginosa</i> was found to be highly abundant in the fecal flora of treated mice, and <i>Pseudomonas aeruginosa</i> mannose-sensitive hemagglutinin (PA-MSHA) in combination with αPD-1 was effective in the treatment of CRC through the induction of CD8<sup>+</sup> T-cell immunological effects.</p><p><strong>Conclusion: </strong>The clinical drug PA-MSHA can be used in combination with αPD-1 for the treatment of CRC as a potential clinical therapeutic option.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1553757"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968734/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1558867
Chen-Hao Yang, Yan Xu, Si-Yuan Fan
{"title":"Case Report: Anti-Yo antibody mediated paraneoplastic cerebellar degeneration in a patient with squamous cell lung carcinoma.","authors":"Chen-Hao Yang, Yan Xu, Si-Yuan Fan","doi":"10.3389/fimmu.2025.1558867","DOIUrl":"10.3389/fimmu.2025.1558867","url":null,"abstract":"<p><p>Paraneoplastic cerebellar degeneration (PCD) is a heterogeneous group of neurologic syndromes associated with primary tumors. It is postulated that the immune system targets a tumor antigen that is also expressed endogenously in the nervous system. The majority of these patients are diagnosed with breast cancer or gynecological cancer, while it is exceedingly rare in lung squamous cell carcinoma (LUSC) patients. Here we reported a rare case of anti-Yo antibody-positive PCD in a patient with LUSC and got successfully treated via immunotherapy and oncological treatment. The patient's ataxia symptoms alleviated following the administered treatments, suggesting that early immunotherapeutic intervention may have potential value in mitigating neurological deterioration. Furthermore, active and timely management of the primary carcinoma is crucial.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1558867"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968391/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Characterizing macrophage diversity in colorectal malignancies through single-cell genomics.","authors":"Tingshuo Zhao, Yinyi Luo, Yuanjie Sun, Zhigang Wei","doi":"10.3389/fimmu.2025.1526668","DOIUrl":"10.3389/fimmu.2025.1526668","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract, with increasing incidence and mortality rates, posing a significant burden on human health. Its progression relies on various mechanisms, among which the tumor microenvironment and tumor-associated macrophages (TAMs) have garnered increasing attention. Macrophage infiltration in various solid tumors is associated with poor prognosis and is linked to chemotherapy resistance in many cancers. These significant biological behaviors depend on the heterogeneity of macrophages. Tumor-promoting TAMs comprise subpopulations characterized by distinct markers and unique transcriptional profiles, rendering them potential targets for anticancer therapies through either depletion or reprogramming from a pro-tumoral to an anti-tumoral state. Single-cell RNA sequencing technology has significantly enhanced our research resolution, breaking the traditional simplistic definitions of macrophage subtypes and deepening our understanding of the diversity within TAMs. However, a unified elucidation of the nomenclature and molecular characteristics associated with this diversity remains lacking. In this review, we assess the application of conventional macrophage polarization subtypes in colorectal malignancies and explore several unique subtypes defined from a single-cell omics perspective in recent years, categorizing them based on their potential functions.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1526668"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968368/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1511385
Eduardo Liquidano-Perez, Gibert Maza-Ramos, Marco Yamazaki-Nakashimada, Rodolfo Rodríguez-Jurado, Alfonso G Ramírez Ristori, Juan Carlos Bustamante-Ogando, Mario Ernesto Cruz-Munoz, Arturo Gutierrez-Guerrero, Marimar Saez-de-Ocariz, Sara Espinosa-Padilla, Nideshda Ramirez-Uribe, Selma C Scheffler-Mendoza
{"title":"Case Report: Hydroa vacciniforme-like lymphoproliferative disorder, an EBV-associated disease, successfully treated with hematopoietic stem cell transplantation.","authors":"Eduardo Liquidano-Perez, Gibert Maza-Ramos, Marco Yamazaki-Nakashimada, Rodolfo Rodríguez-Jurado, Alfonso G Ramírez Ristori, Juan Carlos Bustamante-Ogando, Mario Ernesto Cruz-Munoz, Arturo Gutierrez-Guerrero, Marimar Saez-de-Ocariz, Sara Espinosa-Padilla, Nideshda Ramirez-Uribe, Selma C Scheffler-Mendoza","doi":"10.3389/fimmu.2025.1511385","DOIUrl":"10.3389/fimmu.2025.1511385","url":null,"abstract":"<p><strong>Introduction: </strong>The hydroa-vacciniforme-like lymphoproliferative disorder (HVLD) is a rare NK/T-cell condition affecting children in Latin America and Asia. It often progresses to systemic lymphoma, with Latin American patients experiencing worse outcomes compared to East Asians. Understanding viral and host genetic interactions is crucial for advancing targeted therapies. Here, we report a male patient with HVLD successfully treated with hematopoietic stem cell transplantation, highlighting its potential as a therapeutic approach for this aggressive disease.</p><p><strong>Case description: </strong>An 8-year-old boy presented with persistent skin lesions, fever, and pain. Biopsy confirmed a diagnosis of HVLD. Initial treatments with thalidomide and steroids provided temporary relief. At 12, lymphoma progression led to rituximab and CHOP chemotherapy. Further investigations revealed persistent EBV infection and lymphoma; hence, a haploidentical stem cell transplant was performed at 15. The procedure was successful, achieving complete immune reconstitution and viral clearance. Three years post-transplant, the patient remains in good health with no detectable EBV and complete vaccinations.</p><p><strong>Discussion: </strong>While EBV infection is common, only specific immunodeficiency states seem to enable EBV-related lymphoproliferative disorders. The exact mechanism leading to this immunosuppressive environment in HVLD remains unclear. Clinically, HVLD resembles specific inborn errors of immunity with EBV susceptibility. Additionally, cases of GATA2 and TACI deficiency presenting with HVLD suggest a potential link to underlying immune dysfunction. Further research in this area is crucial to understand the immunological basis of HVLD. Treatment options for HVLD are diverse and lack standardized protocols. Our case demonstrates the potential of HSCT with reduced-intensity conditioning and EBV-specific T-cell infusion as an effective cure. Given the limited understanding of HVLD, an immunological approach to characterizing patient profiles and prolonged follow-up are essential. While diverse therapies exist, HSCT offers the best hope for a cure. Further research towards tailored treatment strategies holds significant promise for improved patient outcomes.</p><p><strong>Conclusion: </strong>HVLD presents a complex and multifaceted challenge; our case demonstrates the potential of HSCT as a curative treatment. Unveiling the underlying immunology and tailoring therapies to patient profiles are crucial for improved outcomes. Further research is key to refining treatment strategies and offering hope for this rare and severe disease.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1511385"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968379/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1543649
Luis Brieva, Carmen Calles, Lamberto Landete, Celia Oreja-Guevara
{"title":"Current challenges in secondary progressive multiple sclerosis: diagnosis, activity detection and treatment.","authors":"Luis Brieva, Carmen Calles, Lamberto Landete, Celia Oreja-Guevara","doi":"10.3389/fimmu.2025.1543649","DOIUrl":"10.3389/fimmu.2025.1543649","url":null,"abstract":"<p><p>Approximately 50% diagnosed with relapsing-remitting multiple sclerosis (RRMS) transition to secondary progressive multiple sclerosis (SPMS) within 20 years following disease onset. However, early diagnosis of SPMS and effective treatment remain important clinical challenges. The lack of established diagnostic criteria often leads to delays in identifying SPMS. Also, there are limited disease-modifying therapies (DMTs) available for progressive forms of MS, and these therapies require evidence of disease activity to be initiated. This review examines the challenges in diagnosing SPMS at an early stage and summarizes the current and potential use of biomarkers of disease progression in clinical practice. We also discuss the difficulties in initiating the DMTs indicated for active SPMS (aSPMS), particularly in patients already undergoing treatment with DMTs that suppress disease activity, which may mask the presence of inflammatory activity required for the therapy switch. The article also addresses the DMTs available for both active and non-active SPMS, along with the clinical trials that supported the approval of DMTs indicated for aSPMS or relapsing MS in Europe, which includes aSPMS. We also offer insights on when discontinuing these treatments may be appropriate.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1543649"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968352/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795272","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1561152
F Javier Morán-Plata, Noemí Muñoz-García, Susana Barrena, Ana Yeguas, Ana Balanzategui, Sonia Carretero-Domínguez, Quentin Lécrevisse, María González-González, Sheila Mateos, Lidia Silos, Miguel Alcoceba, Fernando Solano, Miriam López-Parra, Vitor Botafogo, Alberto Orfao, Julia Almeida
{"title":"Altered immune cell profiles in blood of mature/peripheral T-cell leukemia/lymphoma patients: an EuroFlow study.","authors":"F Javier Morán-Plata, Noemí Muñoz-García, Susana Barrena, Ana Yeguas, Ana Balanzategui, Sonia Carretero-Domínguez, Quentin Lécrevisse, María González-González, Sheila Mateos, Lidia Silos, Miguel Alcoceba, Fernando Solano, Miriam López-Parra, Vitor Botafogo, Alberto Orfao, Julia Almeida","doi":"10.3389/fimmu.2025.1561152","DOIUrl":"10.3389/fimmu.2025.1561152","url":null,"abstract":"<p><strong>Introduction: </strong>The interactions between T-cell chronic lymphoproliferative disorder (T-CLPD) tumor cells and the bystander immune cells may play a critical role in the failure of immune surveillance and disease progression, but the altered blood immune profiles of T-CLPD remain unknown.</p><p><strong>Methods: </strong>Here we analyzed the distribution of residual non-tumoral immune cells in blood of 47 T-CLPD patients -14 T-prolymphocytic leukemia (T-PLL), 7 Sézary syndrome/mycosis fungoides (SS/MF) and 26 T-large granular lymphocytic leukemia (T-LGLL)-, as tumor models of neoplastic T-cells that resemble naive/central memory (N/CM), memory and terminal effector T-cells, respectively, compared to 110 age- and sex-matched healthy donors, using spectral flow cytometry.</p><p><strong>Results: </strong>Overall, our results showed deeply altered immune cell profiles in T-PLL, characterized by significantly increased counts of monocytes, dendritic cells, B-cells, NK-cells and innate lymphoid cells (ILC) -particularly ILC3-, together with reduced normal T-cells. In contrast, SS/MF showed neutrophilia, associated with decreased numbers of dendritic cells and NK-cells, potentially reflecting their increased migration from blood to the skin. In turn, T-LGLL displayed the mildest immune impairment, dependent on the TCD4+ <i>vs</i>. TCD8+ nature of the clonal T-cells and presence of <i>STAT3</i> mutations among TαβCD8+ T-LGLL cases. Further dissection of the normal T-cell compartment showed a significant reduction of the earliest T-cell maturation compartments (N/CM) in T-PLL and SS/MF, whereas T-cells remained within normal ranges in T-LGLL, with only a minor reduction of N/CM T-cells.</p><p><strong>Conclusion: </strong>These findings point out the existence of differentially altered innate and adaptive immune cell profiles in the distinct diagnostic subtypes of T-CLPD, with progressively less pronounced alterations from T-PLL and SS/MF to T-LGLL.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1561152"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968749/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795136","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1556857
Yating Ji, Qingyao Xie, Wei Wei, Zhen Huang, Xuhui Liu, Qi Ye, Yanping Liu, Xiaoyu Lu, Yixiao Lu, Renjie Hou, Qingping Zhang, Yanzi Xu, Jianhui Yuan, Shuihua Lu, Chongguang Yang
{"title":"Association between blood inflammatory status and the survival of tuberculosis: a five-year cohort study.","authors":"Yating Ji, Qingyao Xie, Wei Wei, Zhen Huang, Xuhui Liu, Qi Ye, Yanping Liu, Xiaoyu Lu, Yixiao Lu, Renjie Hou, Qingping Zhang, Yanzi Xu, Jianhui Yuan, Shuihua Lu, Chongguang Yang","doi":"10.3389/fimmu.2025.1556857","DOIUrl":"10.3389/fimmu.2025.1556857","url":null,"abstract":"<p><strong>Background: </strong>Blood inflammatory status is closely associated with tuberculosis (TB) progression. Emerging inflammatory indices from different leukocyte subtypes have become a prognostic hotspot for various diseases, yet their application in TB prognosis remains limited. This study aims to assess the impact of inflammatory status on TB patients' prognosis and its potential as a prognostic indicator to optimize prognostic assessment and therapeutic strategies.</p><p><strong>Methods: </strong>This study included 4027 TB patients admitted to a tuberculosis-designated hospital in Shenzhen from January 2017 to December 2022. Patients were classified into three inflammatory statuses (Q1-Q3) based on each index's level. We conducted Cox regression and restricted cubic splines (RCS) analyses to evaluate the association between inflammatory status and unfavorable outcome, subgroup analyses to understand heterogeneous associations among subpopulations, and receiver operating characteristic (ROC) analyses to evaluate the prognostic performance of inflammatory status on TB treatment outcomes.</p><p><strong>Results: </strong>During 48991.79 person-months of follow-up involving 4027 patients, 225 unfavorable outcomes occurred. Multivariable Cox regression indicated that the Q3 levels of CAR, CLR, dNLR, NLR, SII, and SIRI increased the risk of unfavorable outcome by 45%-99% (HR: 1.45-1.99, all <i>P</i><0.050), whereas ENR reduced the risk by 29% (HR: 0.71, <i>P</i>=0.040) compared to Q1. RCS curves revealed linear associations with unfavorable outcome that were positive for CAR, CLR, dNLR, SII, and SIRI, negative for ENR (all <i>P</i> for nonlinear>0.050), and nonlinear for MLR, NLR, and PNI (all <i>P</i> for nonlinear<0.050). Subgroup analyses identified heterogeneous associations across age, sex, BMI, comorbidities, and drug resistance (all <i>P</i> for interaction<0.050), with attenuated risk effects of CAR, CLR, dNLR, and SII in patients aged 30-60 years, male, BMI≥24.0 kg/m², smokers, retreatment cases, and those with tumor. ROC analysis demonstrated stable predictive performances of inflammatory status (AUC: 0.785-0.804 at 6-month, 0.781-0.793 at 9-month, and 0.762-0.773 at 12-month), and the combination of the inflammatory status significantly optimized the prognostic performance of the basic model (9-month AUC: 0.811 vs 0.780, <i>P</i>=0.024; 12-month AUC: 0.794 vs 0.758, <i>P</i>=0.013).</p><p><strong>Conclusion: </strong>Pretreatment blood inflammatory status effectively predicts the treatment outcome of TB patients. Our findings hold significant clinical value for TB patient management and warrant prospective evaluation in future studies.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1556857"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968758/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795244","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1500997
Piia Karisola, Mari Kanerva, Aki Vuokko, Helena Liira, Shuyuan Wang, Kirsi Kvarnström, Mikko Varonen, Hille Suojalehto, Harri Alenius
{"title":"Patients with post-COVID-19 condition show minor blood transcriptomic changes, with altered erythrocyte gene expression in a male subgroup.","authors":"Piia Karisola, Mari Kanerva, Aki Vuokko, Helena Liira, Shuyuan Wang, Kirsi Kvarnström, Mikko Varonen, Hille Suojalehto, Harri Alenius","doi":"10.3389/fimmu.2025.1500997","DOIUrl":"10.3389/fimmu.2025.1500997","url":null,"abstract":"<p><strong>Background: </strong>The mechanisms underlying persistent symptoms after non-severe COVID-19 remain unclear. This study aimed to investigate transcriptomic changes in peripheral blood cells of patients with post-COVID-19 condition (PCC) and assess if distinct clinical subtypes with specific gene signatures could be identified.</p><p><strong>Methods: </strong>The cohort included 111 PCC patients from the SARS-CoV-2 Omicron variant era, with 57 recovered (Recov) and 54 having prolonged symptoms indicative of PCC. The results were compared to 63 healthy controls (Ctrl) without known SARS-CoV-2 infection. Clinical data included patient assessments, laboratory results, comorbidities, and questionnaires on quality of life and functioning. Transcriptomic analysis and cellular deconvolution methods were used on total RNA from peripheral blood mononuclear cells (PBMCs).</p><p><strong>Results: </strong>PCC patients had more comorbidities (mean 1.3) and more frequently (59%) at least one comorbidity than recovered patients (31%) and controls (24%). Overall, past COVID-19 illness or current PCC symptoms caused minimal changes in the blood cell transcriptome, with only 3-6 differentially expressed genes (DEGs) identified across comparisons. However, a subset of male PCC patients exhibited an increased fraction of deconvoluted erythroblasts and significant genome-wide gene expression changes, with 399 DEGs compared to recovered and control males. These genes were enriched in pathways related to heme metabolism and gas exchange in erythrocytes.</p><p><strong>Conclusions: </strong>Persistent symptoms in PCC are multifactorial and not directly linked to peripheral blood cell gene expression changes. However, a subgroup of male PCC patients shows distinct erythrocyte responses that may contribute to long-term symptoms.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1500997"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968430/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Assessing the impact of probiotics on immunotherapy effectiveness and antibiotic-mediated resistance in cancer: a systematic review and meta-analysis.","authors":"Shuya Zhao, Zian Lu, Fangmin Zhao, Shihuan Tang, Lishan Zhang, Cuiling Feng","doi":"10.3389/fimmu.2025.1538969","DOIUrl":"10.3389/fimmu.2025.1538969","url":null,"abstract":"<p><strong>Background: </strong>Probiotics have been demonstrated to exert a potential clinical enhancing effect in cancer patients receiving immune checkpoint inhibitors (ICIs), while antibiotics exert a detrimental impact. Prior meta-analysis papers have substantial limitations and are devoid of recent published studies. Therefore, this study aimed to perform an updated meta-analysis and, for the first time, assess whether probiotics can restore the damage of antibiotics to immunotherapy.</p><p><strong>Methods: </strong>A comprehensive literature search was conducted in three English databases and three Chinese databases with a cutoff date of August 11, 2024. The methodological quality of the studies was evaluated using the Newcastle-Ottawa Quality Assessment Scale (NOS) or the Revised Cochrane risk-of-bias tool (RoB 2). Engauge Digitizer v12.1 was employed to extract hazard ratios (HRs) with 95% confidence interval (CI) for survival outcomes when these data were not explicitly provided in the manuscripts. Meta-analysis was conducted using Stata 14 software.</p><p><strong>Results: </strong>The study sample comprised eight retrospective and four prospective studies, involving a total of 3,142 participants. The findings indicate that probiotics significantly prolong the overall survival (OS) (I<sup>2</sup> = 31.2%; HR=0.58, 95% CI: 0.46-0.73, p < 0.001) and progression-free survival (PFS) (I<sup>2</sup> = 65.2%; HR=0.66, 95% CI: 0.54-0.81, p < 0.001) in cancer patients receiving ICIs, enhance the objective response rate (ORR) (I<sup>2</sup> = 33.5%; OR=1.75, 95% CI: 1.27-2.40, p = 0.001) and disease control rate (DCR) (I<sup>2</sup> = 50.0%; OR=1.93, 95% CI: 1.11-3.35, p = 0.002). For non-small cell lung cancer (NSCLC) patients exposed to antibiotics, the use of probiotics was associated with superior OS (I<sup>2</sup> = 0.0%; HR=0.45, 95% CI: 0.34-0.59, p < 0.001) and PFS (I<sup>2</sup> = 0.0%; HR=0.48, 95% CI: 0.38-0.62, p < 0.001) when compared to non-users. Subgroup differences were observed regarding the cancer type (P=0.006) and ethnic backgrounds (P=0.011) in OS.</p><p><strong>Conclusions: </strong>The meta-analysis findings suggest that probiotics can effectively extend the survival of cancer treated with ICIs. In NSCLC, probiotics appear to mitigate the negative impact of antibiotics on immunotherapy effectiveness, which has profound clinical significance. Nevertheless, additional large-scale, high-quality randomized controlled trials are necessary to further validate these findings.</p><p><strong>Systematic review registration: </strong>https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=579047, identifier CRD42024579047.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1538969"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968366/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in ImmunologyPub Date : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fimmu.2025.1530305
Aly Ismailov, Aldo Spallone, Alexey Belogurov, Alan Herbert, Maria Poptsova
{"title":"Molecular biology of the deadliest cancer - glioblastoma: what do we know?","authors":"Aly Ismailov, Aldo Spallone, Alexey Belogurov, Alan Herbert, Maria Poptsova","doi":"10.3389/fimmu.2025.1530305","DOIUrl":"10.3389/fimmu.2025.1530305","url":null,"abstract":"<p><p>Glioblastomas are the most prevalent primary brain tumors and are associated with a dramatically poor prognosis. Despite an intensive treatment approach, including maximal surgical tumor removal followed by radio- and chemotherapy, the median survival for glioblastoma patients has remained around 18 months for decades. Glioblastoma is distinguished by its highly complex mechanisms of immune evasion and pronounced heterogeneity. This variability is apparent both within the tumor itself, which can exhibit multiple phenotypes simultaneously, and in its surrounding microenvironment. Another key feature of glioblastoma is its \"cold\" microenvironment, characterized by robust immunosuppression. Recent advances in single-cell RNA sequencing have uncovered new promising insights, revealing previously unrecognized aspects of this tumor. In this review, we consolidate current knowledge on glioblastoma cells and its microenvironment, with an emphasis on their biological properties and unique patterns of molecular communication through signaling pathways. The evidence underscores the critical need for personalized poly-immunotherapy and other approaches to overcome the plasticity of glioblastoma stem cells. Analyzing the tumor microenvironment of individual patients using single-cell transcriptomics and implementing a customized immunotherapeutic strategy could potentially improve survival outcomes for those facing this formidable disease.</p>","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":"16 ","pages":"1530305"},"PeriodicalIF":5.7,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795425","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}