首页 > 最新文献

Cancer Immunology, Immunotherapy最新文献

英文 中文
Revealing the role of regulatory b cells in cancer: development, function and treatment significance. 揭示调节性b细胞在癌症中的作用:发展、功能和治疗意义。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03973-w
Ruyu Ye, Sijia Li, Yuxiao Li, Kaixin Shi, Li Li

B cells are essential components of the immune response, primarily recognized for their ability to produce antibodies. However, emerging research reveals their important roles in regulating immune responses and influencing tumor development, independent of antibodies. The connection between tumor progression and alterations in the tumor microenvironment is well-established, as immune infiltrating cells can enhance the survival of tumor cells by modifying their surroundings. Despite this, the majority of studies have focused on T cells and macrophages, creating a gap in our understanding of B cells. Regulatory B cells (Bregs) represent a crucial subpopulation that plays a significant role in maintaining immune balance. They may have a substantial impact on tumor immunity by negatively regulating tumor-infiltrating immune cells. This paper reviews the existing literature on Bregs, examining their development, phenotypes, functions, and the mechanisms through which they exert their regulatory effects. Furthermore, we highlight their potential interventional roles and prognostic significance in cancer therapy. By addressing the current gaps in knowledge regarding Bregs within tumors, we hope to inspire further research that could lead to innovative cancer treatments and improved outcomes for patients.

B细胞是免疫反应的重要组成部分,主要因其产生抗体的能力而被认可。然而,新兴的研究揭示了它们在调节免疫反应和影响肿瘤发展方面的重要作用,独立于抗体。肿瘤进展与肿瘤微环境改变之间的联系已经确立,因为免疫浸润细胞可以通过改变其环境来增强肿瘤细胞的存活。尽管如此,大多数研究都集中在T细胞和巨噬细胞上,这给我们对B细胞的理解带来了空白。调节性B细胞(Bregs)是一个重要的亚群,在维持免疫平衡中起着重要作用。它们可能通过负向调节肿瘤浸润性免疫细胞对肿瘤免疫产生实质性影响。本文就Bregs的发育、表型、功能及其调控机制进行综述。此外,我们强调了它们在癌症治疗中的潜在介入作用和预后意义。通过解决目前关于肿瘤内Bregs的知识差距,我们希望能激发进一步的研究,从而带来创新的癌症治疗方法,改善患者的预后。
{"title":"Revealing the role of regulatory b cells in cancer: development, function and treatment significance.","authors":"Ruyu Ye, Sijia Li, Yuxiao Li, Kaixin Shi, Li Li","doi":"10.1007/s00262-025-03973-w","DOIUrl":"10.1007/s00262-025-03973-w","url":null,"abstract":"<p><p>B cells are essential components of the immune response, primarily recognized for their ability to produce antibodies. However, emerging research reveals their important roles in regulating immune responses and influencing tumor development, independent of antibodies. The connection between tumor progression and alterations in the tumor microenvironment is well-established, as immune infiltrating cells can enhance the survival of tumor cells by modifying their surroundings. Despite this, the majority of studies have focused on T cells and macrophages, creating a gap in our understanding of B cells. Regulatory B cells (Bregs) represent a crucial subpopulation that plays a significant role in maintaining immune balance. They may have a substantial impact on tumor immunity by negatively regulating tumor-infiltrating immune cells. This paper reviews the existing literature on Bregs, examining their development, phenotypes, functions, and the mechanisms through which they exert their regulatory effects. Furthermore, we highlight their potential interventional roles and prognostic significance in cancer therapy. By addressing the current gaps in knowledge regarding Bregs within tumors, we hope to inspire further research that could lead to innovative cancer treatments and improved outcomes for patients.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"125"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861783/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490871","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}
引用次数: 0
Study on pathological factors affecting the sensitivity of neoadjuvant therapy in hypopharyngeal cancer. 影响下咽癌新辅助治疗敏感性的病理因素研究。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03957-w
Gaofei Yin, Nuan Li, Xiaohong Chen, Yang Zhang, Zhigang Huang, Wei Guo

Objective: Hypopharyngeal squamous cell carcinoma (HSCC) is a highly malignant tumor type in the head and neck region. In recent years, neoadjuvant therapy has significantly improved the laryngeal preservation rate among patients. However, there are variations in the efficacy of neoadjuvant therapy, and the identification of reliable predictive biomarkers for its efficacy remains a challenging research focus. This study aims to explore the correlation between the expression of P53, P16, and Ki67 and the response to neoadjuvant therapy.

Methods: A retrospective analysis was conducted on 120 patients who underwent neoadjuvant therapy at Beijing Tongren Hospital, Capital Medical University, from January 2021 to June 2024. Patients were grouped based on their treatment response and the type of neoadjuvant therapy received. Group analysis were employed to assess the predictive value of P53, P16, and Ki67 for treatment efficacy across different neoadjuvant therapy groups.

Results: A total of 120 patients were included in the study, with 60 patients each in the good response and poor response groups. The study comprised three neoadjuvant therapy groups: neoadjuvant chemotherapy, neoadjuvant targeted therapy combined with chemotherapy, and neoadjuvant immunotherapy combined with chemotherapy. Each group had 20 patients with good and poor responses, respectively. Among patients with P53 positivity, the effective rate after neoadjuvant therapy was 35.94%, significantly lower than the 66.07% observed in P53negative patients (p = 0.0017). For P16 positivity, the effective rate was 28.57%, lower than the 52.83% in P16negative patients (p = 0.0880). For patients with Ki67 < 60%, the effective rate was 33.33%, significantly lower than the 66.67% in those with Ki67 ≥ 60% (p = 0.0005). Comprehensively evaluate confirmed that P53 positivity and Ki67 < 60% were associated with poor response to neoadjuvant therapy, while P53 negativity and Ki67 ≥ 60% were associated with good response.

Conclusion: Our findings suggest that the combination of P53 and P16 can be used as a preliminary tool to assess the efficacy of neoadjuvant therapy in patients with HSCC. Specifically, patients with P53 positivity and Ki67 < 60% tend to have lower sensitivity to neoadjuvant therapy. Further research may be needed to clarify the precise role of P16 in HSCC.

目的:下咽鳞状细胞癌(HSCC)是一种发生在头颈部的高度恶性肿瘤。近年来,新辅助治疗显著提高了患者的喉保管率。然而,新辅助治疗的疗效存在差异,确定可靠的预测其疗效的生物标志物仍然是一个具有挑战性的研究焦点。本研究旨在探讨P53、P16和Ki67的表达与新辅助治疗反应的关系。方法:回顾性分析2021年1月至2024年6月在首都医科大学附属北京同仁医院接受新辅助治疗的120例患者。根据患者的治疗效果和接受的新辅助治疗类型对患者进行分组。采用分组分析的方法评估不同新辅助治疗组中P53、P16和Ki67对治疗效果的预测价值。结果:共纳入120例患者,反应良好组和反应不良组各60例。本研究分为新辅助化疗、新辅助靶向治疗联合化疗、新辅助免疫治疗联合化疗三个新辅助治疗组。每组分别有20例反应良好和较差的患者。在P53阳性患者中,新辅助治疗后的有效率为35.94%,显著低于P53阴性患者66.07% (p = 0.0017)。P16阳性组有效率为28.57%,低于P16阴性组的52.83% (p = 0.0880)。结论:我们的研究结果表明P53和P16的联合可以作为评估hsc患者新辅助治疗效果的初步工具。具体来说,P53阳性和Ki67的患者
{"title":"Study on pathological factors affecting the sensitivity of neoadjuvant therapy in hypopharyngeal cancer.","authors":"Gaofei Yin, Nuan Li, Xiaohong Chen, Yang Zhang, Zhigang Huang, Wei Guo","doi":"10.1007/s00262-025-03957-w","DOIUrl":"10.1007/s00262-025-03957-w","url":null,"abstract":"<p><strong>Objective: </strong>Hypopharyngeal squamous cell carcinoma (HSCC) is a highly malignant tumor type in the head and neck region. In recent years, neoadjuvant therapy has significantly improved the laryngeal preservation rate among patients. However, there are variations in the efficacy of neoadjuvant therapy, and the identification of reliable predictive biomarkers for its efficacy remains a challenging research focus. This study aims to explore the correlation between the expression of P53, P16, and Ki67 and the response to neoadjuvant therapy.</p><p><strong>Methods: </strong>A retrospective analysis was conducted on 120 patients who underwent neoadjuvant therapy at Beijing Tongren Hospital, Capital Medical University, from January 2021 to June 2024. Patients were grouped based on their treatment response and the type of neoadjuvant therapy received. Group analysis were employed to assess the predictive value of P53, P16, and Ki67 for treatment efficacy across different neoadjuvant therapy groups.</p><p><strong>Results: </strong>A total of 120 patients were included in the study, with 60 patients each in the good response and poor response groups. The study comprised three neoadjuvant therapy groups: neoadjuvant chemotherapy, neoadjuvant targeted therapy combined with chemotherapy, and neoadjuvant immunotherapy combined with chemotherapy. Each group had 20 patients with good and poor responses, respectively. Among patients with P53 positivity, the effective rate after neoadjuvant therapy was 35.94%, significantly lower than the 66.07% observed in P53negative patients (p = 0.0017). For P16 positivity, the effective rate was 28.57%, lower than the 52.83% in P16negative patients (p = 0.0880). For patients with Ki67 < 60%, the effective rate was 33.33%, significantly lower than the 66.67% in those with Ki67 ≥ 60% (p = 0.0005). Comprehensively evaluate confirmed that P53 positivity and Ki67 < 60% were associated with poor response to neoadjuvant therapy, while P53 negativity and Ki67 ≥ 60% were associated with good response.</p><p><strong>Conclusion: </strong>Our findings suggest that the combination of P53 and P16 can be used as a preliminary tool to assess the efficacy of neoadjuvant therapy in patients with HSCC. Specifically, patients with P53 positivity and Ki67 < 60% tend to have lower sensitivity to neoadjuvant therapy. Further research may be needed to clarify the precise role of P16 in HSCC.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"118"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861825/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490886","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}
引用次数: 0
Microsatellite instability-high status as a pan-cancer biomarker for immunotherapy efficacy. 微卫星不稳定性-作为免疫治疗疗效的泛癌生物标志物的高地位。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03980-x
Thierry Landre, Gaëtan Des Guetz

Background: Microsatellite instability-high (MSI-H) cancers are linked to exceptional benefit from immune checkpoint inhibitors (ICIs), but studies on their efficacy across various MSI-H cancer types are limited.

Methods: Randomized clinical trials (RCTs) comparing ICIs to chemotherapy in advanced MSI-H/dMMR cancers were systematically reviewed. Eligible studies included 13 RCTs with 1633 MSI-H patients across colorectal, gastric, and endometrial cancers. Data were analyzed using hazard ratios for progression-free survival (PFS) and overall survival (OS), with subgroup analyses by tumor type. Statistical heterogeneity was assessed using Cochrane's Q and I2.

Results: Immunotherapy significantly improved PFS and OS in MSI-H patients, with an HR for OS of 0.35 (95% CI 0.27-0.46; p < 0.00001) versus 0.81 for MSS patients. PFS showed a 64% reduced risk of progression (HR = 0.36, 95% CI 0.28-0.46; p < 0.0001). Subgroup analyses highlighted PFS benefits across tumor types: colorectal (HR = 0.28, 95% CI 0.11-0.73), gastric (HR = 0.43, 95% CI 0.27-0.68), and endometrial cancers (HR = 0.34, 95% CI 0.27-0.42).

Conclusions: This meta-analysis establishes MSI-H as a predictive biomarker for ICIs, supporting its role in therapy selection and underscoring the need for MSI-H/dMMR-focused clinical trials.

背景:微卫星不稳定性高(MSI-H)癌症与免疫检查点抑制剂(ICIs)的特殊益处有关,但对其在各种MSI-H癌症类型中的疗效的研究有限。方法:系统回顾比较晚期MSI-H/dMMR癌症中ICIs与化疗的随机临床试验(rct)。符合条件的研究包括13项随机对照试验,包括1633例结肠直肠癌、胃癌和子宫内膜癌的MSI-H患者。数据分析采用无进展生存期(PFS)和总生存期(OS)的风险比,并按肿瘤类型进行亚组分析。采用Cochrane's Q和I2评估统计异质性。结果:免疫治疗显著改善MSI-H患者的PFS和OS, OS的HR为0.35 (95% CI 0.27-0.46;结论:本荟萃分析确立了MSI-H作为ICIs的预测性生物标志物,支持其在治疗选择中的作用,并强调了以MSI-H/ dmmr为重点的临床试验的必要性。
{"title":"Microsatellite instability-high status as a pan-cancer biomarker for immunotherapy efficacy.","authors":"Thierry Landre, Gaëtan Des Guetz","doi":"10.1007/s00262-025-03980-x","DOIUrl":"10.1007/s00262-025-03980-x","url":null,"abstract":"<p><strong>Background: </strong>Microsatellite instability-high (MSI-H) cancers are linked to exceptional benefit from immune checkpoint inhibitors (ICIs), but studies on their efficacy across various MSI-H cancer types are limited.</p><p><strong>Methods: </strong>Randomized clinical trials (RCTs) comparing ICIs to chemotherapy in advanced MSI-H/dMMR cancers were systematically reviewed. Eligible studies included 13 RCTs with 1633 MSI-H patients across colorectal, gastric, and endometrial cancers. Data were analyzed using hazard ratios for progression-free survival (PFS) and overall survival (OS), with subgroup analyses by tumor type. Statistical heterogeneity was assessed using Cochrane's Q and I<sup>2</sup>.</p><p><strong>Results: </strong>Immunotherapy significantly improved PFS and OS in MSI-H patients, with an HR for OS of 0.35 (95% CI 0.27-0.46; p < 0.00001) versus 0.81 for MSS patients. PFS showed a 64% reduced risk of progression (HR = 0.36, 95% CI 0.28-0.46; p < 0.0001). Subgroup analyses highlighted PFS benefits across tumor types: colorectal (HR = 0.28, 95% CI 0.11-0.73), gastric (HR = 0.43, 95% CI 0.27-0.68), and endometrial cancers (HR = 0.34, 95% CI 0.27-0.42).</p><p><strong>Conclusions: </strong>This meta-analysis establishes MSI-H as a predictive biomarker for ICIs, supporting its role in therapy selection and underscoring the need for MSI-H/dMMR-focused clinical trials.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"122"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861458/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490867","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}
引用次数: 0
CDX2 and SATB2 loss are associated with myeloid cell infiltration and poor survival in colorectal cancer. CDX2和SATB2的缺失与结直肠癌骨髓细胞浸润和生存不良相关。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03964-x
Päivi Sirniö, Hanna Elomaa, Anne Tuomisto, Ville K Äijälä, Henna Karjalainen, Meeri Kastinen, Vilja V Tapiainen, Onni Sirkiä, Maarit Ahtiainen, Olli Helminen, Erkki-Ville Wirta, Jukka Rintala, Sanna Meriläinen, Juha Saarnio, Tero Rautio, Toni T Seppälä, Jan Böhm, Jukka-Pekka Mecklin, Markus J Mäkinen, Juha P Väyrynen

Background: Caudal-type homeobox 2 (CDX2) and special AT-rich sequence-binding protein 2 (SATB2) are transcription factors playing important roles in intestinal homeostasis and participating in the regulation of intestinal inflammation. In colorectal cancer (CRC), reduced expression levels of CDX2 and SATB2 have been associated with poor differentiation and worse survival. However, their prognostic significance still needs further clarification, and the associations between CDX2 and SATB2 and immune cell infiltration into the CRC microenvironment are largely unknown.

Methods: We analyzed CDX2 and SATB2 expression in two large cohorts of stages I-IV CRC patients (N = 2302) and analyzed their associations with clinicopathologic parameters, the density of local immune cells (determined with three multiplex immunohistochemistry panels and conventional immunohistochemistry), and survival.

Results: In mismatch repair-proficient tumors, reduced CDX2 and SATB2 expression were associated with higher densities of immature monocytic cells, macrophages, and M2-like macrophages. Low expression of CDX2 was associated with shorter cancer-specific survival independent of conventional prognostic parameters in both cohorts. In the larger cohort, adjusted hazard ratio (HR) for negative (vs. high) CDX2 expression was 3.62 (95% CI 2.08-6.31, ptrend < 0.0001), and adjusted HR for negative (vs. high) SATB2 level was 1.61 (95% CI 0.97-2.67, ptrend = 0.002).

Conclusion: This study indicates that reduced CDX2 and SATB2 expression levels are associated with myeloid cell infiltration in the CRC microenvironment and represent markers for poor outcome. These findings highlight the potential of CDX2 and SATB2 as biomarkers for classifying CRC patients and support their role in regulating the tumor microenvironment.

背景:尾型同源盒2 (CDX2)和特殊AT-rich sequence-binding protein 2 (SATB2)是在肠道内稳态和参与肠道炎症调控中起重要作用的转录因子。在结直肠癌(CRC)中,CDX2和SATB2表达水平降低与分化不良和生存率降低相关。然而,它们的预后意义仍需要进一步澄清,CDX2和SATB2与免疫细胞浸润CRC微环境之间的关系在很大程度上是未知的。方法:我们分析了两个大队列I-IV期CRC患者(N = 2302)的CDX2和SATB2表达,并分析了它们与临床病理参数、局部免疫细胞密度(通过三个多重免疫组化小组和常规免疫组化小组测定)和生存的关系。结果:在错配修复熟练的肿瘤中,CDX2和SATB2表达的降低与未成熟单核细胞、巨噬细胞和m2样巨噬细胞的高密度相关。在两个队列中,CDX2的低表达与较短的癌症特异性生存相关,与传统预后参数无关。在更大的队列中,CDX2阴性(vs.高)表达的校正风险比(HR)为3.62 (95% CI 2.08-6.31, ptrend趋势= 0.002)。结论:本研究表明,CDX2和SATB2表达水平降低与CRC微环境中髓系细胞浸润有关,是预后不良的标志。这些发现突出了CDX2和SATB2作为结直肠癌患者分类生物标志物的潜力,并支持它们在调节肿瘤微环境中的作用。
{"title":"CDX2 and SATB2 loss are associated with myeloid cell infiltration and poor survival in colorectal cancer.","authors":"Päivi Sirniö, Hanna Elomaa, Anne Tuomisto, Ville K Äijälä, Henna Karjalainen, Meeri Kastinen, Vilja V Tapiainen, Onni Sirkiä, Maarit Ahtiainen, Olli Helminen, Erkki-Ville Wirta, Jukka Rintala, Sanna Meriläinen, Juha Saarnio, Tero Rautio, Toni T Seppälä, Jan Böhm, Jukka-Pekka Mecklin, Markus J Mäkinen, Juha P Väyrynen","doi":"10.1007/s00262-025-03964-x","DOIUrl":"10.1007/s00262-025-03964-x","url":null,"abstract":"<p><strong>Background: </strong>Caudal-type homeobox 2 (CDX2) and special AT-rich sequence-binding protein 2 (SATB2) are transcription factors playing important roles in intestinal homeostasis and participating in the regulation of intestinal inflammation. In colorectal cancer (CRC), reduced expression levels of CDX2 and SATB2 have been associated with poor differentiation and worse survival. However, their prognostic significance still needs further clarification, and the associations between CDX2 and SATB2 and immune cell infiltration into the CRC microenvironment are largely unknown.</p><p><strong>Methods: </strong>We analyzed CDX2 and SATB2 expression in two large cohorts of stages I-IV CRC patients (N = 2302) and analyzed their associations with clinicopathologic parameters, the density of local immune cells (determined with three multiplex immunohistochemistry panels and conventional immunohistochemistry), and survival.</p><p><strong>Results: </strong>In mismatch repair-proficient tumors, reduced CDX2 and SATB2 expression were associated with higher densities of immature monocytic cells, macrophages, and M2-like macrophages. Low expression of CDX2 was associated with shorter cancer-specific survival independent of conventional prognostic parameters in both cohorts. In the larger cohort, adjusted hazard ratio (HR) for negative (vs. high) CDX2 expression was 3.62 (95% CI 2.08-6.31, p<sub>trend</sub> < 0.0001), and adjusted HR for negative (vs. high) SATB2 level was 1.61 (95% CI 0.97-2.67, p<sub>trend </sub>= 0.002).</p><p><strong>Conclusion: </strong>This study indicates that reduced CDX2 and SATB2 expression levels are associated with myeloid cell infiltration in the CRC microenvironment and represent markers for poor outcome. These findings highlight the potential of CDX2 and SATB2 as biomarkers for classifying CRC patients and support their role in regulating the tumor microenvironment.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"111"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861821/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490772","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}
引用次数: 0
Comparison of neoadjuvant chemoimmunotherapy with planned surgery and concurrent chemoradiation followed by immunotherapy for potentially resectable stage III non-small-cell lung cancer: a retrospective study. 对可能切除的III期非小细胞肺癌,新辅助化疗免疫治疗与计划手术和同步放化疗后免疫治疗的比较:一项回顾性研究。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03961-0
Yana Qi, Xiaoyang Zhai, Qinhao Xu, Yuqin Jin, Yingfan Guo, Miaoqing Zhao, Hui Zhu, Hongbo Guo

Objective: Despite the promising potential of neoadjuvant chemoimmunotherapy for non-small cell lung cancer (NSCLC), there is limited consensus on the optimal treatment strategy for potentially resectable NSCLC. This study aimed to evaluate the efficacy and safety of neoadjuvant chemoimmunotherapy (neoCT/IO) with planned surgery versus definitive concurrent chemoradiation followed by immunotherapy (cCRT + IO) in potentially resectable stage III NSCLC.

Methods: This retrospective study analyzed data from patients with potentially resectable stage III NSCLC who underwent neoCT/IO with planned surgery or cCRT + IO between March 2020 and June 2023. Propensity score matching (PSM) was used to balance heterogeneity between groups. Efficacy outcomes, safety profiles and patterns of disease recurrence were assessed.

Results: A total of 308 eligible patients were included in this study, of whom 195 (63.3%) underwent neoCT/IO and 113 (36.7%) received cCRT + IO. The neoCT/IO group consisted of patients who underwent neoCT/IO + Surgery and neoCT/IO + Radiotherapy. After 1:1 PSM, each group consisted of 105 patients. The median progression-free survival (PFS) was 25.9 months in the cCRT + IO group and not reached (NR) in the neoCT/IO group (hazard ratio: 2.91, 95% confidence interval: 1.77-4.78; p < 0.001). Median overall survival (OS) was NR in either group, with 3-year OS rates of 87.5% in the neoCT/IO group and 75.0% in the cCRT + IO group (p = 0.22). The incidence of grade 3/4 treatment-related adverse events was similar in both groups, except for a higher incidence of grade 3/4 hematological toxicity in the cCRT + IO group.

Conclusions: For patients with potentially resectable stage III NSCLC, neoCT/IO appears to be a safe approach and may offer better survival outcomes compared with cCRT + IO. Prospective randomized trials are needed to further validate these findings.

目的:尽管新辅助化疗免疫治疗非小细胞肺癌(NSCLC)的潜力很大,但对于可能可切除的非小细胞肺癌的最佳治疗策略,人们的共识有限。本研究旨在评估计划手术的新辅助化疗免疫治疗(neoCT/IO)与确定同步放化疗后免疫治疗(cCRT + IO)在潜在可切除的III期NSCLC中的疗效和安全性。方法:本回顾性研究分析了2020年3月至2023年6月期间接受neoCT/IO计划手术或cCRT + IO的可能可切除的III期NSCLC患者的数据。倾向得分匹配(PSM)用于平衡组间异质性。评估了疗效、结果、安全性和疾病复发模式。结果:本研究共纳入308例符合条件的患者,其中195例(63.3%)接受了neoCT/IO, 113例(36.7%)接受了cCRT + IO。neoCT/IO组由接受neoCT/IO +手术和neoCT/IO +放疗的患者组成。经1:1 PSM后,每组105例。cCRT + IO组的中位无进展生存期(PFS)为25.9个月,neoCT/IO组未达到(NR)(风险比:2.91,95%可信区间:1.77-4.78;结论:对于可能可切除的III期NSCLC患者,neoCT/IO似乎是一种安全的方法,与cCRT + IO相比,可能提供更好的生存结果。需要前瞻性随机试验来进一步验证这些发现。
{"title":"Comparison of neoadjuvant chemoimmunotherapy with planned surgery and concurrent chemoradiation followed by immunotherapy for potentially resectable stage III non-small-cell lung cancer: a retrospective study.","authors":"Yana Qi, Xiaoyang Zhai, Qinhao Xu, Yuqin Jin, Yingfan Guo, Miaoqing Zhao, Hui Zhu, Hongbo Guo","doi":"10.1007/s00262-025-03961-0","DOIUrl":"10.1007/s00262-025-03961-0","url":null,"abstract":"<p><strong>Objective: </strong>Despite the promising potential of neoadjuvant chemoimmunotherapy for non-small cell lung cancer (NSCLC), there is limited consensus on the optimal treatment strategy for potentially resectable NSCLC. This study aimed to evaluate the efficacy and safety of neoadjuvant chemoimmunotherapy (neoCT/IO) with planned surgery versus definitive concurrent chemoradiation followed by immunotherapy (cCRT + IO) in potentially resectable stage III NSCLC.</p><p><strong>Methods: </strong>This retrospective study analyzed data from patients with potentially resectable stage III NSCLC who underwent neoCT/IO with planned surgery or cCRT + IO between March 2020 and June 2023. Propensity score matching (PSM) was used to balance heterogeneity between groups. Efficacy outcomes, safety profiles and patterns of disease recurrence were assessed.</p><p><strong>Results: </strong>A total of 308 eligible patients were included in this study, of whom 195 (63.3%) underwent neoCT/IO and 113 (36.7%) received cCRT + IO. The neoCT/IO group consisted of patients who underwent neoCT/IO + Surgery and neoCT/IO + Radiotherapy. After 1:1 PSM, each group consisted of 105 patients. The median progression-free survival (PFS) was 25.9 months in the cCRT + IO group and not reached (NR) in the neoCT/IO group (hazard ratio: 2.91, 95% confidence interval: 1.77-4.78; p < 0.001). Median overall survival (OS) was NR in either group, with 3-year OS rates of 87.5% in the neoCT/IO group and 75.0% in the cCRT + IO group (p = 0.22). The incidence of grade 3/4 treatment-related adverse events was similar in both groups, except for a higher incidence of grade 3/4 hematological toxicity in the cCRT + IO group.</p><p><strong>Conclusions: </strong>For patients with potentially resectable stage III NSCLC, neoCT/IO appears to be a safe approach and may offer better survival outcomes compared with cCRT + IO. Prospective randomized trials are needed to further validate these findings.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"119"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861850/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490773","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}
引用次数: 0
Continuous immunotherapy beyond disease progression in patients with advanced non-small cell and small cell lung cancer. 晚期非小细胞和小细胞肺癌患者疾病进展后的持续免疫治疗
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03958-9
Jing Cheng, Wenwen Kang, Yueying Chen, Luyun Pan, Hedong Han, Tangfeng Lv

Background: The benefits of continuing immunotherapy beyond disease progression in advanced non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) remain uncertain, along with the specific patient subgroups that may gain the most from this approach. This retrospective study aims to evaluate the efficacy of this approach and identify target patient populations likely to benefit.

Methods: We collected data from patients with NSCLC and SCLC who experienced disease progression following initial immune checkpoint inhibitor (ICI) treatment from January 2020 to December 2023. Patients were categorized based on second-line treatment: those receiving immunotherapy beyond progression (IBP) and those receiving non-immunotherapy beyond progression (NIBP). Survival outcomes and treatment safety were compared between these two groups.

Results: A total of 150 patients were included, with 111 NSCLC patients (IBP: n = 78, NIBP: n = 33) and 39 SCLC patients (IBP: n = 31, NIBP: n = 8). Significant differences in median progression-free survival (PFS) and overall survival (OS) were found in patients with driver gene-negative NSCLC (mPFS: 4.7 vs 1.3 months, HR = 0.29, P < 0.01; mOS: 11.03 vs 2.63 months, HR = 0.13, P < 0.001) and SCLC (mPFS: 3.9 vs 2.1 months, HR = 0.38, P = 0.02; mOS: 9.28 vs 2.27 months, HR = 0.23, P < 0.01). Additionally, among driver gene-negative NSCLC patients, achieving a partial response (PR) or stable disease (SD) during initial immunotherapy was associated with improved effectiveness of continued immunotherapy beyond progression.

Conclusions: Continued immunotherapy as a second-line treatment may benefit patients with driver gene-negative NSCLC and SCLC who have progressed after initial immunotherapy.

背景:在晚期非小细胞肺癌(NSCLC)和小细胞肺癌(SCLC)中,持续免疫治疗超越疾病进展的益处仍然不确定,以及可能从这种方法中获益最多的特定患者亚组。这项回顾性研究旨在评估这种方法的疗效,并确定可能受益的目标患者群体。方法:我们收集了2020年1月至2023年12月期间接受初始免疫检查点抑制剂(ICI)治疗后出现疾病进展的NSCLC和SCLC患者的数据。患者根据二线治疗进行分类:接受进展后免疫治疗(IBP)的患者和接受进展后非免疫治疗(NIBP)的患者。比较两组的生存结局和治疗安全性。结果:共纳入150例患者,其中NSCLC患者111例(IBP: n = 78, NIBP: n = 33), SCLC患者39例(IBP: n = 31, NIBP: n = 8)。驱动基因阴性NSCLC患者的中位无进展生存期(PFS)和总生存期(OS)存在显著差异(mPFS: 4.7 vs 1.3个月,HR = 0.29, P)结论:持续免疫治疗作为二线治疗可能有利于驱动基因阴性NSCLC和SCLC患者,这些患者在初始免疫治疗后出现进展。
{"title":"Continuous immunotherapy beyond disease progression in patients with advanced non-small cell and small cell lung cancer.","authors":"Jing Cheng, Wenwen Kang, Yueying Chen, Luyun Pan, Hedong Han, Tangfeng Lv","doi":"10.1007/s00262-025-03958-9","DOIUrl":"10.1007/s00262-025-03958-9","url":null,"abstract":"<p><strong>Background: </strong>The benefits of continuing immunotherapy beyond disease progression in advanced non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) remain uncertain, along with the specific patient subgroups that may gain the most from this approach. This retrospective study aims to evaluate the efficacy of this approach and identify target patient populations likely to benefit.</p><p><strong>Methods: </strong>We collected data from patients with NSCLC and SCLC who experienced disease progression following initial immune checkpoint inhibitor (ICI) treatment from January 2020 to December 2023. Patients were categorized based on second-line treatment: those receiving immunotherapy beyond progression (IBP) and those receiving non-immunotherapy beyond progression (NIBP). Survival outcomes and treatment safety were compared between these two groups.</p><p><strong>Results: </strong>A total of 150 patients were included, with 111 NSCLC patients (IBP: n = 78, NIBP: n = 33) and 39 SCLC patients (IBP: n = 31, NIBP: n = 8). Significant differences in median progression-free survival (PFS) and overall survival (OS) were found in patients with driver gene-negative NSCLC (mPFS: 4.7 vs 1.3 months, HR = 0.29, P < 0.01; mOS: 11.03 vs 2.63 months, HR = 0.13, P < 0.001) and SCLC (mPFS: 3.9 vs 2.1 months, HR = 0.38, P = 0.02; mOS: 9.28 vs 2.27 months, HR = 0.23, P < 0.01). Additionally, among driver gene-negative NSCLC patients, achieving a partial response (PR) or stable disease (SD) during initial immunotherapy was associated with improved effectiveness of continued immunotherapy beyond progression.</p><p><strong>Conclusions: </strong>Continued immunotherapy as a second-line treatment may benefit patients with driver gene-negative NSCLC and SCLC who have progressed after initial immunotherapy.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"124"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861770/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490775","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}
引用次数: 0
Comprehensive analysis and experimental validation of disulfidptosis-associated prognostic signature and immune microenvironment characterization of gastric cancer. 胃癌二硫中毒相关预后特征及免疫微环境特征的综合分析与实验验证。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-024-03883-3
Huangjie Zhang, Jinguo Hu, Yuanqiang Li, Yanyang Liu, Huize Shen, Zeng Wang, Qinglin Li

Background: Gastric cancer (GC) is one of the most common causes of cancer-related death worldwide. As a novel form of programmed cell death, disulfidptosis is characterized by excessive cysteine accumulation, disulfide stress and actin destruction. There is evidence that targeting disulfidptosis is a promising anticancer strategy. Further improvement of GC risk stratification based on disulfidptosis has positive clinical significance.

Methods: We analyzed the expression levels of disulfidptosis-associated genes (DPAGs) in normal and GC tissues and characterized the molecular subtypes of GC patients. Based on the characteristics of DPAG subtypes, differentially expressed prognosis-related genes were selected by LASSO-univariate Cox analysis and multivariate Cox analysis analyzed to establish a prognostic model. Using single-cell sequencing analysis reveals the cell subpopulation for GC. The function of the selected target in GC was verified by in vitro experimental means, including siRNA, qRT-PCR, Western blot, CCK-8, and Transwell assay.

Results: DPAG score was verified to be an independent prognostic factor of GC and was significantly associated with poor prognosis of gastric cancer. Subsequent studies on subgroup immunoinfiltration characteristics, drug sensitivity analysis, immunotherapy response and somatic mutation characteristics of DPAG score comprehensively confirmed the potential guiding significance of DPAG score for individualized treatment of gastric cancer patients. Single-cell sequencing analysis revealed the expression characteristics of DPAG-related prognostic signatures across cell subpopulations. In vitro experiments showed APC11, as one of the selected DPAGs, was highly expressed in gastric cancer, and knockdown of APC11 could significantly inhibit the proliferation and migration of GC cells, demonstrating the reliability of bioinformatics results.

Conclusion: The results of this study provide a new perspective for exploring the role of disulfidptosis in the occurrence and development of GC.

背景:胃癌(GC)是世界范围内最常见的癌症相关死亡原因之一。作为一种新的程序性细胞死亡形式,二硫细胞凋亡的特点是过量的半胱氨酸积累、二硫应激和肌动蛋白破坏。有证据表明,靶向双曲下垂是一种很有前途的抗癌策略。进一步完善基于双上睑下垂的GC风险分层具有积极的临床意义。方法:分析正常组织和胃癌组织中二硫中毒相关基因(DPAGs)的表达水平,并对胃癌患者的分子亚型进行表征。根据DPAG亚型的特点,选择差异表达的预后相关基因进行lasso单因素Cox分析和多因素Cox分析,建立预后模型。使用单细胞测序分析揭示了GC的细胞亚群。通过siRNA、qRT-PCR、Western blot、CCK-8、Transwell等体外实验手段验证所选靶点在GC中的功能。结果:证实DPAG评分是胃癌的独立预后因素,且与胃癌的不良预后显著相关。后续对DPAG评分亚组免疫浸润特征、药物敏感性分析、免疫治疗反应、体细胞突变特征的研究,全面证实了DPAG评分对胃癌患者个体化治疗的潜在指导意义。单细胞测序分析揭示了dpag相关预后特征在细胞亚群中的表达特征。体外实验显示,APC11作为所选dpag之一,在胃癌中高表达,敲低APC11可显著抑制胃癌细胞的增殖和迁移,证明了生物信息学结果的可靠性。结论:本研究结果为探讨二睑下垂在胃癌发生发展中的作用提供了新的视角。
{"title":"Comprehensive analysis and experimental validation of disulfidptosis-associated prognostic signature and immune microenvironment characterization of gastric cancer.","authors":"Huangjie Zhang, Jinguo Hu, Yuanqiang Li, Yanyang Liu, Huize Shen, Zeng Wang, Qinglin Li","doi":"10.1007/s00262-024-03883-3","DOIUrl":"10.1007/s00262-024-03883-3","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) is one of the most common causes of cancer-related death worldwide. As a novel form of programmed cell death, disulfidptosis is characterized by excessive cysteine accumulation, disulfide stress and actin destruction. There is evidence that targeting disulfidptosis is a promising anticancer strategy. Further improvement of GC risk stratification based on disulfidptosis has positive clinical significance.</p><p><strong>Methods: </strong>We analyzed the expression levels of disulfidptosis-associated genes (DPAGs) in normal and GC tissues and characterized the molecular subtypes of GC patients. Based on the characteristics of DPAG subtypes, differentially expressed prognosis-related genes were selected by LASSO-univariate Cox analysis and multivariate Cox analysis analyzed to establish a prognostic model. Using single-cell sequencing analysis reveals the cell subpopulation for GC. The function of the selected target in GC was verified by in vitro experimental means, including siRNA, qRT-PCR, Western blot, CCK-8, and Transwell assay.</p><p><strong>Results: </strong>DPAG score was verified to be an independent prognostic factor of GC and was significantly associated with poor prognosis of gastric cancer. Subsequent studies on subgroup immunoinfiltration characteristics, drug sensitivity analysis, immunotherapy response and somatic mutation characteristics of DPAG score comprehensively confirmed the potential guiding significance of DPAG score for individualized treatment of gastric cancer patients. Single-cell sequencing analysis revealed the expression characteristics of DPAG-related prognostic signatures across cell subpopulations. In vitro experiments showed APC11, as one of the selected DPAGs, was highly expressed in gastric cancer, and knockdown of APC11 could significantly inhibit the proliferation and migration of GC cells, demonstrating the reliability of bioinformatics results.</p><p><strong>Conclusion: </strong>The results of this study provide a new perspective for exploring the role of disulfidptosis in the occurrence and development of GC.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"116"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861452/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490774","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}
引用次数: 0
Monitoring peripheral blood data supports the prediction of immunotherapy response in advanced non-small cell lung cancer based on real-world data. 监测外周血数据支持基于真实世界数据的晚期非小细胞肺癌免疫治疗反应预测。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03966-9
Ana D Ramos-Guerra, Benito Farina, Jaime Rubio Pérez, Anna Vilalta-Lacarra, Jon Zugazagoitia, Germán Peces-Barba, Luis M Seijo, Luis Paz-Ares, Ignacio Gil-Bazo, Manuel Dómine Gómez, María J Ledesma-Carbayo

The identification of non-small cell lung cancer (NSCLC) patients who will benefit from immunotherapy remains a clinical challenge. Monitoring real-world data (RWD) in the first cycles of therapy may provide a more accurate representation of response patterns in a real-world setting. We propose a multivariate Bayesian joint model using generalized linear mixed effects, trained and validated on RWD from 424 advanced NSCLC patients retrospectively collected from three clinical centers. Center1 was used as training ( N = 212 ), while Center2 and Center3 were used as independent testing sets ( N = 137 and N = 75 , respectively). Peripheral blood data (PBD) were collected at baseline and at three follow-up time points, alongside demographic and epidemiologic features. Six models were trained to predict progression-free survival at 6 months, PFS(6), using different number of longitudinal samples (baseline, two, or four time points) of the neutrophil-to-lymphocyte ratio (NLR) or a multivariate feature selection. Long-term predictions at 12 and 24 months were also evaluated. Prediction accuracy was measured using the area under the receiver operating characteristic curve (AUC). The proposed model significantly improved prediction performance, achieving AUCs of 0.870, 0.804 and 0.827 at 6, 12 and 24 months for Center2, and 0.824, 0.822 and 0.667 for Center3. There was also a significant difference in PFS and overall survival (OS) between predicted response groups, defined by a 6-month PFS cutoff (log-rank test p < 0.001 ). Our study suggests that the integration of multiple biomarkers and monitored PBD in an RWD-based Bayesian joint model framework significantly improves immunotherapy response prediction in advanced NSCLC compared to conventional approaches involving biomarker data at baseline only.

确定非小细胞肺癌(NSCLC)患者将受益于免疫治疗仍然是一个临床挑战。在治疗的第一个周期中监测真实世界数据(RWD)可以更准确地反映真实世界环境中的反应模式。我们提出了一个多元贝叶斯联合模型,使用广义线性混合效应,对来自三个临床中心的424名晚期非小细胞肺癌患者的RWD进行了训练和验证。Center1作为训练集(N = 212), Center2和Center3作为独立测试集(N = 137和N = 75)。在基线和三个随访时间点收集外周血数据(PBD),以及人口统计学和流行病学特征。使用中性粒细胞与淋巴细胞比率(NLR)的不同纵向样本数量(基线、2个或4个时间点)或多变量特征选择,训练6个模型来预测6个月时的无进展生存期(PFS)。对12个月和24个月的长期预测也进行了评估。预测精度采用受试者工作特征曲线下面积(AUC)测量。该模型显著提高了预测性能,Center2在6、12和24个月的auc分别为0.870、0.804和0.827,Center3在6、12和24个月的auc分别为0.824、0.822和0.667。预测缓解组之间的PFS和总生存期(OS)也有显著差异,由6个月PFS截止定义(log-rank检验p 0.001)。我们的研究表明,在基于rwd的贝叶斯联合模型框架中整合多种生物标志物和监测PBD,与仅涉及基线生物标志物数据的传统方法相比,可显著提高晚期非小细胞肺癌的免疫治疗反应预测。
{"title":"Monitoring peripheral blood data supports the prediction of immunotherapy response in advanced non-small cell lung cancer based on real-world data.","authors":"Ana D Ramos-Guerra, Benito Farina, Jaime Rubio Pérez, Anna Vilalta-Lacarra, Jon Zugazagoitia, Germán Peces-Barba, Luis M Seijo, Luis Paz-Ares, Ignacio Gil-Bazo, Manuel Dómine Gómez, María J Ledesma-Carbayo","doi":"10.1007/s00262-025-03966-9","DOIUrl":"10.1007/s00262-025-03966-9","url":null,"abstract":"<p><p>The identification of non-small cell lung cancer (NSCLC) patients who will benefit from immunotherapy remains a clinical challenge. Monitoring real-world data (RWD) in the first cycles of therapy may provide a more accurate representation of response patterns in a real-world setting. We propose a multivariate Bayesian joint model using generalized linear mixed effects, trained and validated on RWD from 424 advanced NSCLC patients retrospectively collected from three clinical centers. Center1 was used as training ( <math><mrow><mi>N</mi> <mo>=</mo> <mn>212</mn></mrow> </math> ), while Center2 and Center3 were used as independent testing sets ( <math><mrow><mi>N</mi> <mo>=</mo> <mn>137</mn></mrow> </math> and <math><mrow><mi>N</mi> <mo>=</mo> <mn>75</mn></mrow> </math> , respectively). Peripheral blood data (PBD) were collected at baseline and at three follow-up time points, alongside demographic and epidemiologic features. Six models were trained to predict progression-free survival at 6 months, PFS(6), using different number of longitudinal samples (baseline, two, or four time points) of the neutrophil-to-lymphocyte ratio (NLR) or a multivariate feature selection. Long-term predictions at 12 and 24 months were also evaluated. Prediction accuracy was measured using the area under the receiver operating characteristic curve (AUC). The proposed model significantly improved prediction performance, achieving AUCs of 0.870, 0.804 and 0.827 at 6, 12 and 24 months for Center2, and 0.824, 0.822 and 0.667 for Center3. There was also a significant difference in PFS and overall survival (OS) between predicted response groups, defined by a 6-month PFS cutoff (log-rank test <math><mrow><mi>p</mi> <mo><</mo> <mn>0.001</mn></mrow> </math> ). Our study suggests that the integration of multiple biomarkers and monitored PBD in an RWD-based Bayesian joint model framework significantly improves immunotherapy response prediction in advanced NSCLC compared to conventional approaches involving biomarker data at baseline only.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"120"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861465/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490869","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}
引用次数: 0
Exploring correlations between gut mycobiome and lymphocytes in melanoma patients undergoing anti-PD-1 therapy. 探索接受抗pd -1治疗的黑色素瘤患者肠道菌群和淋巴细胞之间的相关性。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-024-03918-9
Natalia Szóstak, Michał Budnik, Katarzyna Tomela, Luiza Handschuh, Anna Samelak-Czajka, Bernadeta Pietrzak, Marcin Schmidt, Mariusz Kaczmarek, Łukasz Galus, Jacek Mackiewicz, Andrzej Mackiewicz, Piotr Kozlowski, Anna Philips

Research has shown that the microbiome can influence how the immune system responds to melanoma cells, affecting the course of the disease and the outcome of the therapy. Here, we used the metagenomic approach and flow cytometry analyses of blood cells to discover correlations between gut fungi of metastatic melanoma patients enrolled in anti-PD-1 therapy and lymphocytes in their blood.We analyzed the patterns of associations before the first administration of anti-PD-1 therapy (BT, n = 61) and in the third month of the therapy (T3, n = 37), allowing us to track changes during treatment. To understand the possible impact of gut fungi on the efficacy of anti-PD-1 therapy, we analyzed the associations in clinical beneficiaries (CB, n = 37) and non-beneficiaries (NB, n = 24), as well as responders (R, n = 28) and non-responders (NR, n = 33).Patients with LDH < 338 units/L, overall survival (OS) > 12, CB, as well as R, had lower levels of Shannon diversity (p = 0.02, p = 0.05, p = 0.05, and p = 0.03, respectively). We found that the correlation pattern between intestinal fungi and lymphocytes was specific to the type of response, positive or negative. When comparing CB and NB groups, correlations with opposite directions were detected for C. albicans, suggesting a response-specific immune reaction. For CB, M. restricta exhibited a set of correlations with different types of lymphocytes, with prevalent positive correlations, suggesting a robust immune response in the CB group. This result extends our former research, where M. restricta and C. albicans were associated with an increased risk of melanoma progression and a poorer response to anti-PD-1 treatment.

研究表明,微生物组可以影响免疫系统对黑色素瘤细胞的反应,影响疾病的进程和治疗的结果。在这里,我们使用宏基因组方法和血细胞流式细胞术分析来发现参与抗pd -1治疗的转移性黑色素瘤患者的肠道真菌与其血液中的淋巴细胞之间的相关性。我们分析了首次给予抗pd -1治疗(BT, n = 61)之前和治疗的第三个月(T3, n = 37)的关联模式,使我们能够跟踪治疗期间的变化。为了了解肠道真菌对抗pd -1治疗疗效的可能影响,我们分析了临床受益者(CB, n = 37)和非受益者(NB, n = 24)以及应答者(R, n = 28)和无应答者(NR, n = 33)之间的相关性。LDH 12、CB、R患者Shannon多样性水平较低(p = 0.02、p = 0.05、p = 0.05、p = 0.03)。我们发现肠道真菌和淋巴细胞之间的相关模式是特定于反应类型的,阳性或阴性。当比较CB组和NB组时,白色念珠菌检测到相反方向的相关性,提示反应特异性免疫反应。对于CB, M. restricta与不同类型的淋巴细胞表现出一组相关性,普遍呈正相关,表明CB组存在强大的免疫应答。这一结果扩展了我们之前的研究,即限制性分枝杆菌和白色念珠菌与黑色素瘤进展风险增加和抗pd -1治疗反应较差相关。
{"title":"Exploring correlations between gut mycobiome and lymphocytes in melanoma patients undergoing anti-PD-1 therapy.","authors":"Natalia Szóstak, Michał Budnik, Katarzyna Tomela, Luiza Handschuh, Anna Samelak-Czajka, Bernadeta Pietrzak, Marcin Schmidt, Mariusz Kaczmarek, Łukasz Galus, Jacek Mackiewicz, Andrzej Mackiewicz, Piotr Kozlowski, Anna Philips","doi":"10.1007/s00262-024-03918-9","DOIUrl":"10.1007/s00262-024-03918-9","url":null,"abstract":"<p><p>Research has shown that the microbiome can influence how the immune system responds to melanoma cells, affecting the course of the disease and the outcome of the therapy. Here, we used the metagenomic approach and flow cytometry analyses of blood cells to discover correlations between gut fungi of metastatic melanoma patients enrolled in anti-PD-1 therapy and lymphocytes in their blood.We analyzed the patterns of associations before the first administration of anti-PD-1 therapy (BT, n = 61) and in the third month of the therapy (T3, n = 37), allowing us to track changes during treatment. To understand the possible impact of gut fungi on the efficacy of anti-PD-1 therapy, we analyzed the associations in clinical beneficiaries (CB, n = 37) and non-beneficiaries (NB, n = 24), as well as responders (R, n = 28) and non-responders (NR, n = 33).Patients with LDH < 338 units/L, overall survival (OS) > 12, CB, as well as R, had lower levels of Shannon diversity (p = 0.02, p = 0.05, p = 0.05, and p = 0.03, respectively). We found that the correlation pattern between intestinal fungi and lymphocytes was specific to the type of response, positive or negative. When comparing CB and NB groups, correlations with opposite directions were detected for C. albicans, suggesting a response-specific immune reaction. For CB, M. restricta exhibited a set of correlations with different types of lymphocytes, with prevalent positive correlations, suggesting a robust immune response in the CB group. This result extends our former research, where M. restricta and C. albicans were associated with an increased risk of melanoma progression and a poorer response to anti-PD-1 treatment.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"110"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861499/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490777","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}
引用次数: 0
From silent partners to potential therapeutic targets: macrophages in colorectal cancer. 从沉默伙伴到潜在治疗靶点:巨噬细胞在结直肠癌中的作用
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s00262-025-03965-w
Hayat Khizar, Kamran Ali, Jianwei Wang

Cancer cells grow and survive in the tumor microenvironment, which is a complicated process. As a key part of how colorectal cancer (CRC) progresses, tumor-associated macrophages (TAMs) exhibit a double role. Through angiogenesis, this TAM can promote the growth of cancers. Although being able to modify and adjust immune cells is a great advantage, these cells can also exhibit anti-cancer properties including direct killing of cancer cells, presenting antigens, and aiding T cell-mediated responses. The delicate regulatory mechanisms between the immune system and tumors are composed of a complex network of pathways regulated by several factors including hypoxia, metabolic reprogramming, cytokine/chemokine signaling, and cell interactions. Decoding and figuring out these complex systems become significant in building targeted treatment programs. Targeting TAMs in CRC involves disrupting chemokine signaling or adhesion molecules, reprogramming them to an anti-tumor phenotype using TLR agonists, CD40 agonists, or metabolic modulation, and selectively removing TAM subsets that promote tumor growth. Multi-drug resistance, the absence of an accurate biomarker, and drug non-specificity are also major problems. Combining macrophage-targeted therapies with chemotherapy and immunotherapy may revolutionize treatment. Macrophage studies will advance with new technology and multi-omics methodologies to help us understand CRC and build specific and efficient treatments.

癌细胞在肿瘤微环境中生长和生存是一个复杂的过程。作为结直肠癌(CRC)进展的关键部分,肿瘤相关巨噬细胞(tam)表现出双重作用。通过血管生成,这种TAM可以促进肿瘤的生长。虽然能够修饰和调节免疫细胞是一个很大的优势,但这些细胞也可以表现出抗癌特性,包括直接杀死癌细胞、呈递抗原和帮助T细胞介导的反应。免疫系统和肿瘤之间微妙的调节机制是由多种因素调控的复杂通路网络组成的,包括缺氧、代谢重编程、细胞因子/趋化因子信号传导和细胞相互作用。解码和弄清楚这些复杂的系统对于建立有针对性的治疗方案非常重要。靶向CRC中的TAM包括破坏趋化因子信号或粘附分子,使用TLR激动剂、CD40激动剂或代谢调节将其重新编程为抗肿瘤表型,并选择性地去除促进肿瘤生长的TAM亚群。多药耐药、缺乏准确的生物标志物和药物非特异性也是主要问题。将巨噬细胞靶向治疗与化疗和免疫治疗相结合可能会给治疗带来革命性的变化。巨噬细胞研究将随着新技术和多组学方法的发展,帮助我们了解结直肠癌并建立特异性和有效的治疗方法。
{"title":"From silent partners to potential therapeutic targets: macrophages in colorectal cancer.","authors":"Hayat Khizar, Kamran Ali, Jianwei Wang","doi":"10.1007/s00262-025-03965-w","DOIUrl":"10.1007/s00262-025-03965-w","url":null,"abstract":"<p><p>Cancer cells grow and survive in the tumor microenvironment, which is a complicated process. As a key part of how colorectal cancer (CRC) progresses, tumor-associated macrophages (TAMs) exhibit a double role. Through angiogenesis, this TAM can promote the growth of cancers. Although being able to modify and adjust immune cells is a great advantage, these cells can also exhibit anti-cancer properties including direct killing of cancer cells, presenting antigens, and aiding T cell-mediated responses. The delicate regulatory mechanisms between the immune system and tumors are composed of a complex network of pathways regulated by several factors including hypoxia, metabolic reprogramming, cytokine/chemokine signaling, and cell interactions. Decoding and figuring out these complex systems become significant in building targeted treatment programs. Targeting TAMs in CRC involves disrupting chemokine signaling or adhesion molecules, reprogramming them to an anti-tumor phenotype using TLR agonists, CD40 agonists, or metabolic modulation, and selectively removing TAM subsets that promote tumor growth. Multi-drug resistance, the absence of an accurate biomarker, and drug non-specificity are also major problems. Combining macrophage-targeted therapies with chemotherapy and immunotherapy may revolutionize treatment. Macrophage studies will advance with new technology and multi-omics methodologies to help us understand CRC and build specific and efficient treatments.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":"74 4","pages":"121"},"PeriodicalIF":4.6,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11861851/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490778","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}
引用次数: 0
期刊
Cancer Immunology, Immunotherapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1