Cancer immunology, immunotherapy : CII最新文献

筛选
英文 中文
NLRP3 activation promotes cGAS/STING signaling and antitumor immunity by colorectal cancer cells. NLRP3激活可促进结直肠癌细胞的cGAS/STING信号传导和抗肿瘤免疫。
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-07 DOI: 10.1007/s00262-025-04088-y
Courtney Mowat, Daniel Schiller, Kristi Baker
{"title":"NLRP3 activation promotes cGAS/STING signaling and antitumor immunity by colorectal cancer cells.","authors":"Courtney Mowat, Daniel Schiller, Kristi Baker","doi":"10.1007/s00262-025-04088-y","DOIUrl":"10.1007/s00262-025-04088-y","url":null,"abstract":"<p><strong>Introduction: </strong>Colorectal cancer (CRC) is a highly prevalent and deadly disease that is largely refractory to immunotherapy. The only CRC subset that responds to these therapies is characterized by prevalent microsatellite instability (MSI), extensive  CD8+ T cell infiltration and high expression of innate immune signaling pathways. Endogenous activation of the cGAS/STING pathway is essential for this CD8+ T cell antitumor response in MSI CRCs, suggesting that activating it in other CRCs could boost immunotherapy response rates. In contrast, activation of the NLRP3 inflammasome is typically associated with tumor-promoting inflammation although this has primarily been studied in immune cells.</p><p><strong>Methods: </strong>We used a mixture of flow cytometry, activation assays, in vivo orthotopic models and patient-derived organoids to investigate the effect of NLRP3 activation in CRC cells on cGAS/STING-mediated antitumor immunity.</p><p><strong>Results: </strong>Our results show that activation of the NLRP3 inflammasome specifically in CRC cells boosts cGAS/STING signaling in both MSI and non-MSI CRCs and that dual stimulation increases CD8+ T cell-mediated antitumor immunity. The ability of NLRP3 to enhance cGAS/STING signaling was specific and did not occur with activation of other innate immune pathways such as AIM2 or TLRs. Enhancement of cGAS/STING signaling by NLRP3 proceeded via a positive feedback loop that was inflammasome-independent and required early crosstalk between the signaling mediators and regulation of their gene expression.</p><p><strong>Conclusions: </strong>Activation of NLRP3 specifically in CRC cells could be a promising strategy to boost antitumor immunity in otherwise immunotherapy resistant CRCs.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 8","pages":"238"},"PeriodicalIF":0.0,"publicationDate":"2025-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12145389/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144251872","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Glutamate transporter SLC1A6 promotes resistance to immunotherapy in cancer. 谷氨酸转运体SLC1A6促进癌症免疫治疗的抵抗。
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-07 DOI: 10.1007/s00262-025-04074-4
Chenchen Li, Yi Lin, Haoran Zheng, Hengda Zeng, Longhao Xu, Daqin Wu, Jianwen Lao, Peicong Cai, Shuai Liang, Chunhui Wang, Tianxin Lin, Wenlong Zhong
{"title":"Glutamate transporter SLC1A6 promotes resistance to immunotherapy in cancer.","authors":"Chenchen Li, Yi Lin, Haoran Zheng, Hengda Zeng, Longhao Xu, Daqin Wu, Jianwen Lao, Peicong Cai, Shuai Liang, Chunhui Wang, Tianxin Lin, Wenlong Zhong","doi":"10.1007/s00262-025-04074-4","DOIUrl":"10.1007/s00262-025-04074-4","url":null,"abstract":"<p><strong>Background: </strong>Resistance to immune checkpoint inhibitors remains a significant challenge in the treatment of cancer. Emerging evidence suggests that metabolic reprogramming plays a crucial role in tumor metabolism and progression. Our study strived to investigate the role and underlying mechanisms of the glutamate transporter SLC1A6 in resistance to immunotherapy of cancer.</p><p><strong>Methods: </strong>Single-cell RNA sequencing was performed on bladder cancer patients receiving neoadjuvant immunotherapy to identify the expression of SLC1A6 in treatment-resistant cases. The clinical prognostic value of SLC1A6 in cancer was validated using publicly available lung cancer single-cell datasets, as well as transcriptomic data from both bladder and lung cancer cohorts. Flow cytometry was employed to assess the impact of SLC1A6 knockdown on the effector function of CD8⁺ T cell. In vivo tumor models were used to evaluate the role of SLC1A6 in immunotherapy resistance, with immunofluorescence staining performed to examine GZMB⁺ CD8⁺ T cell infiltration.</p><p><strong>Results: </strong>SLC1A6 was highly expressed in bladder cancer patients resistant to neoadjuvant immunotherapy, and its expression was associated with disease progression, poor prognosis, and low immune infiltration. Knockdown of SLC1A6 in tumor cells enhanced CD8⁺ T cell effector function. SLC1A6 knockdown also improved the efficacy of immunotherapy and increased the infiltration of GZMB⁺ CD8⁺ T cells within the tumor microenvironment.</p><p><strong>Conclusions: </strong>SLC1A6 plays a critical role in resistance to immunotherapy in cancer. Targeting SLC1A6 may provide a promising therapeutic strategy for improving responses to neoadjuvant immunotherapy and advancing combination treatment approaches.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 8","pages":"240"},"PeriodicalIF":0.0,"publicationDate":"2025-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12145344/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144251871","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Assessment of compartment-specific CD103-positive cells for prognosis prediction of colorectal cancer. 评估室特异性cd103阳性细胞对结直肠癌预后的预测作用。
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-07 DOI: 10.1007/s00262-025-04087-z
Anpei Huang, Yuanhui Wu, Ji Cui, Muyan Cai, Yumo Xie, Jialin Zou, Maram Alenzi, Hui Yang, Pinzhu Huang, Meijin Huang
{"title":"Assessment of compartment-specific CD103-positive cells for prognosis prediction of colorectal cancer.","authors":"Anpei Huang, Yuanhui Wu, Ji Cui, Muyan Cai, Yumo Xie, Jialin Zou, Maram Alenzi, Hui Yang, Pinzhu Huang, Meijin Huang","doi":"10.1007/s00262-025-04087-z","DOIUrl":"10.1007/s00262-025-04087-z","url":null,"abstract":"<p><strong>Background: </strong>CD103<sup>+</sup> tissue-resident memory T cells was detected in various solid malignancies, like colorectal cancer (CRC), and associated with improved survival. However, clinical significance of CD103<sup>+</sup> cells in specific intratumor compartment remains unclear.</p><p><strong>Methods: </strong>The abundance and distribution of CD103<sup>+</sup> cells were assessed using immunohistochemistry and quantified separately for 3 compartments, including intraepithelial compartments at center of tumor (CT-IEL), stromal compartments at center of tumor (CT-ST) and invasive margin (IM) in a cohort of 224 CRC patients under radical surgery and correlated with outcome. Findings in each compartment were then validated in an external validation cohort comprising 294 CRC patients.</p><p><strong>Results: </strong>Elevated density of CD103<sup>+</sup> cells infiltration in the CT-IEL, CT-ST or IM compartment was correlated with favorable survival in both the initial discovery cohort and subsequent validation cohort. Notably, abundant CD103<sup>+</sup> cells located in the CT-IEL compartment was remained an independent prognostic indicator for CRC patients by multivariant analysis. Characterization study showed that intraepithelial CD103<sup>+</sup> cells were predominantly single positive CD8 T cells. Conversely, CD103<sup>+</sup> cells exhibited a heterogeneous population comprising CD103<sup>+</sup>CD8<sup>+</sup> cells, CD103<sup>+</sup>CD4<sup>+</sup> cells, and nonconventional CD103<sup>+</sup>CD4<sup>+</sup>CD8<sup>+</sup> cells in the CT-ST and IM compartments. Finally, a CD103 score was generated comprising abundance of CD103<sup>+</sup> cells in the 3 compartments. This score had the highest relative contribution to the risk of all clinical parameters for prognosis in both cohorts.</p><p><strong>Conclusion: </strong>This study supported a phenotypic heterogeneity of CD103<sup>+</sup> cells in CRC, and provided a reliable estimate of the risk of death and recurrence in CRC patients based on combined analysis of CD103<sup>+</sup> cells within 3 intratumor compartments.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 8","pages":"237"},"PeriodicalIF":0.0,"publicationDate":"2025-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12145388/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144251869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Characteristics and outcomes of primary and secondary resistance to immune checkpoint inhibitors in hepatocellular carcinoma. 肝细胞癌免疫检查点抑制剂原发性和继发性耐药的特点和结果
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-07 DOI: 10.1007/s00262-025-04089-x
Xiaowen Cui, Minghao Ruan, Yao Li, Cheng Yang, Jin Zhang, Riming Jin, Dong Wu, Wen Sun, Ruoyu Wang
{"title":"Characteristics and outcomes of primary and secondary resistance to immune checkpoint inhibitors in hepatocellular carcinoma.","authors":"Xiaowen Cui, Minghao Ruan, Yao Li, Cheng Yang, Jin Zhang, Riming Jin, Dong Wu, Wen Sun, Ruoyu Wang","doi":"10.1007/s00262-025-04089-x","DOIUrl":"10.1007/s00262-025-04089-x","url":null,"abstract":"<p><p>Resistance limits the efficacy and durability of immune checkpoint inhibitors (ICIs) in hepatocellular carcinoma (HCC). Therefore, we conducted a retrospective cohort study to investigate the outcomes and characteristics of HCC patients with resistance to immunotherapy. Patients with HCC who have received ICIs at Eastern Hepatobiliary Surgery Hospital between 2016 and 2021 were retrospectively screened and divided into primary resistance, secondary resistance, and durable response group. Time to progression (TTP), overall survival (OS), subsequent management and post-progression survival (PPS) were analyzed. Of 496 patients included, 229 (46.2%) and 141 (28.4%) patients developed primary and secondary resistance, and 126 (25.4%) patients achieved a durable response, the median TTP was 2.83 [2.56-3.09] months, 11.93 [10.45-13.40] months, and not reached, respectively, whereas the median OS was 12.83 [10.36-15.30] months, 31.53 [28.09-34.97] and not reached, respectively. Multivariate logistic regression revealed that Child-Pugh score, BCLC stage, and combined systemic therapies (ICI plus bevacizumab or lenvatinib versus ICI monotherapy) were independently associated with primary resistance, and only combined systemic therapies (ICI plus bevacizumab versus ICI monotherapy) were independently associated with secondary resistance. AFP levels were independently associated with PPS in patients with primary resistance, while post-progression therapies (ICI-based therapies versus others) were independently associated with PPS in patients with resistance. The risk of resistance was notably lower in patients receiving the combination of ICI plus bevacizumab. High AFP levels were associated with the survival of patients with primary resistance. ICI-based maintenance therapy after resistance may provide a significant survival advantage for HCC patients.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 8","pages":"239"},"PeriodicalIF":0.0,"publicationDate":"2025-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12145340/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144251870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Single-cell RNA sequencing analysis reveals a lack of CXCL13+ T cell subsets associated with the recurrence of cervical squamous cell carcinoma following concurrent chemoradiotherapy. 单细胞RNA测序分析显示,缺乏CXCL13+ T细胞亚群与同步放化疗后宫颈鳞状细胞癌复发相关。
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-04 DOI: 10.1007/s00262-025-04083-3
Xia Li, Yanmei Cheng, Mei Ji, Junqi Liu, Zhao Zhao, Qitai Zhao
{"title":"Single-cell RNA sequencing analysis reveals a lack of CXCL13<sup>+</sup> T cell subsets associated with the recurrence of cervical squamous cell carcinoma following concurrent chemoradiotherapy.","authors":"Xia Li, Yanmei Cheng, Mei Ji, Junqi Liu, Zhao Zhao, Qitai Zhao","doi":"10.1007/s00262-025-04083-3","DOIUrl":"10.1007/s00262-025-04083-3","url":null,"abstract":"<p><p>Concurrent chemoradiotherapy (CCRT) is the standard treatment for advanced cervical cancer, but tumor recurrence within 3-5 years remains a significant challenge. In this study, using 10 × single-cell sequencing, we constructed a cellular atlas of the tumor microenvironment from six CSCC patients, including three with recurrence and three without, prior to CCRT. We analyzed cellular subsets, focusing on T cells, myeloid cells and cancer associated fibroblasts (CAFs), and their interactions within the tumor. Key findings revealed that CXCL13<sup>+</sup> T cell subsets were significantly increased in non-recurrent tumors and acted as major signal senders. In recurrent tumors, FOXP3<sup>+</sup> and IL2RA<sup>+</sup> Tregs were the primary mediators of cell communications. CXCL13<sup>+</sup> CD8<sup>+</sup> T cells interacted with SPP1<sup>+</sup> tumor-associated macrophages (TAMs) in non-recurrent tumors, while in recurrent tumors, they interacted with CD163<sup>+</sup> TAMs. Moreover, in recurrent tumor tissues, this subset demonstrates a preferential interaction with MMP3<sup>+</sup> CAFs. The study also identified five genes (PDCD1, CXCL13, TOX, RGS1, and ALOX5AP) based on CXCL13<sup>+</sup> T cell signature to construct predictive models for recurrence, with the random forest model showing the best performance. This study provides new insights into recurrence mechanisms in CSCC and suggests that increasing CXCL13<sup>+</sup> T cells could be a potential therapeutic strategy.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 7","pages":"235"},"PeriodicalIF":0.0,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12137842/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144218532","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Infectious complications distribution following CLL1 CAR-T cell therapy for acute myeloid leukemia. 更正:CLL1 CAR-T细胞治疗急性髓性白血病后感染性并发症分布。
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-04 DOI: 10.1007/s00262-025-04066-4
Jianmei Xu, Huan Zhang, Yifan Zhao, Xiaomei Zhang, Shujing Guo, Xiaoxue Shi, Xia Xiao, Hairong Lyu, Yu Zhang, Xiaoyuan He, Mingfeng Zhao
{"title":"Correction: Infectious complications distribution following CLL1 CAR-T cell therapy for acute myeloid leukemia.","authors":"Jianmei Xu, Huan Zhang, Yifan Zhao, Xiaomei Zhang, Shujing Guo, Xiaoxue Shi, Xia Xiao, Hairong Lyu, Yu Zhang, Xiaoyuan He, Mingfeng Zhao","doi":"10.1007/s00262-025-04066-4","DOIUrl":"10.1007/s00262-025-04066-4","url":null,"abstract":"","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 7","pages":"232"},"PeriodicalIF":0.0,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12137827/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144218529","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oncolytic adenovirus encoding variant interleukin-2 combined with chemotherapy enables PD-L1 inhibition in pancreatic cancer models. 溶瘤腺病毒编码变异型白细胞介素-2联合化疗可抑制胰腺癌模型中的PD-L1。
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-04 DOI: 10.1007/s00262-025-04072-6
Santeri A Pakola, Nea Ojala, Tatiana V Kudling, James H A Clubb, Elise Jirovec, Mirte van der Heijden, Victor Arias, Lyna Haybout, Saru Basnet, Susanna Grönberg-Vähä-Koskela, Dafne C A Quixabeira, Joao M Santos, Victor Cervera-Carrascon, Otto Hemminki, Anna Kanerva, Harri Mustonen, Pauli Puolakkainen, Hanna Seppänen, Akseli Hemminki
{"title":"Oncolytic adenovirus encoding variant interleukin-2 combined with chemotherapy enables PD-L1 inhibition in pancreatic cancer models.","authors":"Santeri A Pakola, Nea Ojala, Tatiana V Kudling, James H A Clubb, Elise Jirovec, Mirte van der Heijden, Victor Arias, Lyna Haybout, Saru Basnet, Susanna Grönberg-Vähä-Koskela, Dafne C A Quixabeira, Joao M Santos, Victor Cervera-Carrascon, Otto Hemminki, Anna Kanerva, Harri Mustonen, Pauli Puolakkainen, Hanna Seppänen, Akseli Hemminki","doi":"10.1007/s00262-025-04072-6","DOIUrl":"10.1007/s00262-025-04072-6","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is a cancer with dismal prognosis due to resistance to most current therapies. Although immunotherapy has improved the treatment of many solid cancers, pancreatic cancer remains resistant to immunotherapy due to immunosuppressive tumor microenvironment, limited lymphocyte infiltration and lack of neoantigens. Oncolytic adenoviruses are a possible solution to treatment resistance in PDAC due to their ability to elicit lymphocyte trafficking and epitope spreading. Herein, we tested if an oncolytic adenovirus encoding a variant interleukin-2 molecule (Ad5/3-E2F-d24-vIL2), could enable immune checkpoint inhibitor (ICI) therapy in PDAC when combined with chemotherapy. Rationale for Ad5/3-E2F-d24-vIL2 was tested in vitro, where increase in programmed death ligand 1 (PD-L1) expression was seen after virotherapy and chemotherapy. Expression of other B7 family proteins was characterized in mono- and co-culture settings of cancer cells, fibroblasts, and macrophages. The combination therapy of virotherapy, chemotherapy and ICI was characterized in freshly resected ex vivo pancreatic tumor samples. Combination of ICI with virotherapy showed increased interferon and chemokine production in samples, with expansion of cytotoxic CD8 + T cells seen by flow cytometry. In vivo evaluation of the triple combination therapy in a Syrian hamster model showed improved tumor growth control and overall survival, with concurrent increase in intratumoral lymphocytes during therapy. Animals cured with the therapy showed resistance to re-challenge with the same cell line, supportive of successful generation of anti-tumor immunity in the animals. The combination treatment of Ad5/3-E2F-d24-vIL2, chemotherapy, and checkpoint inhibition is a promising treatment modality to tackle treatment resistance in PDAC.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 7","pages":"234"},"PeriodicalIF":0.0,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12137825/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144218530","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Predictive factors and nomogram for the risk of cytokine release syndrome with anti-CD3 × CD20 bispecific antibodies for Chinese patients. 中国患者抗cd3 × CD20双特异性抗体细胞因子释放综合征风险的预测因素和nomogram
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-04 DOI: 10.1007/s00262-025-04080-6
Peng Sun, Cui Chen, Bai-Tian Zhao, Bo-Yan Zhang, Hang Yang, Yu Wang, Fei Xu, Kang-Ming Huang, Yin-Ting Liu, Mei-Shuo Ouyang, Jia-Jia Huang, Pan-Pan Liu, Ying Guo, Zhi-Ming Li
{"title":"Predictive factors and nomogram for the risk of cytokine release syndrome with anti-CD3 × CD20 bispecific antibodies for Chinese patients.","authors":"Peng Sun, Cui Chen, Bai-Tian Zhao, Bo-Yan Zhang, Hang Yang, Yu Wang, Fei Xu, Kang-Ming Huang, Yin-Ting Liu, Mei-Shuo Ouyang, Jia-Jia Huang, Pan-Pan Liu, Ying Guo, Zhi-Ming Li","doi":"10.1007/s00262-025-04080-6","DOIUrl":"10.1007/s00262-025-04080-6","url":null,"abstract":"<p><strong>Background: </strong>Targeting CD3 × CD20 bispecific antibodies (BsAbs) represents a new milestone in the salvage therapy of relapsed/relapsed large B cell lymphoma and follicular lymphoma. However, cytokine release syndrome (CRS) remains one of the major concerns in clinical practice of CD3 × CD20 BsAbs. This study aimed to identify the potential predictive factors and to construct a nomogram of grade ≥ 2 CRS in CD3 × CD20 BsAbs in Chinese patients.</p><p><strong>Methods: </strong>A total of 87 consecutive patients with B-NHL who received CD3 × CD20 BsAbs at Sun Yat-sen University Cancer Center from January 2021 to December 2023 were included and analyzed. Clinical data were collected, and various methods including machine learning algorithms were introduced.</p><p><strong>Results: </strong>The median age of the patients was 55 years, and the median number of previous treatment lines was 2. CRS occurred in 42 patients (48.3%), including 27 cases of grade 1, 9 cases of grade 2, 5 cases of grade 3, and 1 case of grade 5. Four variables were revealed and selected: bulky disease (> 5 cm), number of previous treatment lines (≥ 3), monocyte-to-lymphocyte ratio (MLR), and platelet count (PLT). Bootstrap resampling was introduced for both internal validation and model building. A predictive nomogram was ultimately established, with the ROC-AUC of 0.867 (95% CI 0.719-0.954).</p><p><strong>Conclusion: </strong>This study suggests four potential predictive factors and provides a feasible and easy-to-use nomogram for grade ≥ 2 CRS in Chinese patients for the first time, which is important for guiding personalized management and early intervention of CRS.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 7","pages":"236"},"PeriodicalIF":0.0,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12137836/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144218531","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Gut microbiota affects PD-L1 therapy and its mechanism in melanoma. 纠正:肠道微生物群影响PD-L1治疗及其在黑色素瘤中的机制。
Cancer immunology, immunotherapy : CII Pub Date : 2025-06-04 DOI: 10.1007/s00262-025-04092-2
Shiqi Liu, Jiahui Liu, Yingwu Mei, Wenjuan Zhang
{"title":"Correction: Gut microbiota affects PD-L1 therapy and its mechanism in melanoma.","authors":"Shiqi Liu, Jiahui Liu, Yingwu Mei, Wenjuan Zhang","doi":"10.1007/s00262-025-04092-2","DOIUrl":"10.1007/s00262-025-04092-2","url":null,"abstract":"","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 7","pages":"233"},"PeriodicalIF":0.0,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12137826/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144218528","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cancer therapy via neoepitope-specific monoclonal antibody cocktails. 新表位特异性单克隆抗体鸡尾酒疗法治疗癌症。
Cancer immunology, immunotherapy : CII Pub Date : 2025-05-31 DOI: 10.1007/s00262-025-04075-3
Colin J Hartman, Asmaa O Mohamed, Girja S Shukla, Stephanie C Pero, Yu-Jing Sun, Roberto S Rodríguez-Jimenez, Nicholas F Genovese, Nico M Kohler, Thomas R Hemphill, Yina H Huang, David N Krag, Margaret E Ackerman
{"title":"Cancer therapy via neoepitope-specific monoclonal antibody cocktails.","authors":"Colin J Hartman, Asmaa O Mohamed, Girja S Shukla, Stephanie C Pero, Yu-Jing Sun, Roberto S Rodríguez-Jimenez, Nicholas F Genovese, Nico M Kohler, Thomas R Hemphill, Yina H Huang, David N Krag, Margaret E Ackerman","doi":"10.1007/s00262-025-04075-3","DOIUrl":"10.1007/s00262-025-04075-3","url":null,"abstract":"<p><p>Cellular heterogeneity presents a significant challenge to cancer treatment. Antibody therapies targeting individual tumor-associated antigens can be extremely effective but are not suited for all patients and often fail against tumors with heterogeneous expression as tumor cells with low or no antigen expression escape targeting and develop resistance. Simultaneously targeting multiple tumor-specific proteins with multiple antibodies has the potential to overcome this barrier and improve efficacy, but relatively few widely expressed cancer-specific antigens are known. In contrast, neoepitopes, which arise from mutations unique to tumor cells, are considerably more abundant. However, since neoepitopes are not commonly shared between individuals, a patient-customized approach is necessary and motivates efforts to develop an efficient means to identify suitable target mutations and isolate neoepitope-specific monoclonal antibodies. Here, focusing on the latter goal, we use directed evolution in yeast and phage display systems to engineer antibodies from nonimmune, human antibody fragment libraries that are specific for neoepitopes previously reported in the B16F10 melanoma model. We demonstrate proof-of-concept for a pipeline that supports rapid isolation and functional enhancement of multiple neoepitope peptide-targeted monoclonal antibodies and demonstrate their robust binding to B16F10 cells and potent effector functions in vitro. These antibodies were combined and evaluated in vivo for anticancer activity in tumor-bearing mice, where they suppressed B16F10 tumor growth and prolonged survival. These findings emphasize the potential for clinical application of patient-customized antibody cocktails in the treatment of the many cancers poorly addressed by current therapies.</p>","PeriodicalId":520581,"journal":{"name":"Cancer immunology, immunotherapy : CII","volume":"74 7","pages":"231"},"PeriodicalIF":0.0,"publicationDate":"2025-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12126454/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144192656","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信