首页 > 最新文献

Journal for Immunotherapy of Cancer最新文献

英文 中文
Anti-HER2×CCR4 bispecific antibody enhances antitumor immunity in advanced HER2-positive tumors by chemotaxis blockade and depletion of tumor-associated Tregs, without inducing systemic toxicity. Anti-HER2×CCR4双特异性抗体通过趋化性阻断和肿瘤相关Tregs的消耗,增强晚期her2阳性肿瘤的抗肿瘤免疫,而不诱导全身毒性。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-012829
Chaokun Wang, Jiachang Li, Yue Tong, Haotian Chang, Jingyi Xu, Hanchen Zhu, Yongmei Yin, Meng Meng, Rimo Xi

Background: CCR4+ regulatory T cells (Tregs), which are widely present in peripheral circulation and tumor microenvironment (TME), promote tumor immune evasion by infiltrating human epidermal growth factor receptor 2-positive (HER2+) tumors in a chemokine-driven manner. However, therapies targeting CCR4 (eg, mogamulizumab) for systemic Treg depletion risk significant toxicity and have far been confined to hematological malignancies. Notably, tumor-chemotactic peripheral Tregs-key precursors to tumor-infiltrating Tregs (tumor-infiltrating lymphocyte (TIL)-Tregs)-remain overlooked.

Methods: We designed and expressed four candidate anti-HER2×CCR4 DVD-Ig bispecific antibodies with varying degrees of anti-CCR4 domain masking, while leaving anti-HER2 domains fully exposed, to preferentially deplete tumor-associated Tregs, including TIL-Tregs and tumor-chemotactic peripheral Tregs. Stability and antitumor activity were assessed in vitro. In human immune system-reconstituted NOG mice, we systematically: (1) conducted a comprehensive dose-response evaluation of the XL-11 to characterize pharmacological efficacy and potential systemic toxicity, (2) assessed the immune memory, (3) studied the synergy of XL-11 with programmed cell death protein-1/programmed death-ligand 1 (PD-1/L1) blocker, and (4) analyzed pharmacokinetic profile (PK).

Results: The lower affinity for CCR4 compared with HER2 enables anti-HER2×CCR4 DVD-Igs to priority target TIL-Tregs, and reduce binding to peripheral Tregs compared with mogamulizumab. Anti-HER2×CCR4 DVD-Igs inhibit Treg chemotaxis to TME, and killing Tregs and HER2+ tumor cells through antibody-dependent cellular cytotoxicity. Among the candidates, XL-11, which had the greatest exposure of the anti-CCR4 domain, was selected for in vivo evaluation due to its superior stability and potent antitumor activity. In vivo models of gastric cancer and breast cancer, XL-11, by reducing TIL-Tregs and increasing CD8+/Tregs ratios, induces potent antitumor activity even in advanced stages, with no evidence of metastasis. Concurrently, XL-11 specifically depletes tumor-chemotactic peripheral Tregs, further enhancing antitumor immunity while avoiding reducing Tregs throughout the body even at a high dose (10 mg/kg). There was no increase in memory T cells. In addition, XL-11 enhances the antitumor activity of anti-PD-1 antibodies and shows superior PK properties.

Conclusions: XL-11 mediates potent antitumor immunity in advanced HER2+ tumors while avoiding reducing Tregs throughout the body. XL-11 also acts synergistically with anti-PD-1 therapy, and exhibits favorable stability and PK supporting clinical translation. This work advances Treg-targeted therapies in HER2+ tumors and overcomes the therapeutic limitations of mogamulizumab.

背景:CCR4+调节性T细胞(Tregs)广泛存在于外周循环和肿瘤微环境(TME)中,通过趋化因子驱动的方式浸润人表皮生长因子受体2阳性(HER2+)肿瘤,促进肿瘤免疫逃避。然而,针对CCR4(例如mogamulizumab)的系统性Treg耗竭疗法存在显著毒性,并且迄今为止仅限于血液系统恶性肿瘤。值得注意的是,肿瘤趋化外周Tregs——肿瘤浸润性Tregs的关键前体(肿瘤浸润性淋巴细胞(TIL)-Tregs)——仍然被忽视。方法:我们设计并表达了四种候选的anti-HER2×CCR4 DVD-Ig双特异性抗体,它们具有不同程度的抗ccr4结构域屏蔽,同时使抗her2结构域完全暴露,以优先消耗肿瘤相关的treg,包括til - treg和肿瘤趋化外周treg。体外稳定性和抗肿瘤活性评估。在人类免疫系统重构的NOG小鼠中,我们系统地:(1)对XL-11进行了全面的剂量反应评估,以表征药理功效和潜在的全身毒性;(2)评估了免疫记忆;(3)研究了XL-11与程序性细胞死亡蛋白-1/程序性死亡配体1 (PD-1/L1)阻滞剂的协同作用;(4)分析了药代动力学谱(PK)。结果:与HER2相比,对CCR4的亲和力较低,使得anti-HER2×CCR4 DVD-Igs优先靶向til - treg,与mogamulizumab相比,与外周treg的结合减少。Anti-HER2×CCR4 DVD-Igs抑制Treg对TME的趋化性,并通过抗体依赖性细胞毒性杀死Treg和HER2+肿瘤细胞。在候选药物中,抗ccr4结构域暴露最多的XL-11因其优越的稳定性和强大的抗肿瘤活性而被选中进行体内评价。在胃癌和乳腺癌的体内模型中,XL-11通过降低TIL-Tregs和增加CD8+/Tregs的比例,即使在晚期也能诱导出强大的抗肿瘤活性,没有转移的证据。同时,XL-11特异性地消耗肿瘤趋化外周treg,进一步增强抗肿瘤免疫,同时即使在高剂量(10 mg/kg)下也避免降低全身treg。记忆T细胞没有增加。此外,XL-11还能增强抗pd -1抗体的抗肿瘤活性,并表现出较好的PK特性。结论:XL-11在晚期HER2+肿瘤中介导了有效的抗肿瘤免疫,同时避免了全身Tregs的降低。XL-11还与抗pd -1治疗协同作用,并表现出良好的稳定性和支持临床翻译的PK。这项工作推进了HER2+肿瘤的treg靶向治疗,克服了mogamulizumab的治疗局限性。
{"title":"Anti-HER2×CCR4 bispecific antibody enhances antitumor immunity in advanced HER2-positive tumors by chemotaxis blockade and depletion of tumor-associated Tregs, without inducing systemic toxicity.","authors":"Chaokun Wang, Jiachang Li, Yue Tong, Haotian Chang, Jingyi Xu, Hanchen Zhu, Yongmei Yin, Meng Meng, Rimo Xi","doi":"10.1136/jitc-2025-012829","DOIUrl":"10.1136/jitc-2025-012829","url":null,"abstract":"<p><strong>Background: </strong>CCR4<sup>+</sup> regulatory T cells (Tregs), which are widely present in peripheral circulation and tumor microenvironment (TME), promote tumor immune evasion by infiltrating human epidermal growth factor receptor 2-positive (HER2<sup>+</sup>) tumors in a chemokine-driven manner. However, therapies targeting CCR4 (eg, mogamulizumab) for systemic Treg depletion risk significant toxicity and have far been confined to hematological malignancies. Notably, tumor-chemotactic peripheral Tregs-key precursors to tumor-infiltrating Tregs (tumor-infiltrating lymphocyte (TIL)-Tregs)-remain overlooked.</p><p><strong>Methods: </strong>We designed and expressed four candidate anti-HER2×CCR4 DVD-Ig bispecific antibodies with varying degrees of anti-CCR4 domain masking, while leaving anti-HER2 domains fully exposed, to preferentially deplete tumor-associated Tregs, including TIL-Tregs and tumor-chemotactic peripheral Tregs. Stability and antitumor activity were assessed in vitro. In human immune system-reconstituted NOG mice, we systematically: (1) conducted a comprehensive dose-response evaluation of the XL-11 to characterize pharmacological efficacy and potential systemic toxicity, (2) assessed the immune memory, (3) studied the synergy of XL-11 with programmed cell death protein-1/programmed death-ligand 1 (PD-1/L1) blocker, and (4) analyzed pharmacokinetic profile (PK).</p><p><strong>Results: </strong>The lower affinity for CCR4 compared with HER2 enables anti-HER2×CCR4 DVD-Igs to priority target TIL-Tregs, and reduce binding to peripheral Tregs compared with mogamulizumab. Anti-HER2×CCR4 DVD-Igs inhibit Treg chemotaxis to TME, and killing Tregs and HER2<sup>+</sup> tumor cells through antibody-dependent cellular cytotoxicity. Among the candidates, XL-11, which had the greatest exposure of the anti-CCR4 domain, was selected for in vivo evaluation due to its superior stability and potent antitumor activity. In vivo models of gastric cancer and breast cancer, XL-11, by reducing TIL-Tregs and increasing CD8<sup>+</sup>/Tregs ratios, induces potent antitumor activity even in advanced stages, with no evidence of metastasis. Concurrently, XL-11 specifically depletes tumor-chemotactic peripheral Tregs, further enhancing antitumor immunity while avoiding reducing Tregs throughout the body even at a high dose (10 mg/kg). There was no increase in memory T cells. In addition, XL-11 enhances the antitumor activity of anti-PD-1 antibodies and shows superior PK properties.</p><p><strong>Conclusions: </strong>XL-11 mediates potent antitumor immunity in advanced HER2<sup>+</sup> tumors while avoiding reducing Tregs throughout the body. XL-11 also acts synergistically with anti-PD-1 therapy, and exhibits favorable stability and PK supporting clinical translation. This work advances Treg-targeted therapies in HER2<sup>+</sup> tumors and overcomes the therapeutic limitations of mogamulizumab.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719890/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804281","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Interpretable multimodal radiopathomics model predicting pathological complete response to neoadjuvant chemoimmunotherapy in esophageal squamous cell carcinoma. 可解释的多模态放射病理学模型预测食管鳞状细胞癌对新辅助化疗免疫治疗的病理完全反应。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-013840
Baojia Qi, Zhaoyu Jiang, Haixia Shen, Jiacheng Li, Zhixiang Wang, Min Fang, Changchun Wang, Youhua Jiang, Jingping Yuan, Inigo Bermejo, Andre Dekker, Dirk De Ruysscher, Leonard Wee, Wencheng Zhang, Yongling Ji, Zhen Zhang

Background: Accurate preoperative prediction of pathological complete response (pCR) following neoadjuvant chemoimmunotherapy (nCIT) could help individualize treatment for patients with esophageal squamous cell carcinoma (ESCC). This study aimed to develop and externally validate an interpretable multimodal machine learning framework that integrates CT radiomics and H&E-stained whole-slide images pathomics to predict pCR.

Methods: In this multicenter, retrospective study, 335 patients with ESCC who received nCIT followed by esophagectomy were enrolled from three institutions. Patients from one center were divided into a training set (181 patients) and an internal test set (115 patients), while data from the other two centers comprised an external test set (39 patients). We developed unimodal radiomics and pathomics models, and two multimodal fusion models-an intermediate fusion model (MIFM) and a late fusion model (MLFM). Model performance was evaluated using the area under the curve (AUC), accuracy, sensitivity, specificity, and F1 score, with exploratory survival stratification by observed and model-predicted pCR status. Interpretability was treated as a design constraint and operationalized at both the feature and model levels.

Results: The MIFM outperformed unimodal models and the MLFM across all cohorts, achieving AUC/accuracy/sensitivity/specificity/F1 score of 0.97/0.93/0.84/0.96/0.86 (training set), 0.78/0.87/0.62/0.93/0.63 (internal test set), and 0.76/0.77/0.54/0.88/0.61 (external test set). Both observed and predicted pCR status showed exploratory prognostic stratification for overall survival. Feature definitions were mathematically or morphologically explicit, and case-level/cohort-level explanations together with decision-pathway views provided insights into model reasoning. We additionally provide a user-friendly Graphical User Interface to facilitate clinical practice.

Conclusions: We developed and externally validated an interpretable radiopathomics fusion framework that predicts pCR after nCIT in ESCC using standard-of-care data. This model holds promise as an effective tool for guiding individualized decisions between surveillance and timely surgery.

背景:准确预测新辅助化疗免疫治疗(nCIT)后的术前病理完全缓解(pCR),有助于食管鳞状细胞癌(ESCC)患者的个体化治疗。本研究旨在开发和外部验证一个可解释的多模式机器学习框架,该框架集成了CT放射组学和h&e染色的全切片图像病理,以预测pCR。方法:在这项多中心回顾性研究中,335名ESCC患者接受nCIT后食管切除术,来自三个机构。来自一个中心的患者被分为一个训练集(181例患者)和一个内部测试集(115例患者),而来自另外两个中心的数据包括一个外部测试集(39例患者)。我们开发了单峰放射组学和病理模型,以及两个多峰融合模型——中间融合模型(MIFM)和晚期融合模型(MLFM)。使用曲线下面积(AUC)、准确性、敏感性、特异性和F1评分来评估模型的性能,并通过观察到的和模型预测的pCR状态进行探索性生存分层。可解释性被视为一种设计约束,并在特征和模型级别上进行操作。结果:MIFM在所有队列中均优于单峰模型和MLFM, AUC/准确度/灵敏度/特异性/F1评分分别为0.97/0.93/0.84/0.96/0.86(训练集)、0.78/0.87/0.62/0.93/0.63(内部测试集)和0.76/0.77/0.54/0.88/0.61(外部测试集)。观察到的和预测的pCR状态都显示了总体生存的探索性预后分层。特征定义在数学上或形态学上是明确的,案例级/队列级解释与决策路径视图一起提供了对模型推理的见解。我们还提供了一个用户友好的图形用户界面,以方便临床实践。结论:我们开发并外部验证了一个可解释的放射病理学融合框架,该框架使用标准护理数据预测ESCC中nCIT后的pCR。该模型有望成为指导在监测和及时手术之间做出个性化决策的有效工具。
{"title":"Interpretable multimodal radiopathomics model predicting pathological complete response to neoadjuvant chemoimmunotherapy in esophageal squamous cell carcinoma.","authors":"Baojia Qi, Zhaoyu Jiang, Haixia Shen, Jiacheng Li, Zhixiang Wang, Min Fang, Changchun Wang, Youhua Jiang, Jingping Yuan, Inigo Bermejo, Andre Dekker, Dirk De Ruysscher, Leonard Wee, Wencheng Zhang, Yongling Ji, Zhen Zhang","doi":"10.1136/jitc-2025-013840","DOIUrl":"10.1136/jitc-2025-013840","url":null,"abstract":"<p><strong>Background: </strong>Accurate preoperative prediction of pathological complete response (pCR) following neoadjuvant chemoimmunotherapy (nCIT) could help individualize treatment for patients with esophageal squamous cell carcinoma (ESCC). This study aimed to develop and externally validate an interpretable multimodal machine learning framework that integrates CT radiomics and H&E-stained whole-slide images pathomics to predict pCR.</p><p><strong>Methods: </strong>In this multicenter, retrospective study, 335 patients with ESCC who received nCIT followed by esophagectomy were enrolled from three institutions. Patients from one center were divided into a training set (181 patients) and an internal test set (115 patients), while data from the other two centers comprised an external test set (39 patients). We developed unimodal radiomics and pathomics models, and two multimodal fusion models-an intermediate fusion model (MIFM) and a late fusion model (MLFM). Model performance was evaluated using the area under the curve (AUC), accuracy, sensitivity, specificity, and F1 score, with exploratory survival stratification by observed and model-predicted pCR status. Interpretability was treated as a design constraint and operationalized at both the feature and model levels.</p><p><strong>Results: </strong>The MIFM outperformed unimodal models and the MLFM across all cohorts, achieving AUC/accuracy/sensitivity/specificity/F1 score of 0.97/0.93/0.84/0.96/0.86 (training set), 0.78/0.87/0.62/0.93/0.63 (internal test set), and 0.76/0.77/0.54/0.88/0.61 (external test set). Both observed and predicted pCR status showed exploratory prognostic stratification for overall survival. Feature definitions were mathematically or morphologically explicit, and case-level/cohort-level explanations together with decision-pathway views provided insights into model reasoning. We additionally provide a user-friendly Graphical User Interface to facilitate clinical practice.</p><p><strong>Conclusions: </strong>We developed and externally validated an interpretable radiopathomics fusion framework that predicts pCR after nCIT in ESCC using standard-of-care data. This model holds promise as an effective tool for guiding individualized decisions between surveillance and timely surgery.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719895/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804286","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
AMPK agonism optimizes the in vivo activation and antileukemic efficacy of chimeric antigen receptor T cells. AMPK激动作用优化了嵌合抗原受体T细胞的体内活化和抗白血病功效。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2024-010312
Erica Braverman, Mengtao Qin, Elisabet Ampudia-Mesias, Herbert Tres Schuler, Harrison Brown, Lukas Murdych, Ann Titus, Allison MacLean, Bhavya Kanagala, Christopher Wittmann, Archana Ramgopal, Felicia Kemp, Steven J Mullet, Aaron Yang, Amanda C Poholek, Stacy Gelhaus, Craig Byersdorfer

Background: Chimeric antigen receptor (CAR) T cells have achieved remarkable clinical success. However, up to 50% of patients with CAR T-cell treated leukemia relapse and long-term survivor data indicate that CAR T cell persistence is key to enforcing extended, relapse-free survival. Unfortunately, ex vivo expansion protocols often drive metabolic and functional exhaustion, reducing in vivo efficacy. Preclinical models have demonstrated that redirecting metabolism can improve in vivo T-cell function. Here, we hypothesized that exposure to an agonist targeting AMP-activated protein kinase (AMPK) would create CAR T cells capable of increased in vivo function and enhanced leukemia clearance.

Methods: CAR T cells were generated from healthy human donor T cells via lentiviral transduction, followed by exposure to either Compound 991 or dimethyl sulfoxide (DMSO) for 96 hours. During and after agonist treatment, T cells were harvested for metabolic and functional assessments. To test in vivo efficacy, immunodeficient mice were injected with luciferase+NALM6 leukemia cells, and 1 week later with 991- versus DMSO-expanded CAR T cells. Leukemia burden and antileukemia efficacy were assessed via radiance imaging and overall survival.

Results: Compound 991 treatment activated AMPK without limiting cellular expansion, and increased both mitochondrial density and handling of reactive oxygen species. Mechanistically, 991 treatment mimicked nutrient starvation, with increased autophagy and generation of mitochondrially protective metabolites. Importantly, receipt of 991-exposed CAR Ts significantly improved in vivo leukemia clearance and prolonged recipient survival, likely as a result of elevated activation and increased CD4+T cell yields at early times post-injection.

Discussion: Ex vivo expansion is necessary to generate sufficient cell numbers for in vivo administration, but sustained activation and differentiation often negatively impact in vivo persistence and function. Here, we demonstrate that promoting AMPK activity during in vitro CAR T expansion metabolically reprograms cells without limiting T cell yield, increases early activation following in vivo transfer, and ultimately enhances anti-leukemia efficacy. Importantly, Compound 991 treatment achieves these results without further modifying the expansion media, changing the CAR T construct, or genetically altering the cells. Together, these data highlight AMPK agonism as a potent and readily translatable approach to improve the metabolic profile and in vivo efficacy of adoptively transferred T cells.

背景:嵌合抗原受体(CAR) T细胞已经取得了显著的临床成功。然而,高达50%的CAR - T细胞治疗的白血病复发患者和长期幸存者数据表明,CAR - T细胞的持久性是延长无复发生存期的关键。不幸的是,体外扩张方案往往会导致代谢和功能衰竭,降低体内疗效。临床前模型表明,重定向代谢可以改善体内t细胞的功能。在这里,我们假设暴露于靶向amp活化蛋白激酶(AMPK)的激动剂会产生能够增加体内功能和增强白血病清除能力的CAR - T细胞。方法:将健康的人供体T细胞通过慢病毒转导生成CAR - T细胞,然后将其暴露于化合物991或二甲基亚砜(DMSO)中96小时。在激动剂治疗期间和之后,收集T细胞用于代谢和功能评估。为了测试体内的有效性,免疫缺陷小鼠注射荧光素酶+NALM6白血病细胞,1周后注射991-与dmso扩增的CAR - T细胞。通过放射成像和总生存期评估白血病负担和抗白血病疗效。结果:复方991在不限制细胞扩张的情况下激活AMPK,增加线粒体密度和活性氧处理。在机制上,991处理模拟了营养饥饿,增加了自噬和线粒体保护性代谢物的产生。重要的是,接受991暴露的CAR - T显著改善了体内白血病清除率,延长了受体生存期,这可能是由于注射后早期激活水平升高和CD4+T细胞产量增加。讨论:体外扩增对于体内给药产生足够的细胞数量是必要的,但持续的激活和分化通常会对体内的持久性和功能产生负面影响。在这里,我们证明在体外CAR - T扩增过程中促进AMPK活性代谢重编程细胞而不限制T细胞产量,增加体内转移后的早期激活,并最终提高抗白血病疗效。重要的是,化合物991治疗无需进一步修改扩增介质,改变CAR - T结构或基因改变细胞即可达到这些结果。总之,这些数据强调AMPK激动作用是一种有效且易于翻译的方法,可以改善过继转移T细胞的代谢谱和体内功效。
{"title":"AMPK agonism optimizes the in vivo activation and antileukemic efficacy of chimeric antigen receptor T cells.","authors":"Erica Braverman, Mengtao Qin, Elisabet Ampudia-Mesias, Herbert Tres Schuler, Harrison Brown, Lukas Murdych, Ann Titus, Allison MacLean, Bhavya Kanagala, Christopher Wittmann, Archana Ramgopal, Felicia Kemp, Steven J Mullet, Aaron Yang, Amanda C Poholek, Stacy Gelhaus, Craig Byersdorfer","doi":"10.1136/jitc-2024-010312","DOIUrl":"10.1136/jitc-2024-010312","url":null,"abstract":"<p><strong>Background: </strong>Chimeric antigen receptor (CAR) T cells have achieved remarkable clinical success. However, up to 50% of patients with CAR T-cell treated leukemia relapse and long-term survivor data indicate that CAR T cell persistence is key to enforcing extended, relapse-free survival. Unfortunately, ex vivo expansion protocols often drive metabolic and functional exhaustion, reducing in vivo efficacy. Preclinical models have demonstrated that redirecting metabolism can improve in vivo T-cell function. Here, we hypothesized that exposure to an agonist targeting AMP-activated protein kinase (AMPK) would create CAR T cells capable of increased in vivo function and enhanced leukemia clearance.</p><p><strong>Methods: </strong>CAR T cells were generated from healthy human donor T cells via lentiviral transduction, followed by exposure to either Compound 991 or dimethyl sulfoxide (DMSO) for 96 hours. During and after agonist treatment, T cells were harvested for metabolic and functional assessments. To test in vivo efficacy, immunodeficient mice were injected with luciferase+NALM6 leukemia cells, and 1 week later with 991- versus DMSO-expanded CAR T cells. Leukemia burden and antileukemia efficacy were assessed via radiance imaging and overall survival.</p><p><strong>Results: </strong>Compound 991 treatment activated AMPK without limiting cellular expansion, and increased both mitochondrial density and handling of reactive oxygen species. Mechanistically, 991 treatment mimicked nutrient starvation, with increased autophagy and generation of mitochondrially protective metabolites. Importantly, receipt of 991-exposed CAR Ts significantly improved in vivo leukemia clearance and prolonged recipient survival, likely as a result of elevated activation and increased CD4+T cell yields at early times post-injection.</p><p><strong>Discussion: </strong>Ex vivo expansion is necessary to generate sufficient cell numbers for in vivo administration, but sustained activation and differentiation often negatively impact in vivo persistence and function. Here, we demonstrate that promoting AMPK activity during in vitro CAR T expansion metabolically reprograms cells without limiting T cell yield, increases early activation following in vivo transfer, and ultimately enhances anti-leukemia efficacy. Importantly, Compound 991 treatment achieves these results without further modifying the expansion media, changing the CAR T construct, or genetically altering the cells. Together, these data highlight AMPK agonism as a potent and readily translatable approach to improve the metabolic profile and in vivo efficacy of adoptively transferred T cells.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719903/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803541","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Society for Immunotherapy of Cancer: Standards for Reporting of Multiplex Immunohistochemistry/Immunofluorescence Assays (STORMI). 癌症免疫治疗学会:多重免疫组织化学/免疫荧光测定报告标准(STORMI)。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-012280
Sam Sater, Carlo B Bifulco, Jaime Rodriguez-Canales, Joe Yeong, Guray Akturk, Michael Angelo, Carmen Ballesteros-Merino, Peter Bankhead, Subham Basu, Jorge M Blando, Saska Brajkovic, Marco Cassano, Benjamin J Chen, Ahmet F Coskun, Tricia R Cottrell, Carlos E De Andrea, Robin H Edwards, Colt Egelston, Logan L Engle, Marc S Ernstoff, Rong Fan, Michael Feldman, Bernard A Fox, Jerome Galon, Robyn Gartrell, Sacha Gnjatic, Benjamin F Green, James L Gulley, Anne Hellebust, Stephen Hewitt, Travis J Hollmann, Lucas A Horn, William J Howat, Clifford C Hoyt, Shawn M Jensen, Arutha Kulasinghe, Wiem Lassoued, Steven Lott, James Mansfield, Sebastian Marwitz, George Netto, David B Page, Edwin Parra, David L Rimm, Scott J Rodig, Roberto Salgado, Denis Schapiro, Kurt A Schalper, Joel C Sunshine, Michael J Surace, Alexander S Szalay, Magdalena Thurin, Jose C Villasboas, Keith Wharton, Ignacio I Wistuba, Jennifer H Yearley, Yinyin Yuan, Geroge Zaki, James Ziai, Janis M Taube

Multiplex immunofluorescence and immunohistochemistry (mIF/IHC) are increasingly employed antibody-based technologies that use tissue sparingly and facilitate the detection of co-localized or neighboring biomarkers. Specifically, these platforms enable spatial analyses of the tumor microenvironment as well as extended applications, for example, describing normal tissue anatomy, autoimmunity, infectious diseases, etc. mIF/IHC has greatly enhanced biomarker discovery efforts, and a growing number of studies suggest superiority to traditional IHC. Standardization of staining approaches, reporting of image analysis strategies and resultant data is critical for facilitating cross-study comparisons, validation, deployment, and generalization of findings. To address this challenge, The Society for Immunotherapy of Cancer (SITC) previously published two articles providing best practice guidelines for mIF/IHC staining, image analysis, and data sharing. Here, SITC convened stakeholders to develop the third article in the series, a consensus checklist for scientific reporting of mIF/IHC data to support and complement the best practice guidelines. The checklist includes critical components of mIF/IHC applications to be defined within publications such as detailed descriptions of analytical validation; image acquisition, selection, and registration methods; and cell clustering and spatial analysis strategies, amongst others. Such information will help with data reproducibility and comparison across studies towards future drug and assay development.

多重免疫荧光和免疫组织化学(mIF/IHC)越来越多地采用基于抗体的技术,这些技术使用组织较少,有助于检测共定位或邻近的生物标志物。具体来说,这些平台使肿瘤微环境的空间分析以及扩展应用成为可能,例如描述正常组织解剖,自身免疫,传染病等。mIF/IHC大大增强了生物标志物的发现工作,越来越多的研究表明其优于传统的IHC。标准化染色方法,报告图像分析策略和结果数据对于促进交叉研究比较,验证,部署和推广结果至关重要。为了应对这一挑战,癌症免疫治疗学会(SITC)此前发表了两篇文章,提供了mIF/IHC染色、图像分析和数据共享的最佳实践指南。在此,SITC召集利益攸关方编写了该系列的第三篇文章,即mIF/IHC数据科学报告的共识清单,以支持和补充最佳实践指南。清单包括将在出版物中定义的mIF/IHC应用的关键组成部分,例如分析验证的详细描述;图像采集、选择和配准方法;细胞聚类和空间分析策略等等。这些信息将有助于数据的可重复性和跨研究的比较,以实现未来的药物和检测开发。
{"title":"Society for Immunotherapy of Cancer: Standards for Reporting of Multiplex Immunohistochemistry/Immunofluorescence Assays (STORMI).","authors":"Sam Sater, Carlo B Bifulco, Jaime Rodriguez-Canales, Joe Yeong, Guray Akturk, Michael Angelo, Carmen Ballesteros-Merino, Peter Bankhead, Subham Basu, Jorge M Blando, Saska Brajkovic, Marco Cassano, Benjamin J Chen, Ahmet F Coskun, Tricia R Cottrell, Carlos E De Andrea, Robin H Edwards, Colt Egelston, Logan L Engle, Marc S Ernstoff, Rong Fan, Michael Feldman, Bernard A Fox, Jerome Galon, Robyn Gartrell, Sacha Gnjatic, Benjamin F Green, James L Gulley, Anne Hellebust, Stephen Hewitt, Travis J Hollmann, Lucas A Horn, William J Howat, Clifford C Hoyt, Shawn M Jensen, Arutha Kulasinghe, Wiem Lassoued, Steven Lott, James Mansfield, Sebastian Marwitz, George Netto, David B Page, Edwin Parra, David L Rimm, Scott J Rodig, Roberto Salgado, Denis Schapiro, Kurt A Schalper, Joel C Sunshine, Michael J Surace, Alexander S Szalay, Magdalena Thurin, Jose C Villasboas, Keith Wharton, Ignacio I Wistuba, Jennifer H Yearley, Yinyin Yuan, Geroge Zaki, James Ziai, Janis M Taube","doi":"10.1136/jitc-2025-012280","DOIUrl":"10.1136/jitc-2025-012280","url":null,"abstract":"<p><p>Multiplex immunofluorescence and immunohistochemistry (mIF/IHC) are increasingly employed antibody-based technologies that use tissue sparingly and facilitate the detection of co-localized or neighboring biomarkers. Specifically, these platforms enable spatial analyses of the tumor microenvironment as well as extended applications, for example, describing normal tissue anatomy, autoimmunity, infectious diseases, etc. mIF/IHC has greatly enhanced biomarker discovery efforts, and a growing number of studies suggest superiority to traditional IHC. Standardization of staining approaches, reporting of image analysis strategies and resultant data is critical for facilitating cross-study comparisons, validation, deployment, and generalization of findings. To address this challenge, The Society for Immunotherapy of Cancer (SITC) previously published two articles providing best practice guidelines for mIF/IHC staining, image analysis, and data sharing. Here, SITC convened stakeholders to develop the third article in the series, a consensus checklist for scientific reporting of mIF/IHC data to support and complement the best practice guidelines. The checklist includes critical components of mIF/IHC applications to be defined within publications such as detailed descriptions of analytical validation; image acquisition, selection, and registration methods; and cell clustering and spatial analysis strategies, amongst others. Such information will help with data reproducibility and comparison across studies towards future drug and assay development.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12718562/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804398","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Clinical and molecular characterization of AXL in colorectal cancer, CALGB (Alliance)/SWOG 80405 and real-world data. 结直肠癌AXL的临床和分子特征,CALGB (Alliance)/SWOG 80405和现实世界数据。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-013186
Karam Ashouri, Joshua Millstein, Yan Yang, Joanne Xiu, Shivani Soni, Pooja Mittal, Sandra Algaze, Lesly Torres-Gonzalez, Priya Jayachandran, Wu Zhang, Shannon Mumenthaler, Anthony F Shields, Richard Goldberg, Emil Lou, Benjamin A Weinberg, Thomas G Graeber, John L Marshall, Alan P Venook, Alexander Hoffmann, Stacey D Finley, Aaron S Meyer, Francesca Battaglin, Heinz-Josef Lenz

Background: AXL dysregulation is associated with both intrinsic resistance in tumor cells and reprogramming of the immune microenvironment. However, it is still unclear which patients would benefit from AXL-targeting therapies. We conducted a clinical and molecular characterization of AXL in colorectal cancer (CRC).

Methods: This study integrated real-world molecular profiling (Caris cohort; n=24,257) and randomized clinical trial data (phase III Cancer and Leukemia Group B/Southwest Oncology Group (CALGB/SWOG) 80405; n=433) to assess AXL messenger RNA expression in patients with CRC. Tumor samples underwent RNA sequencing and analysis of the immune microenvironment. AXL expression was categorized into tertiles. We assessed associations between molecular features, immune biomarkers, and clinical outcomes, including overall survival (OS) and progression-free survival (PFS), using Kaplan-Meier and Cox proportional hazards models while adjusting for relevant covariates.

Results: Elevated AXL expression correlated with increased programmed death-ligand 1 immunohistochemistry positivity (6.2% vs 2.5%, q<0.0001), immune checkpoint-related gene expression, and infiltration of immunosuppressive cell populations (T-regulatory cells, M2 macrophages, monocytes, and B cells). Pathway analyses demonstrated links between high AXL expression and epithelial-mesenchymal transition, inflammatory signaling, interferon-gamma response, and tumor necrosis factor alpha signaling. In the Caris cohort, high AXL predicted worse OS in patients treated with fluorouracil, leucovorin, and oxaliplatin/fluorouracil, leucovorin, and irinotecan (38.0 vs 34.7 months, p=0.027; HR 1.07), bevacizumab (36.8 vs 32.6 months, p=0.013; HR 1.21), and anti-epidermal growth factor receptor therapy (28.4 vs 22.2 months, p=0.005; HR 1.21), but profoundly improved OS in KRAS mutant patients treated with immunotherapy (11.6 vs 23.4 months, p=0.012; HR 0.65). CALGB/SWOG 80405 findings confirmed shorter PFS (9.2 vs 12.9 months, p=0.001; HR 1.56) and OS (24.2 vs 34.7 months, p<0.001; HR 1.68) with high AXL expression across treatment arms.

Conclusions: Elevated AXL expression in CRC correlated with an immunosuppressive microenvironment and worse outcomes across standard treatments. However, it identifies a distinct subgroup of KRAS-mutant patients who significantly benefit from immunotherapy, supporting AXL as a context-specific biomarker and therapeutic target.

背景:AXL失调与肿瘤细胞的内在抗性和免疫微环境的重编程有关。然而,目前尚不清楚哪些患者将受益于针对axl的治疗。我们对结直肠癌(CRC)中AXL的临床和分子特征进行了研究。方法:本研究整合了现实世界的分子分析(Caris队列,n= 24257)和随机临床试验数据(III期癌症和白血病B组/西南肿瘤组(CALGB/SWOG) 80405;n=433)评估结直肠癌患者AXL信使RNA的表达。对肿瘤样本进行RNA测序和免疫微环境分析。AXL的表达可分为三层。在调整相关协变量的同时,我们使用Kaplan-Meier和Cox比例风险模型评估了分子特征、免疫生物标志物和临床结果(包括总生存期(OS)和无进展生存期(PFS))之间的关联。结果:AXL表达升高与程序性死亡-配体1免疫组化阳性升高(6.2% vs 2.5%)、qAXL表达与上皮-间质转化、炎症信号、干扰素- γ反应和肿瘤坏死因子α信号传导相关。在Caris队列中,高AXL预测接受氟尿嘧啶、亚叶酸钙和奥沙利铂/氟尿嘧啶、亚叶酸钙和伊替康治疗的患者的OS更差(38.0 vs 34.7个月,p=0.027; HR 1.07)、贝伐单抗(36.8 vs 32.6个月,p=0.013; HR 1.21)和抗表皮生长因子受体治疗(28.4 vs 22.2个月,p=0.005; HR 1.21),但接受免疫治疗的KRAS突变患者的OS显著改善(11.6 vs 23.4个月,p=0.012; HR 0.65)。CALGB/SWOG 80405结果证实PFS (9.2 vs 12.9个月,p=0.001; HR 1.56)和OS (24.2 vs 34.7个月)在治疗组中pAXL表达缩短。结论:CRC中AXL表达升高与免疫抑制微环境相关,标准治疗的结果更差。然而,它确定了kras突变患者的一个独特亚组,这些患者从免疫治疗中显着受益,支持AXL作为特定环境的生物标志物和治疗靶点。
{"title":"Clinical and molecular characterization of <i>AXL</i> in colorectal cancer, CALGB (Alliance)/SWOG 80405 and real-world data.","authors":"Karam Ashouri, Joshua Millstein, Yan Yang, Joanne Xiu, Shivani Soni, Pooja Mittal, Sandra Algaze, Lesly Torres-Gonzalez, Priya Jayachandran, Wu Zhang, Shannon Mumenthaler, Anthony F Shields, Richard Goldberg, Emil Lou, Benjamin A Weinberg, Thomas G Graeber, John L Marshall, Alan P Venook, Alexander Hoffmann, Stacey D Finley, Aaron S Meyer, Francesca Battaglin, Heinz-Josef Lenz","doi":"10.1136/jitc-2025-013186","DOIUrl":"10.1136/jitc-2025-013186","url":null,"abstract":"<p><strong>Background: </strong><i>AXL</i> dysregulation is associated with both intrinsic resistance in tumor cells and reprogramming of the immune microenvironment. However, it is still unclear which patients would benefit from AXL-targeting therapies. We conducted a clinical and molecular characterization of <i>AXL</i> in colorectal cancer (CRC).</p><p><strong>Methods: </strong>This study integrated real-world molecular profiling (Caris cohort; n=24,257) and randomized clinical trial data (phase III Cancer and Leukemia Group B/Southwest Oncology Group (CALGB/SWOG) 80405; n=433) to assess <i>AXL</i> messenger RNA expression in patients with CRC. Tumor samples underwent RNA sequencing and analysis of the immune microenvironment. <i>AXL</i> expression was categorized into tertiles. We assessed associations between molecular features, immune biomarkers, and clinical outcomes, including overall survival (OS) and progression-free survival (PFS), using Kaplan-Meier and Cox proportional hazards models while adjusting for relevant covariates.</p><p><strong>Results: </strong>Elevated <i>AXL</i> expression correlated with increased programmed death-ligand 1 immunohistochemistry positivity (6.2% vs 2.5%, q<0.0001), immune checkpoint-related gene expression, and infiltration of immunosuppressive cell populations (T-regulatory cells, M2 macrophages, monocytes, and B cells). Pathway analyses demonstrated links between high <i>AXL</i> expression and epithelial-mesenchymal transition, inflammatory signaling, interferon-gamma response, and tumor necrosis factor alpha signaling. In the Caris cohort, high <i>AXL</i> predicted worse OS in patients treated with fluorouracil, leucovorin, and oxaliplatin/fluorouracil, leucovorin, and irinotecan (38.0 vs 34.7 months, p=0.027; HR 1.07), bevacizumab (36.8 vs 32.6 months, p=0.013; HR 1.21), and anti-epidermal growth factor receptor therapy (28.4 vs 22.2 months, p=0.005; HR 1.21), but profoundly improved OS in <i>KRAS</i> mutant patients treated with immunotherapy (11.6 vs 23.4 months, p=0.012; HR 0.65). CALGB/SWOG 80405 findings confirmed shorter PFS (9.2 vs 12.9 months, p=0.001; HR 1.56) and OS (24.2 vs 34.7 months, p<0.001; HR 1.68) with high <i>AXL</i> expression across treatment arms.</p><p><strong>Conclusions: </strong>Elevated <i>AXL</i> expression in CRC correlated with an immunosuppressive microenvironment and worse outcomes across standard treatments. However, it identifies a distinct subgroup of <i>KRAS</i>-mutant patients who significantly benefit from immunotherapy, supporting <i>AXL</i> as a context-specific biomarker and therapeutic target.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719898/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Phase 1 study evaluating the safety, preliminary efficacy, and pharmacodynamics of recombinant interleukin-15 in combination with nivolumab and ipilimumab in patients with refractory cancers. 评估重组白细胞介素-15联合纳武单抗和伊匹单抗治疗难治性癌症患者的安全性、初步疗效和药效学的i期研究。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-013252
Jibran Ahmed, Geraldine O'Sullivan Coyne, Larry V Rubinstein, Sarah J Shin, Naoko Takebe, Ashley Bruns, Brooke Augustine, Jessica Mukherjee, Ning Ma, Kristin Fino, King Leung Fung, Katherine V Ferry-Galow, Barry C Johnson, Ralph E Parchment, Howard Streicher, James H Doroshow, Alice P Chen

Purpose: Combinations of immune checkpoint inhibitors and cytokine therapies have shown promise in cancer immunotherapy. We aimed to evaluate the safety, tolerability, and preliminary efficacy of recombinant human interleukin-15 (rhIL-15) in combination with nivolumab and ipilimumab in patients with advanced, refractory cancers.

Methods: This open-label, non-randomized study employed a 3+3 dose-escalation design to determine the maximum tolerated dose (MTD) of rhIL-15 combined with fixed doses of nivolumab and ipilimumab. Safety and tolerability were assessed according to Common Terminology Criteria for Adverse Events (CTCAE) criteria, and preliminary efficacy was assessed according to Response Evaluation Criteria in Solid Tumors (RECIST) and immune-related RECIST (iRECIST) criteria. Pharmacodynamic studies evaluated changes in immune cell populations in peripheral blood and paired baseline and on-treatment tumor biopsies.

Results: Thirty-one patients were enrolled, with a median age of 56 years (range: 24-81 years). Five patients received rhIL-15 with either nivolumab or ipilimumab in safety run-in cohorts, 26 patients received rhIL-15 with nivolumab plus ipilimumab. The most common cancer types were gastrointestinal (n=7) or gynecologic (n=5). The MTD was 1 µg/kg/day rhIL-15, 240 mg nivolumab, and 1 mg/kg ipilimumab. The triplet combination showed a manageable safety profile; the most common treatment-related adverse events (trAEs) were chills (20/26, 77%), fever (18/26, 69%), injection site reaction (15/26, 58%), anemia (14/26, 54%), and fatigue (14/26, 54%). Lymphopenia (4/26, 15%) was the most common grade 3/4 trAE. Pharmacodynamic analysis of tumor biopsies revealed increases in CD8+, CD8+CD3ζpY142+ (ie, activated CD8+T cells), PD-1+CD3+, and CD45RO+CD3+ (ie, memory T cells) in some patients. Tumor infiltration of natural killer (NK) and γδ T cells was also observed. NK cell populations in peripheral blood were modulated by rhIL-15. 11 of 26 patients (42%) had stable disease as a best response on the triplet regimen. A partial response was measured after cycle 1 in one patient (4%) with cholangiocarcinoma and confirmed after cycle 2; this patient completed 16 cycles.

Conclusions: The combination of rhIL-15 with nivolumab and ipilimumab was safe and induced changes in immune cell populations in some patients. However, preliminary signs of efficacy were limited. Pharmacodynamic findings may support further clinical development of this combination with alternative dosing regimens or combinations of rhIL-15 with other therapeutic modalities.

目的:联合免疫检查点抑制剂和细胞因子治疗在癌症免疫治疗中显示出希望。我们旨在评估重组人白细胞介素-15 (rhIL-15)联合nivolumab和ipilimumab治疗晚期难治性癌症患者的安全性、耐受性和初步疗效。方法:这项开放标签、非随机研究采用3+3剂量递增设计来确定rhIL-15联合固定剂量的纳武单抗和伊匹单抗的最大耐受剂量(MTD)。根据不良事件通用术语标准(CTCAE)评估安全性和耐受性,根据实体瘤反应评价标准(RECIST)和免疫相关RECIST (iRECIST)标准评估初步疗效。药效学研究评估了外周血免疫细胞群的变化,并对基线和治疗期间的肿瘤活检进行了配对。结果:31例患者入组,中位年龄56岁(范围:24-81岁)。在安全磨合队列中,5名患者接受了rhIL-15联合纳武单抗或伊匹单抗,26名患者接受了rhIL-15联合纳武单抗和伊匹单抗。最常见的癌症类型是胃肠道(n=7)或妇科(n=5)。MTD为1µg/kg/天rhIL-15、240 mg纳武单抗和1 mg/kg伊匹单抗。三联剂组合显示出可控的安全性;最常见的治疗相关不良事件(trAEs)为寒战(20/ 26,77%)、发热(18/ 26,69%)、注射部位反应(15/ 26,58%)、贫血(14/ 26,54%)和疲劳(14/ 26,54%)。淋巴细胞减少(4/ 26,15 %)是最常见的3/4级trAE。肿瘤活检的药效学分析显示,在一些患者中CD8+、CD8+CD3ζpY142+(即活化的CD8+T细胞)、PD-1+CD3+和CD45RO+CD3+(即记忆T细胞)增加。自然杀伤细胞(NK)和γδ T细胞浸润肿瘤。rhIL-15对外周血NK细胞群有调节作用。26例患者中有11例(42%)病情稳定,这是三联方案的最佳反应。1例胆管癌患者(4%)在第1周期后获得部分缓解,并在第2周期后得到证实;该患者完成了16个周期。结论:rhIL-15联合纳武单抗和伊匹单抗是安全的,并且在一些患者中引起免疫细胞群的变化。然而,初步疗效迹象有限。药效学研究结果可能支持这种与替代给药方案或rhIL-15与其他治疗方式联合使用的进一步临床开发。
{"title":"Phase 1 study evaluating the safety, preliminary efficacy, and pharmacodynamics of recombinant interleukin-15 in combination with nivolumab and ipilimumab in patients with refractory cancers.","authors":"Jibran Ahmed, Geraldine O'Sullivan Coyne, Larry V Rubinstein, Sarah J Shin, Naoko Takebe, Ashley Bruns, Brooke Augustine, Jessica Mukherjee, Ning Ma, Kristin Fino, King Leung Fung, Katherine V Ferry-Galow, Barry C Johnson, Ralph E Parchment, Howard Streicher, James H Doroshow, Alice P Chen","doi":"10.1136/jitc-2025-013252","DOIUrl":"10.1136/jitc-2025-013252","url":null,"abstract":"<p><strong>Purpose: </strong>Combinations of immune checkpoint inhibitors and cytokine therapies have shown promise in cancer immunotherapy. We aimed to evaluate the safety, tolerability, and preliminary efficacy of recombinant human interleukin-15 (rhIL-15) in combination with nivolumab and ipilimumab in patients with advanced, refractory cancers.</p><p><strong>Methods: </strong>This open-label, non-randomized study employed a 3+3 dose-escalation design to determine the maximum tolerated dose (MTD) of rhIL-15 combined with fixed doses of nivolumab and ipilimumab. Safety and tolerability were assessed according to Common Terminology Criteria for Adverse Events (CTCAE) criteria, and preliminary efficacy was assessed according to Response Evaluation Criteria in Solid Tumors (RECIST) and immune-related RECIST (iRECIST) criteria. Pharmacodynamic studies evaluated changes in immune cell populations in peripheral blood and paired baseline and on-treatment tumor biopsies.</p><p><strong>Results: </strong>Thirty-one patients were enrolled, with a median age of 56 years (range: 24-81 years). Five patients received rhIL-15 with either nivolumab or ipilimumab in safety run-in cohorts, 26 patients received rhIL-15 with nivolumab plus ipilimumab. The most common cancer types were gastrointestinal (n=7) or gynecologic (n=5). The MTD was 1 µg/kg/day rhIL-15, 240 mg nivolumab, and 1 mg/kg ipilimumab. The triplet combination showed a manageable safety profile; the most common treatment-related adverse events (trAEs) were chills (20/26, 77%), fever (18/26, 69%), injection site reaction (15/26, 58%), anemia (14/26, 54%), and fatigue (14/26, 54%). Lymphopenia (4/26, 15%) was the most common grade 3/4 trAE. Pharmacodynamic analysis of tumor biopsies revealed increases in CD8+, CD8+CD3ζpY142+ (ie, activated CD8+T cells), PD-1+CD3+, and CD45RO+CD3+ (ie, memory T cells) in some patients. Tumor infiltration of natural killer (NK) and γδ T cells was also observed. NK cell populations in peripheral blood were modulated by rhIL-15. 11 of 26 patients (42%) had stable disease as a best response on the triplet regimen. A partial response was measured after cycle 1 in one patient (4%) with cholangiocarcinoma and confirmed after cycle 2; this patient completed 16 cycles.</p><p><strong>Conclusions: </strong>The combination of rhIL-15 with nivolumab and ipilimumab was safe and induced changes in immune cell populations in some patients. However, preliminary signs of efficacy were limited. Pharmacodynamic findings may support further clinical development of this combination with alternative dosing regimens or combinations of rhIL-15 with other therapeutic modalities.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12718592/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804371","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Etoposide activates CD8+ T cell anti-tumor immunity in osteosarcoma through MHC I upregulation via tumor-secreted IL-33 mediated signaling. 依托泊苷通过肿瘤分泌的IL-33介导的信号通路上调MHC I,激活骨肉瘤CD8+ T细胞抗肿瘤免疫。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-012591
Xin He, Hanjun Li, Haoyu Wang, Dongqing Zuo, Haoru Dong, Haoran Mu, Binghui Yang, Yining Tao, Xiyu Yang, Bowen Zhao, Tao Zhang, Yafei Jiang, Zhuoying Wang, Hongsheng Wang, Liu Yang, Yingqi Hua, Zhengdong Cai, Chongren Wang, Mengxiong Sun, Jun Gui, Wei Sun

Background: Osteosarcoma patients with high propensity for metastasis and recurrence generally encounter a poor prognosis. Despite the extensive exploration of immunotherapy, particularly the anti-programmed cell death protein 1 (anti-PD-1) antibody, in clinical trials, the efficacy remains unsatisfactory. A more profound comprehension of the resistance mechanisms and the development of innovative therapeutic strategies is imperative.

Methods: A screening was performed for drugs capable of upregulating major histocompatibility class I (MHC I) expression among clinically common drugs. The effects of the drug on both T cells and tumor cells, as well as its combination efficacy with anti-PD-1 antibody, were studied in vitro and in vivo osteosarcoma models. The molecular mechanisms underlying these biological processes were explored via RNA sequencing analysis.

Results: Etoposide was shown to upregulate the MHC I expression in osteosarcoma cells, thereby enhancing the cytotoxicity of CD8+ T cells. Interleukin-33 (IL-33) played a dominant role in etoposide-activated anti-tumor immune response. Etoposide promoted the secretion of IL-33 and augmented the expression of IL-33 binding suppression of tumorigenicity 2 (ST2) receptor, which activated the nuclear factor kappa-B signaling pathway and resulted in MHC I upregulation. Furthermore, etoposide was demonstrated to improve the therapeutic efficacy of anti-PD-1 antibody.

Conclusions: This study revealed the molecular mechanism underlying etoposide-activated CD8+ T cell anti-tumor immunity. The combination of Etoposide and anti-PD-1 antibody has the potential to benefit patients with advanced osteosarcoma.

背景:骨肉瘤患者具有高转移和复发倾向,通常预后较差。尽管对免疫疗法,特别是抗程序性细胞死亡蛋白1 (anti-PD-1)抗体进行了广泛的探索,但在临床试验中,其疗效仍不理想。对耐药机制的更深刻理解和创新治疗策略的发展是势在必行的。方法:筛选临床常用药物中能够上调主要组织相容性I类(MHC I)表达的药物。在体外和体内骨肉瘤模型中研究了该药对T细胞和肿瘤细胞的作用,以及与抗pd -1抗体的联合疗效。通过RNA测序分析,探索了这些生物过程的分子机制。结果:依托泊苷可上调骨肉瘤细胞MHC I的表达,从而增强CD8+ T细胞的细胞毒性。白细胞介素-33 (IL-33)在依托泊苷激活的抗肿瘤免疫应答中起主导作用。依托泊苷促进IL-33分泌,增强IL-33结合抑制致瘤性2 (tumor origicity 2, ST2)受体的表达,激活核因子kappa-B信号通路,导致MHC I上调。此外,依托泊苷被证明可以提高抗pd -1抗体的治疗效果。结论:本研究揭示了依托泊苷激活CD8+ T细胞抗肿瘤免疫的分子机制。依托泊苷联合抗pd -1抗体有可能使晚期骨肉瘤患者受益。
{"title":"Etoposide activates CD8<sup>+</sup> T cell anti-tumor immunity in osteosarcoma through MHC I upregulation via tumor-secreted IL-33 mediated signaling.","authors":"Xin He, Hanjun Li, Haoyu Wang, Dongqing Zuo, Haoru Dong, Haoran Mu, Binghui Yang, Yining Tao, Xiyu Yang, Bowen Zhao, Tao Zhang, Yafei Jiang, Zhuoying Wang, Hongsheng Wang, Liu Yang, Yingqi Hua, Zhengdong Cai, Chongren Wang, Mengxiong Sun, Jun Gui, Wei Sun","doi":"10.1136/jitc-2025-012591","DOIUrl":"10.1136/jitc-2025-012591","url":null,"abstract":"<p><strong>Background: </strong>Osteosarcoma patients with high propensity for metastasis and recurrence generally encounter a poor prognosis. Despite the extensive exploration of immunotherapy, particularly the anti-programmed cell death protein 1 (anti-PD-1) antibody, in clinical trials, the efficacy remains unsatisfactory. A more profound comprehension of the resistance mechanisms and the development of innovative therapeutic strategies is imperative.</p><p><strong>Methods: </strong>A screening was performed for drugs capable of upregulating major histocompatibility class I (MHC I) expression among clinically common drugs. The effects of the drug on both T cells and tumor cells, as well as its combination efficacy with anti-PD-1 antibody, were studied in vitro and in vivo osteosarcoma models. The molecular mechanisms underlying these biological processes were explored via RNA sequencing analysis.</p><p><strong>Results: </strong>Etoposide was shown to upregulate the MHC I expression in osteosarcoma cells, thereby enhancing the cytotoxicity of CD8<sup>+</sup> T cells. Interleukin-33 (IL-33) played a dominant role in etoposide-activated anti-tumor immune response. Etoposide promoted the secretion of IL-33 and augmented the expression of IL-33 binding suppression of tumorigenicity 2 (ST2) receptor, which activated the nuclear factor kappa-B signaling pathway and resulted in MHC I upregulation. Furthermore, etoposide was demonstrated to improve the therapeutic efficacy of anti-PD-1 antibody.</p><p><strong>Conclusions: </strong>This study revealed the molecular mechanism underlying etoposide-activated CD8<sup>+</sup> T cell anti-tumor immunity. The combination of Etoposide and anti-PD-1 antibody has the potential to benefit patients with advanced osteosarcoma.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12718598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
TGF-βRII/IL-15 immunotherapeutic complex targets exhausted CD8+ T-cell subsets in lymph nodes and tumors. TGF-βRII/IL-15免疫治疗复合物靶向淋巴结和肿瘤中耗尽的CD8+ t细胞亚群。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-013533
Varghese K George, Hing C Wong, Martin Felices, Mark P Rubinstein, Niraj Shrestha, Natalia Valderrama, Hamidreza Farzaneh, Lin Kong, Crystal Gilkes, Alyssa Thompson, Megan Larson, Bai Liu, Xiaoyun Zhu, Gilles Michel Leclerc, Dongjun Chung, Juan Xie, Jordan Krull, Lucas Gomez, Leah Kanakaraj, Ann Bialik, Jack O Egan, Lijing You, Xianglan Liu, Rachael Teodorescu, Ana K Karen Gutierrez, Christian Echeverri, Reynier Rodriguez, Zheng Wang, Jilan Xing, Neicy Gonzalez, Pallavi Chaturvedi, Manish R Patel, Rose Wangen, Qing Cao, Peter R Rhode, Jeffrey S Miller, Melissa A Geller

Background: Stem-like progenitor exhausted CD8+ T cells (TPEX), located within the tumor-draining lymph nodes (TDLNs), are responsible for maintaining tumor-specific responses in cancer. Although cytokines such as interleukin (IL)-15 are known to expand CD8+ T-cell subsets, transforming growth factor (TGF)-β in the TDLN is known to arrest the egress of these TPEX to the tumor microenvironment. We hypothesized that combining IL-15 stimulatory and TGF-β blocking activity would boost antitumor responses mediated by TPEX in the TDLN.

Methods: We developed a bifunctional TGF-βRII/IL-15 protein complex (HCW9218) and evaluated its antitumor activity in two murine models of melanoma and breast cancer. Peripheral blood, TDLN and tumor-infiltrating CD8+ T cells were characterized by flow cytometry following a single subcutaneous dose (s.c.) of HCW9218. Transcription profiling of CD8+ T cells in both murine models was performed. Synergistic activity of HCW9218 with immune-checkpoint inhibitors (ICIs) was evaluated. Finally, safety and immune profiling in patients with chemo-refractory/relapsed solid tumors was performed in a Phase 1 dose-escalating trial.

Results: HCW9218 was capable of localizing to the TDLNs and tumors after s.c. administration, neutralized TGF-β, expanded TPEX in TDLNs, increased chemokine-expressing effectors in peripheral circulation and promoted their infiltration into murine tumors. These data were corroborated in RNA sequencing analysis of TDLNs. ICIs significantly enhanced the effects of HCW9218 on TPEX and synergistically improved HCW9218 antitumor efficacy in melanoma and reduced spontaneous lung metastasis in breast cancer models. In a Phase 1 clinical trial, HCW9218 monotherapy was well-tolerated, reduced serum TGF-β levels, promoted and sustained CD8+ T-cell expansion in peripheral blood and CD8+ T-cell infiltration in tumor biopsies. Stable disease was reported for four of six subjects (67%) with advanced ovarian cancer treated with HCW9218.

Conclusions: Our findings demonstrate that combination therapy targeting immune cells critical for antitumor responses and blocking immune-suppressive environment significantly improves antitumor therapeutic efficacy. These findings provide a strong basis for using HCW9218 to enhance the efficacy of ICIs against solid tumors in the clinical setting.

背景:位于肿瘤引流淋巴结(tdln)内的干细胞样祖耗尽CD8+ T细胞(TPEX)在癌症中负责维持肿瘤特异性反应。虽然已知白细胞介素(IL)-15等细胞因子可扩增CD8+ t细胞亚群,但已知TDLN中的转化生长因子(TGF)-β可阻止这些TPEX向肿瘤微环境的输出。我们假设结合IL-15刺激和TGF-β阻断活性可以增强TPEX在TDLN中介导的抗肿瘤反应。方法:我们开发了一种双功能TGF-βRII/IL-15蛋白复合物(HCW9218),并在两种小鼠黑色素瘤和乳腺癌模型中评估其抗肿瘤活性。单次皮下注射HCW9218后,流式细胞术检测外周血、TDLN和肿瘤浸润的CD8+ T细胞的特征。对两种小鼠模型的CD8+ T细胞进行转录谱分析。评估了HCW9218与免疫检查点抑制剂(ICIs)的协同活性。最后,在1期剂量递增试验中,对化疗难治性/复发实体瘤患者进行了安全性和免疫分析。结果:HCW9218给药后能够定位于tdln和肿瘤,中和TGF-β,增加tdln中的TPEX,增加外周循环中趋化因子表达效应物,促进其向小鼠肿瘤浸润。这些数据在tdln的RNA测序分析中得到证实。ICIs显著增强了HCW9218对TPEX的作用,协同提高了HCW9218在黑色素瘤中的抗肿瘤疗效,降低了乳腺癌模型的自发性肺转移。在一项1期临床试验中,HCW9218单药治疗耐受性良好,可降低血清TGF-β水平,促进和维持外周血CD8+ t细胞扩增和肿瘤活检中CD8+ t细胞浸润。经HCW9218治疗的6例晚期卵巢癌患者中有4例(67%)病情稳定。结论:我们的研究结果表明,靶向抗肿瘤反应关键免疫细胞和阻断免疫抑制环境的联合治疗可显著提高抗肿瘤治疗效果。这些发现为临床应用HCW9218增强ICIs抗实体瘤的疗效提供了强有力的基础。
{"title":"TGF-βRII/IL-15 immunotherapeutic complex targets exhausted CD8<sup>+</sup> T-cell subsets in lymph nodes and tumors.","authors":"Varghese K George, Hing C Wong, Martin Felices, Mark P Rubinstein, Niraj Shrestha, Natalia Valderrama, Hamidreza Farzaneh, Lin Kong, Crystal Gilkes, Alyssa Thompson, Megan Larson, Bai Liu, Xiaoyun Zhu, Gilles Michel Leclerc, Dongjun Chung, Juan Xie, Jordan Krull, Lucas Gomez, Leah Kanakaraj, Ann Bialik, Jack O Egan, Lijing You, Xianglan Liu, Rachael Teodorescu, Ana K Karen Gutierrez, Christian Echeverri, Reynier Rodriguez, Zheng Wang, Jilan Xing, Neicy Gonzalez, Pallavi Chaturvedi, Manish R Patel, Rose Wangen, Qing Cao, Peter R Rhode, Jeffrey S Miller, Melissa A Geller","doi":"10.1136/jitc-2025-013533","DOIUrl":"10.1136/jitc-2025-013533","url":null,"abstract":"<p><strong>Background: </strong>Stem-like progenitor exhausted CD8<sup>+</sup> T cells (T<sub>PEX</sub>), located within the tumor-draining lymph nodes (TDLNs), are responsible for maintaining tumor-specific responses in cancer. Although cytokines such as interleukin (IL)-15 are known to expand CD8<sup>+</sup> T-cell subsets, transforming growth factor (TGF)-β in the TDLN is known to arrest the egress of these T<sub>PEX</sub> to the tumor microenvironment. We hypothesized that combining IL-15 stimulatory and TGF-β blocking activity would boost antitumor responses mediated by T<sub>PEX</sub> in the TDLN.</p><p><strong>Methods: </strong>We developed a bifunctional TGF-βRII/IL-15 protein complex (HCW9218) and evaluated its antitumor activity in two murine models of melanoma and breast cancer. Peripheral blood, TDLN and tumor-infiltrating CD8<sup>+</sup> T cells were characterized by flow cytometry following a single subcutaneous dose (s.c.) of HCW9218. Transcription profiling of CD8<sup>+</sup> T cells in both murine models was performed. Synergistic activity of HCW9218 with immune-checkpoint inhibitors (ICIs) was evaluated. Finally, safety and immune profiling in patients with chemo-refractory/relapsed solid tumors was performed in a Phase 1 dose-escalating trial.</p><p><strong>Results: </strong>HCW9218 was capable of localizing to the TDLNs and tumors after s.c. administration, neutralized TGF-β, expanded T<sub>PEX</sub> in TDLNs, increased chemokine-expressing effectors in peripheral circulation and promoted their infiltration into murine tumors. These data were corroborated in RNA sequencing analysis of TDLNs. ICIs significantly enhanced the effects of HCW9218 on T<sub>PEX</sub> and synergistically improved HCW9218 antitumor efficacy in melanoma and reduced spontaneous lung metastasis in breast cancer models. In a Phase 1 clinical trial, HCW9218 monotherapy was well-tolerated, reduced serum TGF-β levels, promoted and sustained CD8<sup>+</sup> T-cell expansion in peripheral blood and CD8<sup>+</sup> T-cell infiltration in tumor biopsies. Stable disease was reported for four of six subjects (67%) with advanced ovarian cancer treated with HCW9218.</p><p><strong>Conclusions: </strong>Our findings demonstrate that combination therapy targeting immune cells critical for antitumor responses and blocking immune-suppressive environment significantly improves antitumor therapeutic efficacy. These findings provide a strong basis for using HCW9218 to enhance the efficacy of ICIs against solid tumors in the clinical setting.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719900/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804339","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Multi-omics analysis reveals differential benefits of immunotherapy±chemotherapy based on detailed smoking history in advanced non-small cell lung cancer. 多组学分析显示基于详细吸烟史的免疫治疗±化疗对晚期非小细胞肺癌的不同益处。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-21 DOI: 10.1136/jitc-2025-012205
Xinan Wang, Biagio Ricciuti, Arielle Elkrief, Joao V Alessi, Alessandro Di Federico, Federica Pecci, Mizuki Nishino, Doga Gulhan, Guruprasad Ananda, Scott J Rodig, Lynette Sholl, Bruce Johnson, Xihong Lin, Adam Schoenfeld, Mark M Awad, David Christiani

Background: Despite immunotherapy±chemotherapy has transformed the therapeutic landscape for patients with non-small cell lung cancer (NSCLC), critical questions remain regarding how detailed smoking history affects the evolving treatment options and the underlying molecular mechanisms driving these effects.

Methods: We analyzed 4157 patients with advanced NSCLC who were treated with immunotherapy monotherapy (IO alone) (n=2768) or chemoimmunotherapy (chemo-IO) (n=1389) at the Dana-Farber Cancer Institute and Memorial Sloan Kettering Cancer Center (2010-2023). Associations between detailed smoking history (status and cumulative pack-years) and clinical outcomes were assessed using multivariable analyses. First-line chemo-IO versus IO alone was compared in programmed death receptor ligand 1 (PD-L1) Tumor Proportion Score (TPS) of ≥50% EGFR/ALK wild-type patients. Tobacco smoking-related mutational signature (TSMS) was inferred from the targeted next generation sequencing (NGS) panel. We investigated relationships between detailed smoking history and tumor genomics/transcriptomics, PD-L1 expression, tumor-infiltrating lymphocytes, and circulating plasma proteomics.

Results: In patients receiving IO alone, both smoking status and intensity showed dose-dependent associations with improved response and survival outcomes. In contrast, among patients receiving chemo-IO, smoking history did not affect initial response but patients with active (HR=0.73, 95% CI 0.57 to 0.94, p=0.01) and heavy tobacco use (HR=0.76, 95% CI 0.62 to 0.93, p=0.001) showed improved progression-free survival (PFS), and a trend toward improved overall survival (OS). Importantly, these associations remained independent of STK11, KEAP1, and KRAS co-mutation status. In patients with PD-L1 TPS of ≥50% lacking EGFR/ALK alterations, patients who do not smoke derived significant benefit from first-line chemo-IO versus IO alone with higher response rates (70.0% vs 23.9%, p=0.001), prolonged PFS (median PFS 9.5 vs 3.7 months, HR=0.51, 95% CI 0.27 to 0.95, p=0.04), and a trend toward prolonged OS, while patients who smoke showed comparable outcomes with either strategy. TSMS independently predicted improved outcomes of IO alone, even after tumor mutational burden (TMB) adjustment. Molecular analyses revealed associations between tobacco use and higher TMB, increased tumor-infiltrating lymphocytes (CD8+, PD-1+, CD8+PD-1+, FOXP3+) and distinct plasma protein profiles involved in immune signaling pathways (CCL7, CXCL17, CDCP1, TNFRSF6B).

Conclusions: Detailed smoking history provides crucial insights for optimizing IO selection in advanced NSCLC through mechanistic alterations in both the tumor microenvironment and systemic plasma protein profiles.

背景:尽管免疫治疗+化疗已经改变了非小细胞肺癌(NSCLC)患者的治疗前景,但关于详细的吸烟史如何影响不断发展的治疗方案以及驱动这些效果的潜在分子机制的关键问题仍然存在。方法:我们分析了2010-2023年在Dana-Farber癌症研究所和Memorial Sloan Kettering癌症中心接受免疫治疗单药(IO) (n=2768)或化疗免疫治疗(chemo-IO) (n=1389)的4157例晚期非小细胞肺癌患者。详细的吸烟史(状态和累积包年)与临床结果之间的关系使用多变量分析进行评估。在≥50% EGFR/ALK野生型患者的程序性死亡受体配体1 (PD-L1)肿瘤比例评分(TPS)中,比较一线化疗-IO与单独IO。烟草吸烟相关的突变特征(TSMS)是从目标下一代测序(NGS)面板推断出来的。我们研究了详细的吸烟史与肿瘤基因组学/转录组学、PD-L1表达、肿瘤浸润淋巴细胞和循环血浆蛋白质组学之间的关系。结果:在单独接受IO治疗的患者中,吸烟状态和吸烟强度与改善的反应和生存结果均显示剂量依赖关系。相反,在接受化疗的患者中,吸烟史不影响初始反应,但活跃(HR=0.73, 95% CI 0.57至0.94,p=0.01)和重度吸烟(HR=0.76, 95% CI 0.62至0.93,p=0.001)的患者无进展生存期(PFS)得到改善,总生存期(OS)有改善的趋势。重要的是,这些关联仍然独立于STK11、KEAP1和KRAS共突变状态。在缺乏EGFR/ALK改变的PD-L1 TPS≥50%的患者中,不吸烟的患者从一线化疗-IO与单独IO相比获得了显着益处,具有更高的缓解率(70.0% vs 23.9%, p=0.001),延长PFS(中位PFS 9.5 vs 3.7个月,HR=0.51, 95% CI 0.27至0.95,p=0.04),并有延长OS的趋势,而吸烟患者的结果与两种策略相当。即使在肿瘤突变负荷(TMB)调整后,TSMS也能独立预测IO单独改善的结果。分子分析显示,烟草使用与TMB升高、肿瘤浸润淋巴细胞(CD8+、PD-1+、CD8+PD-1+、FOXP3+)增加以及参与免疫信号通路的不同血浆蛋白谱(CCL7、CXCL17、CDCP1、TNFRSF6B)有关。结论:详细的吸烟史通过肿瘤微环境和全身血浆蛋白谱的机制改变,为优化晚期非小细胞肺癌的IO选择提供了重要的见解。
{"title":"Multi-omics analysis reveals differential benefits of immunotherapy±chemotherapy based on detailed smoking history in advanced non-small cell lung cancer.","authors":"Xinan Wang, Biagio Ricciuti, Arielle Elkrief, Joao V Alessi, Alessandro Di Federico, Federica Pecci, Mizuki Nishino, Doga Gulhan, Guruprasad Ananda, Scott J Rodig, Lynette Sholl, Bruce Johnson, Xihong Lin, Adam Schoenfeld, Mark M Awad, David Christiani","doi":"10.1136/jitc-2025-012205","DOIUrl":"10.1136/jitc-2025-012205","url":null,"abstract":"<p><strong>Background: </strong>Despite immunotherapy±chemotherapy has transformed the therapeutic landscape for patients with non-small cell lung cancer (NSCLC), critical questions remain regarding how detailed smoking history affects the evolving treatment options and the underlying molecular mechanisms driving these effects.</p><p><strong>Methods: </strong>We analyzed 4157 patients with advanced NSCLC who were treated with immunotherapy monotherapy (IO alone) (n=2768) or chemoimmunotherapy (chemo-IO) (n=1389) at the Dana-Farber Cancer Institute and Memorial Sloan Kettering Cancer Center (2010-2023). Associations between detailed smoking history (status and cumulative pack-years) and clinical outcomes were assessed using multivariable analyses. First-line chemo-IO versus IO alone was compared in programmed death receptor ligand 1 (PD-L1) Tumor Proportion Score (TPS) of ≥50% <i>EGFR/ALK</i> wild-type patients. Tobacco smoking-related mutational signature (TSMS) was inferred from the targeted next generation sequencing (NGS) panel. We investigated relationships between detailed smoking history and tumor genomics/transcriptomics, PD-L1 expression, tumor-infiltrating lymphocytes, and circulating plasma proteomics.</p><p><strong>Results: </strong>In patients receiving IO alone, both smoking status and intensity showed dose-dependent associations with improved response and survival outcomes. In contrast, among patients receiving chemo-IO, smoking history did not affect initial response but patients with active (HR=0.73, 95% CI 0.57 to 0.94, p=0.01) and heavy tobacco use (HR=0.76, 95% CI 0.62 to 0.93, p=0.001) showed improved progression-free survival (PFS), and a trend toward improved overall survival (OS). Importantly, these associations remained independent of <i>STK11</i>, <i>KEAP1</i>, and <i>KRAS</i> co-mutation status. In patients with PD-L1 TPS of ≥50% lacking <i>EGFR/ALK</i> alterations, patients who do not smoke derived significant benefit from first-line chemo-IO versus IO alone with higher response rates (70.0% vs 23.9%, p=0.001), prolonged PFS (median PFS 9.5 vs 3.7 months, HR=0.51, 95% CI 0.27 to 0.95, p=0.04), and a trend toward prolonged OS, while patients who smoke showed comparable outcomes with either strategy. TSMS independently predicted improved outcomes of IO alone, even after tumor mutational burden (TMB) adjustment. Molecular analyses revealed associations between tobacco use and higher TMB, increased tumor-infiltrating lymphocytes (CD8<sup>+</sup>, PD-1<sup>+</sup>, CD8<sup>+</sup>PD-1<sup>+</sup>, FOXP3<sup>+</sup>) and distinct plasma protein profiles involved in immune signaling pathways (CCL7, CXCL17, CDCP1, TNFRSF6B).</p><p><strong>Conclusions: </strong>Detailed smoking history provides crucial insights for optimizing IO selection in advanced NSCLC through mechanistic alterations in both the tumor microenvironment and systemic plasma protein profiles.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12719888/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
PINK1 dysfunction in hepatocellular carcinoma fosters immune evasion and disease progression by promoting neutrophil infiltration. 肝细胞癌中的PINK1功能障碍通过促进中性粒细胞浸润促进免疫逃避和疾病进展。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-18 DOI: 10.1136/jitc-2025-013079
Zhiyun Gu, Tongwang Yang, Yuanli Ni, Yuanze Zhou, Cheng Qian, Na Zhuang

Background: Despite breakthroughs in immune checkpoint blockade (ICB) for advanced hepatocellular carcinoma (HCC), tumor immune evasion remains a major limitation to immunotherapy efficacy. PTEN-induced putative kinase 1 (PINK1) has highlighted roles in immune system regulation and tumor progression, but its specific role in HCC and impact on ICB response remains unclear. This study aimed to elucidate how PINK1 dysfunction enables HCC adaptation to ICB-induced immune attacks and identify potential therapeutic targets.

Methods: Online patient dataset analyses were performed to evaluate the impact of reduced PINK1 expression on clinical prognosis in HCC. An orthotopic HCC tumor cell mouse model was established. Single-cell RNA sequencing and flow cytometry were employed to explore the immune characteristics and remodeling of the tumor microenvironment in PINK1-deficient HCC. Neutrophils and T cells were isolated, stimulated, and/or cultured for ex vivo functional assays. Gene Expression Omnibus database analyses were performed to identify key genes involved in HCC resistance to ICB.

Results: We identified PINK1 as a key mediator of ICB resistance in HCC. We found that PINK1 expression was significantly reduced in ICB-resistant HCC and a reduced PINK1 signature was associated with poorer clinical outcomes in patients with HCC. Importantly, PINK1 deficiency serves as a predictive biomarker for diminished responsiveness to immunotherapy. Function experiments have revealed that PINK1 regulates HCC progression and infiltration and activation of neutrophils and T cells. Mechanistically, the tumor cell-intrinsic downregulation of PINK1 activated the production of chemokines involved in neutrophil recruitment, and tumor-infiltrating neutrophils inhibited T cell activity. Additionally, a neutrophil-depleting antibody resensitized tumors to antiprogrammed death-ligand 1 therapy in orthotopic HCC models, further demonstrating that neutrophils are the primary population responsible for the adaptation of cancer cells with reduced PINK1 to ICB-induced immune attacks.

Conclusions: Our study uncovered PINK1 as both a predictor of the ICB response and a key mediator of immune evasion by promoting neutrophil infiltration. These results highlight that the therapeutic targeting of neutrophils may represent a viable strategy to overcome ICB resistance in HCC.

背景:尽管免疫检查点阻断(ICB)治疗晚期肝细胞癌(HCC)取得了突破,但肿瘤免疫逃避仍然是免疫治疗效果的主要限制因素。pten诱导的推定激酶1 (PINK1)在免疫系统调节和肿瘤进展中发挥了重要作用,但其在HCC中的具体作用和对ICB反应的影响尚不清楚。本研究旨在阐明PINK1功能障碍如何使HCC适应icb诱导的免疫攻击,并确定潜在的治疗靶点。方法:通过在线患者数据集分析,评估PINK1表达降低对HCC临床预后的影响。建立原位肝癌小鼠肿瘤细胞模型。采用单细胞RNA测序和流式细胞术探讨pink1缺失HCC的免疫特性和肿瘤微环境的重塑。中性粒细胞和T细胞被分离、刺激和/或培养用于离体功能测定。基因表达Omnibus数据库分析确定HCC对ICB耐药的关键基因。结果:我们发现PINK1是HCC中ICB耐药的关键介质。我们发现,在icb耐药HCC中,PINK1表达显著降低,并且PINK1信号的降低与HCC患者较差的临床结果相关。重要的是,PINK1缺乏可作为免疫治疗反应性降低的预测性生物标志物。功能实验显示,PINK1调节HCC的进展、中性粒细胞和T细胞的浸润和活化。从机制上讲,肿瘤细胞内在的PINK1下调激活了参与中性粒细胞募集的趋化因子的产生,肿瘤浸润的中性粒细胞抑制了T细胞的活性。此外,在原位HCC模型中,中性粒细胞消耗抗体使肿瘤对抗程序性死亡配体1治疗重新敏感,进一步证明中性粒细胞是导致PINK1降低的癌细胞适应icb诱导的免疫攻击的主要人群。结论:我们的研究揭示了PINK1既是ICB反应的预测因子,也是通过促进中性粒细胞浸润来逃避免疫的关键介质。这些结果强调中性粒细胞的靶向治疗可能是克服HCC中ICB耐药的可行策略。
{"title":"PINK1 dysfunction in hepatocellular carcinoma fosters immune evasion and disease progression by promoting neutrophil infiltration.","authors":"Zhiyun Gu, Tongwang Yang, Yuanli Ni, Yuanze Zhou, Cheng Qian, Na Zhuang","doi":"10.1136/jitc-2025-013079","DOIUrl":"10.1136/jitc-2025-013079","url":null,"abstract":"<p><strong>Background: </strong>Despite breakthroughs in immune checkpoint blockade (ICB) for advanced hepatocellular carcinoma (HCC), tumor immune evasion remains a major limitation to immunotherapy efficacy. PTEN-induced putative kinase 1 (PINK1) has highlighted roles in immune system regulation and tumor progression, but its specific role in HCC and impact on ICB response remains unclear. This study aimed to elucidate how PINK1 dysfunction enables HCC adaptation to ICB-induced immune attacks and identify potential therapeutic targets.</p><p><strong>Methods: </strong>Online patient dataset analyses were performed to evaluate the impact of reduced PINK1 expression on clinical prognosis in HCC. An orthotopic HCC tumor cell mouse model was established. Single-cell RNA sequencing and flow cytometry were employed to explore the immune characteristics and remodeling of the tumor microenvironment in PINK1-deficient HCC. Neutrophils and T cells were isolated, stimulated, and/or cultured for ex vivo functional assays. Gene Expression Omnibus database analyses were performed to identify key genes involved in HCC resistance to ICB.</p><p><strong>Results: </strong>We identified PINK1 as a key mediator of ICB resistance in HCC. We found that PINK1 expression was significantly reduced in ICB-resistant HCC and a reduced PINK1 signature was associated with poorer clinical outcomes in patients with HCC. Importantly, PINK1 deficiency serves as a predictive biomarker for diminished responsiveness to immunotherapy. Function experiments have revealed that PINK1 regulates HCC progression and infiltration and activation of neutrophils and T cells. Mechanistically, the tumor cell-intrinsic downregulation of PINK1 activated the production of chemokines involved in neutrophil recruitment, and tumor-infiltrating neutrophils inhibited T cell activity. Additionally, a neutrophil-depleting antibody resensitized tumors to antiprogrammed death-ligand 1 therapy in orthotopic HCC models, further demonstrating that neutrophils are the primary population responsible for the adaptation of cancer cells with reduced PINK1 to ICB-induced immune attacks.</p><p><strong>Conclusions: </strong>Our study uncovered PINK1 as both a predictor of the ICB response and a key mediator of immune evasion by promoting neutrophil infiltration. These results highlight that the therapeutic targeting of neutrophils may represent a viable strategy to overcome ICB resistance in HCC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12716600/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793676","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Journal for Immunotherapy of Cancer
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