首页 > 最新文献

Journal for Immunotherapy of Cancer最新文献

英文 中文
CD300e is a driver of the immunosuppressive tumor microenvironment and colorectal cancer progression via macrophage reprogramming. CD300e是免疫抑制肿瘤微环境和通过巨噬细胞重编程的结直肠癌进展的驱动因素。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-17 DOI: 10.1136/jitc-2025-013249
Annica Barizza, Stefania Vassallo, Laura Masatti, Mattia Laffranchi, Sofia Giacometti, Silvia Lonardi, Mattia Bugatti, Sara Coletta, Chiara Della Bella, Mario Milco D'Elios, Simone Pizzini, Antonio Rosato, William Vermi, Matteo Fassan, Gaya Spolverato, Silvano Sozzani, Enrica Calura, Roberta Sommaggio, Gaia Codolo

Background: Colorectal cancer (CRC) progression is shaped by the tumor microenvironment, particularly tumor-associated macrophages (TAMs), which often adopt immunosuppressive functions. CD300e, a myeloid receptor involved in immune regulation, has an uncharacterized role in CRC.

Methods: Functional studies were conducted in azoxymethane/dextran sodium sulfate and MC38 murine CRC models using CD300e systemic and myeloid-specific CD300e knockout mice, and adoptive transfer experiments assessed macrophage-intrinsic effects. Human studies included analysis of CD300e expression in matched tumor and normal tissue from patients with CRC and in vitro co-culture of patient-derived colon tumor organoids with monocytes to study CD300e induction and TAM polarization.

Results: In vivo, CD300e deficiency led to reduced tumor burden, enhanced major histocompatibility complex expression on TAMs, and improved T-cell responses. CD300e-deficient macrophages exhibited increased phagocytic activity, antigen presentation, and support for T-cell proliferation and cytotoxicity. Adoptive transfer confirmed that macrophage-intrinsic CD300e expression is sufficient to suppress T-cell function and promote tumor growth. In patients with CRC, CD300e is selectively upregulated in tumor-infiltrating monocytes and macrophages, driving a suppressive phenotype marked by impaired antigen presentation. Tumor-derived signals in vitro induce CD300e expression and promote a protumorigenic macrophage profile.

Conclusions: Our findings identify CD300e as a critical regulator of macrophage-mediated immune suppression in CRC and a potential target for reprogramming TAMs to enhance immunotherapy.

背景:结直肠癌(CRC)的进展是由肿瘤微环境决定的,特别是肿瘤相关巨噬细胞(tam),它们通常具有免疫抑制功能。CD300e是一种参与免疫调节的髓样受体,在结直肠癌中具有未知的作用。方法:采用CD300e全体性和骨髓特异性CD300e敲除小鼠,对偶氮甲烷/葡聚糖硫酸钠和MC38小鼠CRC模型进行功能研究,并通过过继转移实验评估巨噬细胞内在效应。人类研究包括分析CRC患者匹配肿瘤和正常组织中的CD300e表达,以及患者来源的结肠肿瘤类器官与单核细胞体外共培养,以研究CD300e诱导和TAM极化。结果:在体内,CD300e缺乏导致肿瘤负荷减轻,主要组织相容性复合物在tam上的表达增强,t细胞反应改善。cd300e缺陷巨噬细胞表现出增加的吞噬活性、抗原呈递、支持t细胞增殖和细胞毒性。过继转移证实巨噬细胞内生性CD300e表达足以抑制t细胞功能,促进肿瘤生长。在结直肠癌患者中,CD300e在肿瘤浸润的单核细胞和巨噬细胞中选择性上调,驱动以抗原呈递受损为标志的抑制性表型。肿瘤来源的信号在体外诱导CD300e表达并促进致瘤性巨噬细胞谱。结论:我们的研究结果确定CD300e是CRC中巨噬细胞介导的免疫抑制的关键调节因子,也是重编程tam以增强免疫治疗的潜在靶标。
{"title":"CD300e is a driver of the immunosuppressive tumor microenvironment and colorectal cancer progression via macrophage reprogramming.","authors":"Annica Barizza, Stefania Vassallo, Laura Masatti, Mattia Laffranchi, Sofia Giacometti, Silvia Lonardi, Mattia Bugatti, Sara Coletta, Chiara Della Bella, Mario Milco D'Elios, Simone Pizzini, Antonio Rosato, William Vermi, Matteo Fassan, Gaya Spolverato, Silvano Sozzani, Enrica Calura, Roberta Sommaggio, Gaia Codolo","doi":"10.1136/jitc-2025-013249","DOIUrl":"10.1136/jitc-2025-013249","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC) progression is shaped by the tumor microenvironment, particularly tumor-associated macrophages (TAMs), which often adopt immunosuppressive functions. CD300e, a myeloid receptor involved in immune regulation, has an uncharacterized role in CRC.</p><p><strong>Methods: </strong>Functional studies were conducted in azoxymethane/dextran sodium sulfate and MC38 murine CRC models using CD300e systemic and myeloid-specific CD300e knockout mice, and adoptive transfer experiments assessed macrophage-intrinsic effects. Human studies included analysis of CD300e expression in matched tumor and normal tissue from patients with CRC and in vitro co-culture of patient-derived colon tumor organoids with monocytes to study CD300e induction and TAM polarization.</p><p><strong>Results: </strong>In vivo, CD300e deficiency led to reduced tumor burden, enhanced major histocompatibility complex expression on TAMs, and improved T-cell responses. CD300e-deficient macrophages exhibited increased phagocytic activity, antigen presentation, and support for T-cell proliferation and cytotoxicity. Adoptive transfer confirmed that macrophage-intrinsic CD300e expression is sufficient to suppress T-cell function and promote tumor growth. In patients with CRC, CD300e is selectively upregulated in tumor-infiltrating monocytes and macrophages, driving a suppressive phenotype marked by impaired antigen presentation. Tumor-derived signals in vitro induce CD300e expression and promote a protumorigenic macrophage profile.</p><p><strong>Conclusions: </strong>Our findings identify CD300e as a critical regulator of macrophage-mediated immune suppression in CRC and a potential target for reprogramming TAMs to enhance immunotherapy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12716572/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145774590","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
Final results of ANICCA-Class II, a single arm, open-label phase II trial assessing nivolumab in tissue-specific class II expressing metastatic microsatellite stable colorectal cancer, with a parallel assessment of the immunoscore-immune checkpoint as a predictive biomarker for single-agent anti-PD-1. ANICCA-Class II的最终结果,这是一项单组,开放标签的II期试验,评估了nivolumab在组织特异性II类表达转移性微卫星稳定结直肠癌中的作用,同时评估了免疫评分-免疫检查点作为单药抗pd -1的预测性生物标志物。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-17 DOI: 10.1136/jitc-2025-012749
Gary Middleton, Charlotte Gaskell, Joshua Savage, John Bridgewater, Paul Ross, Mark Saunders, Daniel Palmer, Ruth Plummer, Sally Clive, Vicky Coyle, Anne Thomas, David Cunningham, Phillipe Taniere, Lucinda Billingham

Background: Neutralization of interferon (IFN)-γ abrogates the efficacy of anti-programmed death-ligand 1 (PD-(L)1) checkpoint inhibitors. Most epithelial cells do not constitutively express major histocompatibility complex (MHC) class II but can be induced to do so by IFN-γ. Inducible tumor-specific MHC class II (tsMHC-II) underlies responsiveness to anti-PD-(L)1. Retrospective studies show that tsMHC-II positivity associates with improved outcomes in patients treated with anti-PD-(L)1. The ANICCA-Class II single-arm Bayesian phase II trial prospectively explored whether positive tsMHC-II status could be a useful selection marker for anti-programmed cell death protein-1 (PD-1) in proficient mismatch repair colorectal cancer (pMMR CRC). In parallel, we retrospectively evaluated the potential predictive power of immunoscore-immune checkpoint (IS-IC) for outcome with single-agent immune checkpoint blockade.

Methods: Patients with histologically confirmed locally advanced/metastatic pMMR CRC with >1% MHC class II expression, Eastern Cooperative Oncology Group performance status 0-2, aged ≥18 years were eligible. Participants received 480 mg nivolumab every 28 days for up to 24 cycles. The primary outcome was durable clinical benefit (DCB) defined as participants remaining progression-free at their third trial-specific scan since treatment start (ie, at approximately 27 weeks). Secondary outcomes included progression-free survival time (PFS) and overall survival time (OS).

Results: 35 participants were treated: 65.7% of participants' cancers were tsMHC-II ≥5%. 3/35 patients achieved DCB (8.6%), estimating the true DCB rate (R) of 11% (95% credible interval 3% to 22%) with 0.002 probability that the true DCBR was >30%, below the required 0.5 to warrant further research. The higher tsMHC-II cut-point ≥5% was not more useful in predicting duration of disease stabilization. All three participants who achieved DCB had no evidence of liver metastases (LM); DCBR 23.1% in those without versus 0% in those with LM. PFS and OS were significantly greater in those without LM. There was no evidence that IS-IC high predicted for prolonged time on treatment or improved tumor growth inhibition.

Conclusions: In pMMR CRC, tsMHC-II positivity fails to identify a subset of patients with metastatic pMMR CRC obtaining potentially meaningful benefit from single-agent anti-PD-1. Although numbers are limited, there is no clear evidence that IS-IC is predictive of outcome with single-agent anti-PD-1. The poor outcome in those with LM underscores the need for therapies that overcome the systemic immunosuppression driven by LM.

背景:干扰素(IFN)-γ的中和可消除抗程序性死亡配体1 (PD-(L)1)检查点抑制剂的功效。大多数上皮细胞不构成性地表达主要组织相容性复合体(MHC) II类,但可以通过IFN-γ诱导表达。诱导肿瘤特异性MHC II类(tsMHC-II)是对抗pd -(L)1的反应性基础。回顾性研究表明,tsMHC-II阳性与抗pd -(L)1治疗的患者预后改善相关。anicca -II类单臂贝叶斯II期试验前瞻性地探讨了tsMHC-II阳性状态是否可以作为抗程序性细胞死亡蛋白-1 (PD-1)在熟练错配修复结直肠癌(pMMR CRC)中有用的选择标记。同时,我们回顾性地评估了免疫评分-免疫检查点(IS-IC)对单药免疫检查点阻断治疗结果的潜在预测能力。方法:组织学证实的局部晚期/转移性pMMR结直肠癌患者,MHCⅱ类表达>.1 %,东部肿瘤合作组表现状态0-2,年龄≥18岁。参与者每28天接受480mg纳武单抗治疗,长达24个周期。主要结果是持久临床获益(DCB),定义为自治疗开始(即约27周)以来参与者在第三次试验特异性扫描时保持无进展。次要终点包括无进展生存时间(PFS)和总生存时间(OS)。结果:35例受试者接受治疗:65.7%的受试者肿瘤tsMHC-II≥5%。3/35例患者实现DCB(8.6%),估计真实DCB率(R)为11%(95%可信区间为3% ~ 22%),真实DCBR为0.30%的概率为0.002,低于进一步研究所需的0.5。较高的tsMHC-II切点(≥5%)在预测疾病稳定持续时间方面并不更有用。所有三个达到DCB的参与者都没有肝转移(LM)的证据;无LM者DCBR为23.1%,有LM者为0%。无LM组PFS和OS显著增高。没有证据表明IS-IC高预测延长治疗时间或改善肿瘤生长抑制。结论:在pMMR CRC中,tsMHC-II阳性不能识别转移性pMMR CRC患者亚群从单药抗pd -1中获得潜在的有意义的益处。虽然数量有限,但没有明确的证据表明is - ic可以预测单药抗pd -1的预后。LM患者的不良预后强调了克服由LM驱动的全身免疫抑制的治疗的必要性。
{"title":"Final results of ANICCA-Class II, a single arm, open-label phase II trial assessing nivolumab in tissue-specific class II expressing metastatic microsatellite stable colorectal cancer, with a parallel assessment of the immunoscore-immune checkpoint as a predictive biomarker for single-agent anti-PD-1.","authors":"Gary Middleton, Charlotte Gaskell, Joshua Savage, John Bridgewater, Paul Ross, Mark Saunders, Daniel Palmer, Ruth Plummer, Sally Clive, Vicky Coyle, Anne Thomas, David Cunningham, Phillipe Taniere, Lucinda Billingham","doi":"10.1136/jitc-2025-012749","DOIUrl":"10.1136/jitc-2025-012749","url":null,"abstract":"<p><strong>Background: </strong>Neutralization of interferon (IFN)-γ abrogates the efficacy of anti-programmed death-ligand 1 (PD-(L)1) checkpoint inhibitors. Most epithelial cells do not constitutively express major histocompatibility complex (MHC) class II but can be induced to do so by IFN-γ. Inducible tumor-specific MHC class II (tsMHC-II) underlies responsiveness to anti-PD-(L)1. Retrospective studies show that tsMHC-II positivity associates with improved outcomes in patients treated with anti-PD-(L)1. The ANICCA-Class II single-arm Bayesian phase II trial prospectively explored whether positive tsMHC-II status could be a useful selection marker for anti-programmed cell death protein-1 (PD-1) in proficient mismatch repair colorectal cancer (pMMR CRC). In parallel, we retrospectively evaluated the potential predictive power of immunoscore-immune checkpoint (IS-IC) for outcome with single-agent immune checkpoint blockade.</p><p><strong>Methods: </strong>Patients with histologically confirmed locally advanced/metastatic pMMR CRC with >1% MHC class II expression, Eastern Cooperative Oncology Group performance status 0-2, aged ≥18 years were eligible. Participants received 480 mg nivolumab every 28 days for up to 24 cycles. The primary outcome was durable clinical benefit (DCB) defined as participants remaining progression-free at their third trial-specific scan since treatment start (ie, at approximately 27 weeks). Secondary outcomes included progression-free survival time (PFS) and overall survival time (OS).</p><p><strong>Results: </strong>35 participants were treated: 65.7% of participants' cancers were tsMHC-II ≥5%. 3/35 patients achieved DCB (8.6%), estimating the true DCB rate (R) of 11% (95% credible interval 3% to 22%) with 0.002 probability that the true DCBR was >30%, below the required 0.5 to warrant further research. The higher tsMHC-II cut-point ≥5% was not more useful in predicting duration of disease stabilization. All three participants who achieved DCB had no evidence of liver metastases (LM); DCBR 23.1% in those without versus 0% in those with LM. PFS and OS were significantly greater in those without LM. There was no evidence that IS-IC high predicted for prolonged time on treatment or improved tumor growth inhibition.</p><p><strong>Conclusions: </strong>In pMMR CRC, tsMHC-II positivity fails to identify a subset of patients with metastatic pMMR CRC obtaining potentially meaningful benefit from single-agent anti-PD-1. Although numbers are limited, there is no clear evidence that IS-IC is predictive of outcome with single-agent anti-PD-1. The poor outcome in those with LM underscores the need for therapies that overcome the systemic immunosuppression driven by LM.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12716601/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145774595","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
Autologous profiling reveals inter-patient heterogeneity in Vδ2+γδTIL responses to glioblastoma driven by extracellular matrix-BTN3A axis. 自体谱分析揭示了由细胞外基质- btn3a轴驱动的胶质母细胞瘤的Vδ2+γδTIL反应的患者间异质性。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-17 DOI: 10.1136/jitc-2025-013018
Mara J T Nicolasen, Lucrezia Cde Gatti, Laia Gasull-Celades, Peter Brazda, Marta Botas, Daniel Zawal, Esmee J van Vliet, Astrid Cleven, Zsolt Sebestyén, Pierre A Robe, Dennis X Beringer, Jurgen Kuball

Background: The effectiveness of immunotherapies against glioblastoma (GB) remains limited. A major obstacle in advancing new strategies is the reliance on non-autologous systems, which do not accurately mimic the true extent of inter-patient heterogeneity in both immune responses and tumor susceptibility. This often leads to misleading conclusions about therapeutic efficacy and targetability.

Methods: In this study, we addressed this critical gap by employing a fully autologous model. We phenotypically characterized primary αβ and γδT cells from the peripheral blood and tumors of 40 brain tumor patients, including 36 with confirmed GB, and expanded and functionally assessed the autologous anti-GB reactivity in a subset of patients.

Results: Notably, only Vδ2+ and Vδ2- γδT cells, but not αβT cells, recognized autologous tumors. While Vδ2- γδT cells showed activity in a subset of patients, Vδ2+ γδTILs from all patients responded to autologous GB cells in the presence of pamidronate. In patients, a higher percentage of Vδ2+ γδTILs was associated with longer overall survival. However, the potency of Vδ2+ γδTILs varied markedly between individuals, highlighting substantial inter-patient heterogeneity in γδT cell-mediated tumor recognition. This variability was driven by differences in both immune cell-intrinsic features and tumor-intrinsic factors, including expression of BTN2A1 and especially BTN3A, the ligands of the Vδ2+ γδTCR. Functional assays revealed that anti-GB reactivity was further modulated by stimulatory and inhibitory co-receptors such as NKG2D, CD94, and TIGIT. Transcriptomic analysis linked Vδ2+ γδT cell reactivity to extracellular matrix (ECM) pathways and disrupting ECM components such as LAMA5 and TGFB1 enhanced T cell responses. Knockout of ITGA3, a LAMA5 receptor, increased BTN2A1 and BTN3A expression on GB cells, improving immune recognition.

Conclusions: This study demonstrates that inter-patient heterogeneity in Vδ2+ γδTIL responses to GB is driven by the extracellular matrix-BTN3A axis. Autologous systems effectively capture this heterogeneity, offering a reliable platform to identify determinants of both immune function and tumor vulnerability, insights that are essential for the rational design of γδTIL-based immunotherapies.

背景:免疫疗法治疗胶质母细胞瘤(GB)的有效性仍然有限。推进新策略的一个主要障碍是对非自体系统的依赖,它不能准确地模拟免疫反应和肿瘤易感性中患者间异质性的真实程度。这常常导致关于治疗效果和靶向性的误导性结论。方法:在本研究中,我们通过采用完全自体模型来解决这一关键差距。我们对40例脑肿瘤患者的外周血和肿瘤中的αβ和γδT细胞进行了表型表征,其中包括36例确诊的GB,并对一部分患者的自体抗GB反应性进行了扩展和功能评估。结果:值得注意的是,只有Vδ2+和Vδ2- γδT细胞能识别自体肿瘤,而αβT细胞不能识别。虽然Vδ2- γδT细胞在一部分患者中表现出活性,但所有患者的Vδ2+ γδTILs在帕米膦酸盐存在下对自体GB细胞有反应。在患者中,较高比例的Vδ2+ γδTILs与较长的总生存期相关。然而,Vδ2+ γδTILs的效力在个体之间存在显著差异,这突出了γδT细胞介导的肿瘤识别在患者之间存在很大的异质性。这种变异是由免疫细胞内在特征和肿瘤内在因素的差异驱动的,包括BTN2A1,尤其是BTN3A的表达,BTN2A1是Vδ2+ γδTCR的配体。功能分析显示,NKG2D、CD94和TIGIT等刺激和抑制性共受体可进一步调节抗gb反应性。转录组学分析将Vδ2+ γδT细胞反应性与细胞外基质(ECM)途径联系起来,破坏ECM成分(如LAMA5和TGFB1)可增强T细胞反应。敲除LAMA5受体ITGA3可增加GB细胞BTN2A1和BTN3A的表达,提高免疫识别能力。结论:本研究表明,Vδ2+ γδTIL对GB反应的患者间异质性是由细胞外基质- btn3a轴驱动的。自体系统有效地捕获了这种异质性,为确定免疫功能和肿瘤易感性的决定因素提供了可靠的平台,这对于合理设计基于γδ til的免疫疗法至关重要。
{"title":"Autologous profiling reveals inter-patient heterogeneity in Vδ2<sup>+</sup>γδTIL responses to glioblastoma driven by extracellular matrix-BTN3A axis.","authors":"Mara J T Nicolasen, Lucrezia Cde Gatti, Laia Gasull-Celades, Peter Brazda, Marta Botas, Daniel Zawal, Esmee J van Vliet, Astrid Cleven, Zsolt Sebestyén, Pierre A Robe, Dennis X Beringer, Jurgen Kuball","doi":"10.1136/jitc-2025-013018","DOIUrl":"10.1136/jitc-2025-013018","url":null,"abstract":"<p><strong>Background: </strong>The effectiveness of immunotherapies against glioblastoma (GB) remains limited. A major obstacle in advancing new strategies is the reliance on non-autologous systems, which do not accurately mimic the true extent of inter-patient heterogeneity in both immune responses and tumor susceptibility. This often leads to misleading conclusions about therapeutic efficacy and targetability.</p><p><strong>Methods: </strong>In this study, we addressed this critical gap by employing a fully autologous model. We phenotypically characterized primary αβ and γδT cells from the peripheral blood and tumors of 40 brain tumor patients, including 36 with confirmed GB, and expanded and functionally assessed the autologous anti-GB reactivity in a subset of patients.</p><p><strong>Results: </strong>Notably, only Vδ2<sup>+</sup> and Vδ2<sup>-</sup> γδT cells, but not αβT cells, recognized autologous tumors. While Vδ2<sup>-</sup> γδT cells showed activity in a subset of patients, Vδ2<sup>+</sup> γδTILs from all patients responded to autologous GB cells in the presence of pamidronate. In patients, a higher percentage of Vδ2<sup>+</sup> γδTILs was associated with longer overall survival. However, the potency of Vδ2<sup>+</sup> γδTILs varied markedly between individuals, highlighting substantial inter-patient heterogeneity in γδT cell-mediated tumor recognition. This variability was driven by differences in both immune cell-intrinsic features and tumor-intrinsic factors, including expression of BTN2A1 and especially BTN3A, the ligands of the Vδ2<sup>+</sup> γδTCR. Functional assays revealed that anti-GB reactivity was further modulated by stimulatory and inhibitory co-receptors such as NKG2D, CD94, and TIGIT. Transcriptomic analysis linked Vδ2<sup>+</sup> γδT cell reactivity to extracellular matrix (ECM) pathways and disrupting ECM components such as LAMA5 and TGFB1 enhanced T cell responses. Knockout of ITGA3, a LAMA5 receptor, increased BTN2A1 and BTN3A expression on GB cells, improving immune recognition.</p><p><strong>Conclusions: </strong>This study demonstrates that inter-patient heterogeneity in Vδ2<sup>+</sup> γδTIL responses to GB is driven by the extracellular matrix-BTN3A axis. Autologous systems effectively capture this heterogeneity, offering a reliable platform to identify determinants of both immune function and tumor vulnerability, insights that are essential for the rational design of γδTIL-based immunotherapies.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12718601/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145774653","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
Selective expansion of T-cell receptor engineered T cells with increased stem-like phenotypes using neoantigen stimulation. 使用新抗原刺激选择性扩增T细胞受体工程T细胞,增加干细胞样表型。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-17 DOI: 10.1136/jitc-2025-013373
Sanghyun P Kim, Noam Levin, Youngseo Jo, Charles Marquardt, Zhiya Yu, Lior Levy, Nolan R Vale, Maria Parkhust, Satyajit Ray, Melinda Magna, Shahram Farid, Mamduh Khateb, Hyunmi Halas, Stephanie L Goff, Nicholas D Klemen, Steven A Rosenberg

Background: Adoptive transfer of T-cell receptor-engineered T cells (TCR-T cells) has shown promising efficacy in solid tumor treatment, but achieving clinical benefit typically requires infusion of tens of billions of cells. The commonly used rapid expansion protocol (REP), based on the CD3-agonistic OKT3 antibody and irradiated allogeneic feeder cells, exponentially expands tumor-infiltrating lymphocytes (TILs). However, the effect of REP on TCR-T cell frequency and phenotype remains unclear. This study aimed to evaluate the impact of REP on TCR-T cells and to assess the potential of a neoantigen-specific stimulation platform, NeoExpand, as a strategy to selectively expand TCR-T cells with favorable phenotypes.

Methods: We compared the effects of REP and NeoExpand on the frequency, yield, and phenotype of TCR-T cells engineered against shared neoantigens. Various autologous antigen-presenting cell (APC) types, including dendritic cells and bulk peripheral blood mononuclear cells (PBMCs), were tested as stimulators in NeoExpand. Additionally, we combined NeoExpand with CD3 activation, with or without allogeneic feeders, to improve scalability. The role of exogenous interleukin (IL)-21 in shaping TCR-T cell phenotypes was also investigated.

Results: REP impaired the frequency and phenotype of TCR-T cells, particularly CD4+ and CD8+ subsets matched to their engineered TCRs. In contrast, NeoExpand selectively increased TCR-T cell frequency and improved their phenotypes but did not consistently achieve higher total yields compared with REP. Among the tested autologous APCs, PBMCs effectively supported selective expansion. Further optimization using PBMCs as APCs revealed that combining NeoExpand with CD3 activation enabled exponential expansion without compromising selectivity. Inclusion of IL-21 during NeoExpand promoted a naïve/stem-like phenotype in CD8+ TCR-T cells, with minimal effect on CD4+ TCR-T cells.

Conclusions: These findings demonstrate that NeoExpand enables selective expansion of TCR-T cells while preserving favorable phenotypes and scalability. The approach supports further clinical evaluation of NeoExpand as a strategy to generate high-quality TCR-T cell products for adoptive cell therapy.

背景:T细胞受体工程T细胞(TCR-T细胞)过继转移在实体瘤治疗中显示出良好的疗效,但要达到临床效果通常需要注入数百亿细胞。常用的快速扩增方案(REP),基于cd3激动性OKT3抗体和辐照的异体饲养细胞,呈指数级扩增肿瘤浸润淋巴细胞(til)。然而,REP对TCR-T细胞频率和表型的影响尚不清楚。本研究旨在评估REP对TCR-T细胞的影响,并评估新抗原特异性刺激平台NeoExpand作为选择性扩增具有有利表型的TCR-T细胞的策略的潜力。方法:我们比较了REP和NeoExpand对共同新抗原介导的TCR-T细胞的频率、产量和表型的影响。各种自体抗原呈递细胞(APC)类型,包括树突状细胞和散装外周血单个核细胞(PBMCs),在NeoExpand中作为刺激物进行了测试。此外,我们将NeoExpand与CD3激活相结合,无论是否使用同种异体喂料,都可以提高可扩展性。外源性白细胞介素(IL)-21在形成TCR-T细胞表型中的作用也进行了研究。结果:REP损害了TCR-T细胞的频率和表型,特别是与它们的工程tcr匹配的CD4+和CD8+亚群。相比之下,NeoExpand选择性地增加了TCR-T细胞的频率并改善了它们的表型,但与rep相比,并不能始终保持更高的总产量。使用pbmc作为apc进一步优化表明,将NeoExpand与CD3激活结合可以在不影响选择性的情况下实现指数扩展。在NeoExpand期间,IL-21在CD8+ TCR-T细胞中促进naïve/干细胞样表型,对CD4+ TCR-T细胞的影响最小。结论:这些发现表明NeoExpand能够选择性扩增TCR-T细胞,同时保持有利的表型和可扩展性。该方法支持NeoExpand作为一种策略的进一步临床评估,以产生用于过继细胞治疗的高质量TCR-T细胞产品。
{"title":"Selective expansion of T-cell receptor engineered T cells with increased stem-like phenotypes using neoantigen stimulation.","authors":"Sanghyun P Kim, Noam Levin, Youngseo Jo, Charles Marquardt, Zhiya Yu, Lior Levy, Nolan R Vale, Maria Parkhust, Satyajit Ray, Melinda Magna, Shahram Farid, Mamduh Khateb, Hyunmi Halas, Stephanie L Goff, Nicholas D Klemen, Steven A Rosenberg","doi":"10.1136/jitc-2025-013373","DOIUrl":"10.1136/jitc-2025-013373","url":null,"abstract":"<p><strong>Background: </strong>Adoptive transfer of T-cell receptor-engineered T cells (TCR-T cells) has shown promising efficacy in solid tumor treatment, but achieving clinical benefit typically requires infusion of tens of billions of cells. The commonly used rapid expansion protocol (REP), based on the CD3-agonistic OKT3 antibody and irradiated allogeneic feeder cells, exponentially expands tumor-infiltrating lymphocytes (TILs). However, the effect of REP on TCR-T cell frequency and phenotype remains unclear. This study aimed to evaluate the impact of REP on TCR-T cells and to assess the potential of a neoantigen-specific stimulation platform, NeoExpand, as a strategy to selectively expand TCR-T cells with favorable phenotypes.</p><p><strong>Methods: </strong>We compared the effects of REP and NeoExpand on the frequency, yield, and phenotype of TCR-T cells engineered against shared neoantigens. Various autologous antigen-presenting cell (APC) types, including dendritic cells and bulk peripheral blood mononuclear cells (PBMCs), were tested as stimulators in NeoExpand. Additionally, we combined NeoExpand with CD3 activation, with or without allogeneic feeders, to improve scalability. The role of exogenous interleukin (IL)-21 in shaping TCR-T cell phenotypes was also investigated.</p><p><strong>Results: </strong>REP impaired the frequency and phenotype of TCR-T cells, particularly CD4<sup>+</sup> and CD8<sup>+</sup> subsets matched to their engineered TCRs. In contrast, NeoExpand selectively increased TCR-T cell frequency and improved their phenotypes but did not consistently achieve higher total yields compared with REP. Among the tested autologous APCs, PBMCs effectively supported selective expansion. Further optimization using PBMCs as APCs revealed that combining NeoExpand with CD3 activation enabled exponential expansion without compromising selectivity. Inclusion of IL-21 during NeoExpand promoted a naïve/stem-like phenotype in CD8<sup>+</sup> TCR-T cells, with minimal effect on CD4<sup>+</sup> TCR-T cells.</p><p><strong>Conclusions: </strong>These findings demonstrate that NeoExpand enables selective expansion of TCR-T cells while preserving favorable phenotypes and scalability. The approach supports further clinical evaluation of NeoExpand as a strategy to generate high-quality TCR-T cell products for adoptive cell therapy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12716566/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145774587","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
TIM-3 blockade enhances ex vivo stimulated allogeneic NK cell therapy for relapsed murine neuroblastoma after hematopoietic cell transplant. TIM-3阻断增强体外刺激异体NK细胞治疗小鼠造血细胞移植后复发的神经母细胞瘤。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-12 DOI: 10.1136/jitc-2024-010239
Aicha E Quamine, Evan L Dray, Nicholas R Mohrdieck, Chloe A King, Anastasia A Griggs, Jillian M Kline, Monica M Cho, Sean P Rinella, Katharine E Tippins, Paul D Bates, Lei Shi, Longzhen Song, Nicholas J Hess, Tyce J Kearl, Bryon D Johnson, Christian M Capitini

Background: High-risk neuroblastoma (HR-NBL) is an aggressive tumor of the sympathetic nervous system with high risk of relapse and poor overall survival. Allogeneic hematopoietic cell transplant (allo-HCT) has been used previously in patients with HR-NBL; however, graft-versus-host disease (GVHD) and disease progression have limited clinical application. Ex vivo stimulated allogeneic natural killer (NK) cells represent an approach to enhance the graft-versus-tumor (GVT) effect without exacerbation of GVHD but have not shown efficacy in NBL.

Methods: Ex vivo stimulated NK cells from C57BL/6NCr (B6) mice were expanded with soluble interleukin-15 (IL-15) and IL-15 receptor alpha (IL-15Rα) alone or with irradiated CD137L/CD54+ aggressive variant of the Neuro-2a murine neuroblastoma cell line (15-4P) at a 1:1 ratio for 10-12 days. Allogeneic NK cells were then analyzed for activation, proliferation, cytokine production, and cytotoxicity against two murine NBL cell lines, Neuro2a and NXS2, in the absence or presence of anti-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3). Lethally irradiated B6AJF1 mice received allo-HCT from B6 donors followed by NBL challenge after 7 days to mimic tumor relapse. Select groups received anti-TIM-3 starting on day 9 for every 4 days with/without infusions of 15-4P B6 NK cells on days 14, 21, and 28. In select experiments, T cell and NK cells were selectively depleted to establish contribution to the GVT effect. All groups were analyzed for tumor growth, GVHD and survival.

Results: Co-culturing NK cells with 15-4P results in 78-fold expansion with increased expression of Kiel-67 (Ki-67) and Natural Killer Group 2, Member D (NKG2D), NKp46, TNF-Related Apoptosis-Inducing Ligand (TRAIL) and TIM-3. 15-4P stimulated allogeneic NK cells showed enhanced cytotoxicity against NBL compared with IL-15 NK cells alone but was limited in part due to high expression of TIM-3 ligands on Neuro-2a compared with NXS2. The addition of TIM-3 blockade further enhanced NK cytotoxicity versus Neuro-2a, with enhanced 15-4P NK cell degranulation, Eomesodermin, TRAIL and Fas Ligand expression observed. In vivo, the combination of 15-4P stimulated allogeneic NK cells and TIM-3 blockade after allo-HCT resulted in prolonged survival against NBL with decreased tumor burden compared with NK cells or anti-TIM-3 alone. Depletion of NK cells, but not T cells, abrogated the GVT effect.

Conclusion: Allo-HCT can be a platform for treating NBL using combination ex vivo stimulated allogeneic NK cell therapy with TIM-3 blockade to enhance the GVT effect without inducing GVHD.

背景:高危神经母细胞瘤(HR-NBL)是一种交感神经系统侵袭性肿瘤,复发风险高,总生存率差。同种异体造血细胞移植(alloo - hct)以前曾用于HR-NBL患者;然而,移植物抗宿主病(GVHD)和疾病进展的临床应用有限。体外刺激的同种异体自然杀伤(NK)细胞是一种增强移植物抗肿瘤(GVT)效果而不加重GVHD的方法,但在NBL中尚未显示出疗效。方法:用可溶性白细胞介素-15 (IL-15)和IL-15受体α (IL-15Rα)单独或用CD137L/CD54+侵袭性变异的neuro2a小鼠神经母细胞瘤细胞系(15-4P)按1:1的比例扩增体外刺激的C57BL/6NCr (B6)小鼠NK细胞10-12天。然后分析同种异体NK细胞在缺乏或存在抗t细胞免疫球蛋白和粘蛋白结构域-3 (TIM-3)的情况下对两种小鼠NBL细胞系Neuro2a和NXS2的活化、增殖、细胞因子产生和细胞毒性。致命照射后的B6AJF1小鼠接受来自B6供体的同种异体hct,并在7天后接受NBL攻击以模拟肿瘤复发。选择组从第9天开始注射抗tim -3,每4天注射1次,第14、21、28天注射15-4P B6 NK细胞。在选定的实验中,T细胞和NK细胞被选择性地耗尽,以确定对GVT效应的贡献。分析各组肿瘤生长、GVHD及生存情况。结果:NK细胞与15-4P共培养,增殖78倍,Kiel-67 (Ki-67)和自然杀伤组2、成员D (NKG2D)、NKp46、tnf相关凋亡诱导配体(TRAIL)和TIM-3的表达增加。与单独使用IL-15 NK细胞相比,15-4P刺激的同种异体NK细胞对NBL的细胞毒性增强,但部分原因是与NXS2相比,神经2a上TIM-3配体的高表达。TIM-3阻断剂的加入进一步增强了NK细胞对神经-2a的毒性,观察到15-4P NK细胞脱粒、Eomesodermin、TRAIL和Fas配体表达增强。在体内,与单独使用NK细胞或抗TIM-3相比,15-4P刺激的同种异体NK细胞和TIM-3阻断联合使用可延长抗NBL的生存时间,并降低肿瘤负荷。NK细胞的消耗,而不是T细胞,消除了GVT的作用。结论:Allo-HCT可作为治疗NBL的平台,体外刺激的同种异体NK细胞联合TIM-3阻断治疗可增强GVT效果,但不诱导GVHD。
{"title":"TIM-3 blockade enhances ex vivo stimulated allogeneic NK cell therapy for relapsed murine neuroblastoma after hematopoietic cell transplant.","authors":"Aicha E Quamine, Evan L Dray, Nicholas R Mohrdieck, Chloe A King, Anastasia A Griggs, Jillian M Kline, Monica M Cho, Sean P Rinella, Katharine E Tippins, Paul D Bates, Lei Shi, Longzhen Song, Nicholas J Hess, Tyce J Kearl, Bryon D Johnson, Christian M Capitini","doi":"10.1136/jitc-2024-010239","DOIUrl":"10.1136/jitc-2024-010239","url":null,"abstract":"<p><strong>Background: </strong>High-risk neuroblastoma (HR-NBL) is an aggressive tumor of the sympathetic nervous system with high risk of relapse and poor overall survival. Allogeneic hematopoietic cell transplant (allo-HCT) has been used previously in patients with HR-NBL; however, graft-versus-host disease (GVHD) and disease progression have limited clinical application. Ex vivo stimulated allogeneic natural killer (NK) cells represent an approach to enhance the graft-versus-tumor (GVT) effect without exacerbation of GVHD but have not shown efficacy in NBL.</p><p><strong>Methods: </strong>Ex vivo stimulated NK cells from C57BL/6NCr (B6) mice were expanded with soluble interleukin-15 (IL-15) and IL-15 receptor alpha (IL-15Rα) alone or with irradiated CD137L/CD54<sup>+</sup> aggressive variant of the Neuro-2a murine neuroblastoma cell line (15-4P) at a 1:1 ratio for 10-12 days. Allogeneic NK cells were then analyzed for activation, proliferation, cytokine production, and cytotoxicity against two murine NBL cell lines, Neuro2a and NXS2, in the absence or presence of anti-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3). Lethally irradiated B6AJF1 mice received allo-HCT from B6 donors followed by NBL challenge after 7 days to mimic tumor relapse. Select groups received anti-TIM-3 starting on day 9 for every 4 days with/without infusions of 15-4P B6 NK cells on days 14, 21, and 28. In select experiments, T cell and NK cells were selectively depleted to establish contribution to the GVT effect. All groups were analyzed for tumor growth, GVHD and survival.</p><p><strong>Results: </strong>Co-culturing NK cells with 15-4P results in 78-fold expansion with increased expression of Kiel-67 (Ki-67) and Natural Killer Group 2, Member D (NKG2D), NKp46, TNF-Related Apoptosis-Inducing Ligand (TRAIL) and TIM-3. 15-4P stimulated allogeneic NK cells showed enhanced cytotoxicity against NBL compared with IL-15 NK cells alone but was limited in part due to high expression of TIM-3 ligands on Neuro-2a compared with NXS2. The addition of TIM-3 blockade further enhanced NK cytotoxicity versus Neuro-2a, with enhanced 15-4P NK cell degranulation, Eomesodermin, TRAIL and Fas Ligand expression observed. In vivo, the combination of 15-4P stimulated allogeneic NK cells and TIM-3 blockade after allo-HCT resulted in prolonged survival against NBL with decreased tumor burden compared with NK cells or anti-TIM-3 alone. Depletion of NK cells, but not T cells, abrogated the GVT effect.</p><p><strong>Conclusion: </strong>Allo-HCT can be a platform for treating NBL using combination ex vivo stimulated allogeneic NK cell therapy with TIM-3 blockade to enhance the GVT effect without inducing GVHD.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12699621/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742228","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
Aberrant lipid metabolism reshapes the immune landscape in bone metastasis of nasopharyngeal carcinoma. 异常脂质代谢重塑鼻咽癌骨转移的免疫景观。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-12 DOI: 10.1136/jitc-2025-012134
Yuanyuan Feng, Hongmei Wang, Ziyan Zhu, Wenxiang Deng, Xiaoyue He, Yingying Yao, Miao Song, Xiaohong Peng, Yanling Lin, Longmei Cai

Background: Bone metastasis (BM) drives therapeutic resistance and mortality in nasopharyngeal carcinoma (NPC). Tumor metabolites are crucial for NPC metastasis; however, the mechanisms by which these metabolites synergistically alter the immune microenvironment to promote BM remain unclear.

Methods: In this study, limited immune infiltration was observed in the NPC BM tumor microenvironment. Multiomics analysis has identified sphingosine kinase 1 (SPHK1) as a pivotal mediator driving BM and immune evasion in NPC, orchestrating the production of 1-phosphorylated sphingosine (S1P), which is critical for NPC pathogenesis.

Results: The aberrant buildup of lipid metabolites, along with immune microenvironment shifts, serves as a critical driver of NPC BM. Mechanistically, S1P enhanced osteoclast recruitment via S1PR3 binding and activated the Hippo pathway, worsening bone colonization and facilitating immune evasion by expanding the exhausted CD8+ T cell population.

Conclusions: The synergy between the SPHK1 inhibitor PF543 and anti-programmed cell death protein 1 therapy amplified treatment effectiveness beyond standalone approaches. Overall, the SPHK1/S1P pathway advances NPC growth and aids in suppressing immune defense. Regulation of lipid metabolism may be a therapeutic target against BM in NPC and may improve the effectiveness of immunotherapy.

背景:骨转移(BM)导致鼻咽癌(NPC)的治疗抵抗和死亡率。肿瘤代谢物对鼻咽癌转移至关重要;然而,这些代谢物协同改变免疫微环境以促进脑转移的机制尚不清楚。方法:在本研究中,在NPC BM肿瘤微环境中观察到有限的免疫浸润。多组学分析发现,鞘氨醇激酶1 (SPHK1)是鼻咽癌中驱动BM和免疫逃避的关键介质,协调1-磷酸化鞘氨醇(S1P)的产生,这对鼻咽癌的发病至关重要。结果:脂质代谢物的异常积累,以及免疫微环境的改变,是鼻咽癌BM的关键驱动因素。机制上,S1P通过S1PR3结合增强破骨细胞募集,激活Hippo通路,通过扩大耗尽的CD8+ T细胞群,恶化骨定植并促进免疫逃避。结论:SPHK1抑制剂PF543与抗程序性细胞死亡蛋白1疗法之间的协同作用提高了治疗效果。总的来说,SPHK1/S1P通路促进NPC生长并有助于抑制免疫防御。调节脂质代谢可能是鼻咽癌治疗BM的靶点,并可能提高免疫治疗的有效性。
{"title":"Aberrant lipid metabolism reshapes the immune landscape in bone metastasis of nasopharyngeal carcinoma.","authors":"Yuanyuan Feng, Hongmei Wang, Ziyan Zhu, Wenxiang Deng, Xiaoyue He, Yingying Yao, Miao Song, Xiaohong Peng, Yanling Lin, Longmei Cai","doi":"10.1136/jitc-2025-012134","DOIUrl":"10.1136/jitc-2025-012134","url":null,"abstract":"<p><strong>Background: </strong>Bone metastasis (BM) drives therapeutic resistance and mortality in nasopharyngeal carcinoma (NPC). Tumor metabolites are crucial for NPC metastasis; however, the mechanisms by which these metabolites synergistically alter the immune microenvironment to promote BM remain unclear.</p><p><strong>Methods: </strong>In this study, limited immune infiltration was observed in the NPC BM tumor microenvironment. Multiomics analysis has identified sphingosine kinase 1 (SPHK1) as a pivotal mediator driving BM and immune evasion in NPC, orchestrating the production of 1-phosphorylated sphingosine (S1P), which is critical for NPC pathogenesis.</p><p><strong>Results: </strong>The aberrant buildup of lipid metabolites, along with immune microenvironment shifts, serves as a critical driver of NPC BM. Mechanistically, S1P enhanced osteoclast recruitment via S1PR3 binding and activated the Hippo pathway, worsening bone colonization and facilitating immune evasion by expanding the exhausted CD8<sup>+</sup> T cell population.</p><p><strong>Conclusions: </strong>The synergy between the SPHK1 inhibitor PF543 and anti-programmed cell death protein 1 therapy amplified treatment effectiveness beyond standalone approaches. Overall, the SPHK1/S1P pathway advances NPC growth and aids in suppressing immune defense. Regulation of lipid metabolism may be a therapeutic target against BM in NPC and may improve the effectiveness of immunotherapy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12699725/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742857","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
In situ DC-primed vaccine combined with CD137 agonist elicits long-lasting antitumor immunity through cDC1-mediated tumor antigen-specific CD8+ T cell responses. 原位dc引物疫苗联合CD137激动剂可通过cdc1介导的肿瘤抗原特异性CD8+ T细胞反应获得持久的抗肿瘤免疫。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-12 DOI: 10.1136/jitc-2025-011665
Mengjiao Wang, Li Qiu, Cheng Tang, Danqing Huang, Qiaocong Zheng, Zunyi Li, Zhenhui Zhang, Shupei Wei, Lanfang Chen, Hongxin Huang, Jun Liu, Junfeng Zhou, Tao Lu, Guanxin Liu, Yingjie Nie, Qibin Leng, Tao Chen

Background: Intratumoral vaccines offer a promising avenue in cancer immunotherapy by harnessing the tumor microenvironment to stimulate immune responses. However, challenges persist in maximizing their effectiveness and addressing immune suppression within tumors.

Methods: Conventional in situ vaccine (CisVac) and α-CD137 antibody were administered in implanted mouse models of lung and colon cancer to evaluate therapeutic efficacy. The initiation mechanism of CD8+ T cells was determined using antibody blockade and transgenic mouse models. Single-cell RNA sequencing was used to further characterize the activation mechanism of T cells.

Results: Our findings indicate that this synergistic approach markedly inhibits tumor growth while eliciting a robust antitumor immune response, characterized by heightened activation and cytotoxic differentiation of CD8+ T cells, as well as the polarization of conventional dendritic cells (cDC1). Mechanistically, it promotes the cDC1-dependent proportion and cytokine production of tumor antigen-specific CD8+ T cells, mobilizes and establishes enduring systemic immune memory. Our single-cell transcriptome analyses revealed that the therapy facilitates a functional remodeling of regulatory T cells (Tregs) with upregulated inflammatory genes, potentially attenuating immune suppression. Cell-cell communication analyses highlighted interactions between CD4+ Th1-like/Th17 cells and monocytes/DCs through the CD40L-CD40 pathway, indicating potential intercellular regulatory mechanisms.

Conclusions: Our results highlight the transformative potential of the CisVac/α-CD137 combination in cancer immunotherapy, paving the way for further exploration of its clinical utility and long-term efficacy.

背景:肿瘤内疫苗通过利用肿瘤微环境刺激免疫反应,为癌症免疫治疗提供了一条很有前途的途径。然而,在最大限度地提高其有效性和解决肿瘤内的免疫抑制问题方面,挑战仍然存在。方法:采用常规原位疫苗(CisVac)和α-CD137抗体对小鼠肺癌和结肠癌移植模型进行治疗,观察其疗效。利用抗体阻断和转基因小鼠模型确定CD8+ T细胞的起始机制。单细胞RNA测序用于进一步表征T细胞的激活机制。结果:我们的研究结果表明,这种协同方法显著抑制肿瘤生长,同时引发强大的抗肿瘤免疫反应,其特征是CD8+ T细胞的激活和细胞毒性分化增强,以及常规树突状细胞(cDC1)的极化。在机制上,它促进肿瘤抗原特异性CD8+ T细胞的cdc1依赖性比例和细胞因子的产生,动员并建立持久的全身免疫记忆。我们的单细胞转录组分析显示,该疗法促进了炎症基因上调的调节性T细胞(Tregs)的功能重塑,潜在地减轻了免疫抑制。细胞间通讯分析强调了CD4+ th1样细胞/Th17细胞通过CD40L-CD40途径与单核细胞/ dc之间的相互作用,表明了潜在的细胞间调节机制。结论:我们的研究结果突出了CisVac/α-CD137联合治疗在癌症免疫治疗中的变革潜力,为进一步探索其临床应用和长期疗效铺平了道路。
{"title":"In situ DC-primed vaccine combined with CD137 agonist elicits long-lasting antitumor immunity through cDC1-mediated tumor antigen-specific CD8<sup>+</sup> T cell responses.","authors":"Mengjiao Wang, Li Qiu, Cheng Tang, Danqing Huang, Qiaocong Zheng, Zunyi Li, Zhenhui Zhang, Shupei Wei, Lanfang Chen, Hongxin Huang, Jun Liu, Junfeng Zhou, Tao Lu, Guanxin Liu, Yingjie Nie, Qibin Leng, Tao Chen","doi":"10.1136/jitc-2025-011665","DOIUrl":"10.1136/jitc-2025-011665","url":null,"abstract":"<p><strong>Background: </strong>Intratumoral vaccines offer a promising avenue in cancer immunotherapy by harnessing the tumor microenvironment to stimulate immune responses. However, challenges persist in maximizing their effectiveness and addressing immune suppression within tumors.</p><p><strong>Methods: </strong>Conventional in situ vaccine (CisVac) and α-CD137 antibody were administered in implanted mouse models of lung and colon cancer to evaluate therapeutic efficacy. The initiation mechanism of CD8<sup>+</sup> T cells was determined using antibody blockade and transgenic mouse models. Single-cell RNA sequencing was used to further characterize the activation mechanism of T cells.</p><p><strong>Results: </strong>Our findings indicate that this synergistic approach markedly inhibits tumor growth while eliciting a robust antitumor immune response, characterized by heightened activation and cytotoxic differentiation of CD8<sup>+</sup> T cells, as well as the polarization of conventional dendritic cells (cDC1). Mechanistically, it promotes the cDC1-dependent proportion and cytokine production of tumor antigen-specific CD8<sup>+</sup> T cells, mobilizes and establishes enduring systemic immune memory. Our single-cell transcriptome analyses revealed that the therapy facilitates a functional remodeling of regulatory T cells (Tregs) with upregulated inflammatory genes, potentially attenuating immune suppression. Cell-cell communication analyses highlighted interactions between CD4<sup>+</sup> Th1-like/Th17 cells and monocytes/DCs through the CD40L-CD40 pathway, indicating potential intercellular regulatory mechanisms.</p><p><strong>Conclusions: </strong>Our results highlight the transformative potential of the CisVac/α-CD137 combination in cancer immunotherapy, paving the way for further exploration of its clinical utility and long-term efficacy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12706255/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742941","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
Systemic STING agonist therapy drives expression of interferon stimulated genes and downstream production of cytokines in dogs with solid tumors. 系统性STING激动剂治疗驱动实体瘤犬干扰素刺激基因的表达和下游细胞因子的产生。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-11 DOI: 10.1136/jitc-2025-013715
Jennifer A Lenz, June DiBona, Matthew J Atherton, Sumita Roy-Ghanta, Hank Schmidt, Timothy Hart, Jong W Yu

Background: Stimulator of interferon genes (STING) agonist drugs can induce expression of interferon stimulated genes (ISGs) and proinflammatory cytokine production aimed to enhance antitumor immunity. The purpose of the current study was to determine the safety, pharmacokinetic, and systemic and intratumoral pharmacodynamic properties of a novel, intravenously delivered STING agonist in client-owned dogs with cancer.

Methods: GSK856, a small-molecule dimeric amidobenzimidazole STING agonist, was administered intravenously to dogs with naturally developing tumors. Patients received two doses of GSK856 1 week apart, followed by definitive-intent surgical tumor removal.

Results: 19 dogs diagnosed with various solid tumor types, including malignant melanoma (oral mucosa, n=9; digit, n=1; conjunctiva, n=1), soft tissue sarcoma (5), rhabdomyosarcoma (1), oral fibrosarcoma (1), and mammary squamous cell carcinoma (1), were enrolled. Systemic pharmacokinetic analysis revealed rapid plasma clearance of GSK856 within 30 min of bolus administration. Clinical adverse events of fever, lethargy, and nausea were transient. Concurrent elevation in serum cytokines, including interleukin-6, was consistent with cytokine release syndrome following activation of the STING pathway. Transcriptional analyses of pretreatment and post-treatment blood and tumor tissue revealed robust induction of ISGs.

Conclusions: These data identify tolerated dose levels for a novel, intravenously delivered STING agonist compound that results in on-target effects in systemic and intratumoral immune responses in dogs with solid tumors.

背景:干扰素基因刺激剂(STING)激动剂药物可以诱导干扰素刺激基因(ISGs)的表达和促炎细胞因子的产生,从而增强抗肿瘤免疫。本研究的目的是确定一种新型静脉给药STING激动剂对患有癌症的狗的安全性、药代动力学以及全身和肿瘤内药效学特性。方法:采用小分子二聚体氨基苯并咪唑STING激动剂GSK856,静脉给药于自然发生肿瘤的犬。患者间隔1周接受两剂GSK856,随后进行明确意向的手术肿瘤切除。结果:入选各类实体瘤犬19只,包括恶性黑色素瘤(口腔黏膜,n=9;手指,n=1;结膜,n=1)、软组织肉瘤(5)、横纹肌肉瘤(1)、口腔纤维肉瘤(1)、乳腺鳞癌(1)。系统药代动力学分析显示,GSK856在给药30分钟内快速血浆清除。发热、嗜睡、恶心等临床不良事件均为短暂性。血清细胞因子(包括白细胞介素-6)同时升高与STING通路激活后的细胞因子释放综合征一致。治疗前和治疗后血液和肿瘤组织的转录分析显示,isg具有强大的诱导作用。结论:这些数据确定了一种新型静脉给药STING激动剂化合物的耐受剂量水平,该化合物可在实体瘤犬的全身和肿瘤内免疫反应中产生靶效应。
{"title":"Systemic STING agonist therapy drives expression of interferon stimulated genes and downstream production of cytokines in dogs with solid tumors.","authors":"Jennifer A Lenz, June DiBona, Matthew J Atherton, Sumita Roy-Ghanta, Hank Schmidt, Timothy Hart, Jong W Yu","doi":"10.1136/jitc-2025-013715","DOIUrl":"10.1136/jitc-2025-013715","url":null,"abstract":"<p><strong>Background: </strong>Stimulator of interferon genes (STING) agonist drugs can induce expression of interferon stimulated genes (ISGs) and proinflammatory cytokine production aimed to enhance antitumor immunity. The purpose of the current study was to determine the safety, pharmacokinetic, and systemic and intratumoral pharmacodynamic properties of a novel, intravenously delivered STING agonist in client-owned dogs with cancer.</p><p><strong>Methods: </strong>GSK856, a small-molecule dimeric amidobenzimidazole STING agonist, was administered intravenously to dogs with naturally developing tumors. Patients received two doses of GSK856 1 week apart, followed by definitive-intent surgical tumor removal.</p><p><strong>Results: </strong>19 dogs diagnosed with various solid tumor types, including malignant melanoma (oral mucosa, n=9; digit, n=1; conjunctiva, n=1), soft tissue sarcoma (5), rhabdomyosarcoma (1), oral fibrosarcoma (1), and mammary squamous cell carcinoma (1), were enrolled. Systemic pharmacokinetic analysis revealed rapid plasma clearance of GSK856 within 30 min of bolus administration. Clinical adverse events of fever, lethargy, and nausea were transient. Concurrent elevation in serum cytokines, including interleukin-6, was consistent with cytokine release syndrome following activation of the STING pathway. Transcriptional analyses of pretreatment and post-treatment blood and tumor tissue revealed robust induction of ISGs.</p><p><strong>Conclusions: </strong>These data identify tolerated dose levels for a novel, intravenously delivered STING agonist compound that results in on-target effects in systemic and intratumoral immune responses in dogs with solid tumors.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12699562/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742920","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
Harnessing the innate immune system: a novel bispecific antibody targeting CD47 and CD24 for selective tumor clearance. 利用先天免疫系统:一种新的双特异性抗体靶向CD47和CD24选择性清除肿瘤。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-11 DOI: 10.1136/jitc-2025-013283
Lidi Nan, Liandi Chen, Weifeng Huang, Shaogang Peng, Chao Wang, Huayuan Liao, Yifei Wang, Ziyue Cui, Yanqing Lv, Xiaomu Wang, Yi Luo, Andy Tsun, Xiaoniu Miao, Juan Zhang

Background: Tumor-associated macrophages (TAMs) abundantly infiltrate tumors and possess potent antitumor capabilities. "Don't eat me" signals like CD47 allow tumors to evade macrophages and proliferate unchecked. CD47 is upregulated in many tumors and interacts with the SIRPα expressed on macrophages to restrict effector function. Similarly, CD24 interacts with the Siglec-10 on TAMs to inhibit engulfment. Despite their potential, there is still a lack of effective therapeutics targeting macrophages. Recent clinical trials targeting CD47 have demonstrated limited efficacy and significant side effects in solid tumors, primarily due to the expression of CD47 on healthy cells such as red blood cells (RBCs). We therefore developed novel anti-CD47 variable domain of heavy chain of heavy-chain antibodies (vHHs) with strong ligand-blocking activity while demonstrating minimal binding to RBCs and incorporated these vHHs to generate an anti-CD47/CD24 bispecific antibody that preserves Fc-effector function and achieves improved tumor targeting while maintaining the blockade of antiphagocytic signals elicited by both CD47 and CD24.

Methods: Yeast display was employed to generate vHHs targeting CD47 and fully human monoclonal antibodies against CD24, respectively. The antigen binding epitopes of the vHHs to CD47 were predicted using AlphaFold3. Bispecific antibodies were designed, constructed, and characterized in vitro. Antitumor efficacy was evaluated in a human immune cell reconstitution mouse model, while safety was evaluated using a humanized syngeneic mouse model. Furthermore, the underlying mechanisms and alterations in tumor microenvironment were explored ex vivo.

Results: VHHs targeting CD47 and a fully human antibody against CD24 were identified, all exhibiting potent ligand-blocking activity. The bispecific antibody BiAb-103C, engineered on a human IgG1 scaffold, had strong binding to CD47+CD24+ tumor cells and could effectively inhibit the CD47-SIRPα interaction. Fc-effector activity was observed towards CD24 (but not CD47) single-positive cells to promote phagocytosis and antibody-dependent cellular cytotoxicity of CD47+CD24+ tumor cells. In mice, antibody candidates demonstrated notable antitumor activity alongside favorable safety observations.

Conclusions: Our study presents the discovery of an anti-CD47/CD24 bispecific antibody that offers a promising therapeutic strategy to address the challenges associated with both the efficacy and safety of CD47-targeting agents, offers insight into macrophage-driven cancer immunotherapy, and could potentially provide a therapeutic option for patients non-responsive to immunotherapy.

背景:肿瘤相关巨噬细胞(tumor associated macrophages, tam)大量浸润肿瘤,具有很强的抗肿瘤能力。像CD47这样的“不要吃我”信号允许肿瘤逃避巨噬细胞,不受控制地增殖。CD47在许多肿瘤中上调,并与巨噬细胞上表达的SIRPα相互作用以限制效应功能。同样,CD24与tam上的siglece -10相互作用以抑制吞噬。尽管巨噬细胞具有潜力,但目前仍缺乏针对巨噬细胞的有效治疗方法。最近针对CD47的临床试验表明,在实体肿瘤中,CD47的疗效有限,副作用显著,这主要是由于CD47在健康细胞(如红细胞)上的表达。因此,我们开发了具有强配体阻断活性的重链重链抗体(vHHs)的新型抗CD47可变结构域,同时显示出与红细胞的最小结合,并将这些vHHs结合产生抗CD47/CD24双特异性抗体,该抗体保留了fc效应功能,并在保持对CD47和CD24引发的抗吞噬信号的阻断的同时实现了更好的肿瘤靶向性。方法:利用酵母展示技术分别生成靶向CD47的vhs和针对CD24的全人源单克隆抗体。利用AlphaFold3预测vhs与CD47的抗原结合表位。在体外设计、构建和表征双特异性抗体。在人免疫细胞重建小鼠模型中评估抗肿瘤效果,在人源化同源小鼠模型中评估安全性。此外,我们还在体外探讨了肿瘤微环境的潜在机制和变化。结果:鉴定出靶向CD47的vhs和针对CD24的全人抗体,均表现出有效的配体阻断活性。基于人IgG1支架的双特异性抗体BiAb-103C与CD47+CD24+肿瘤细胞有强结合,能有效抑制CD47- sirp α相互作用。我们观察到fc效应对CD24(而非CD47)单阳性细胞的活性,以促进CD47+CD24+肿瘤细胞的吞噬和抗体依赖性细胞毒性。在小鼠实验中,候选抗体显示出显著的抗肿瘤活性和良好的安全性。结论:我们的研究提出了一种抗cd47 /CD24双特异性抗体的发现,为解决cd47靶向药物的有效性和安全性相关的挑战提供了一种有希望的治疗策略,为巨噬细胞驱动的癌症免疫治疗提供了见解,并可能为免疫治疗无反应的患者提供治疗选择。
{"title":"Harnessing the innate immune system: a novel bispecific antibody targeting CD47 and CD24 for selective tumor clearance.","authors":"Lidi Nan, Liandi Chen, Weifeng Huang, Shaogang Peng, Chao Wang, Huayuan Liao, Yifei Wang, Ziyue Cui, Yanqing Lv, Xiaomu Wang, Yi Luo, Andy Tsun, Xiaoniu Miao, Juan Zhang","doi":"10.1136/jitc-2025-013283","DOIUrl":"10.1136/jitc-2025-013283","url":null,"abstract":"<p><strong>Background: </strong>Tumor-associated macrophages (TAMs) abundantly infiltrate tumors and possess potent antitumor capabilities. \"Don't eat me\" signals like CD47 allow tumors to evade macrophages and proliferate unchecked. CD47 is upregulated in many tumors and interacts with the SIRPα expressed on macrophages to restrict effector function. Similarly, CD24 interacts with the Siglec-10 on TAMs to inhibit engulfment. Despite their potential, there is still a lack of effective therapeutics targeting macrophages. Recent clinical trials targeting CD47 have demonstrated limited efficacy and significant side effects in solid tumors, primarily due to the expression of CD47 on healthy cells such as red blood cells (RBCs). We therefore developed novel anti-CD47 variable domain of heavy chain of heavy-chain antibodies (vHHs) with strong ligand-blocking activity while demonstrating minimal binding to RBCs and incorporated these vHHs to generate an anti-CD47/CD24 bispecific antibody that preserves Fc-effector function and achieves improved tumor targeting while maintaining the blockade of antiphagocytic signals elicited by both CD47 and CD24.</p><p><strong>Methods: </strong>Yeast display was employed to generate vHHs targeting CD47 and fully human monoclonal antibodies against CD24, respectively. The antigen binding epitopes of the vHHs to CD47 were predicted using AlphaFold3. Bispecific antibodies were designed, constructed, and characterized in vitro. Antitumor efficacy was evaluated in a human immune cell reconstitution mouse model, while safety was evaluated using a humanized syngeneic mouse model. Furthermore, the underlying mechanisms and alterations in tumor microenvironment were explored ex vivo.</p><p><strong>Results: </strong>VHHs targeting CD47 and a fully human antibody against CD24 were identified, all exhibiting potent ligand-blocking activity. The bispecific antibody BiAb-103C, engineered on a human IgG1 scaffold, had strong binding to CD47<sup>+</sup>CD24<sup>+</sup> tumor cells and could effectively inhibit the CD47-SIRPα interaction. Fc-effector activity was observed towards CD24 (but not CD47) single-positive cells to promote phagocytosis and antibody-dependent cellular cytotoxicity of CD47<sup>+</sup>CD24<sup>+</sup> tumor cells. In mice, antibody candidates demonstrated notable antitumor activity alongside favorable safety observations.</p><p><strong>Conclusions: </strong>Our study presents the discovery of an anti-CD47/CD24 bispecific antibody that offers a promising therapeutic strategy to address the challenges associated with both the efficacy and safety of CD47-targeting agents, offers insight into macrophage-driven cancer immunotherapy, and could potentially provide a therapeutic option for patients non-responsive to immunotherapy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12699560/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742902","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
Monitoring systemic immune responses to checkpoint inhibition in breast cancer reveals host responses and mechanisms of resistance. 监测乳腺癌对检查点抑制的全身免疫反应,揭示宿主反应和耐药机制。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-10 DOI: 10.1136/jitc-2025-012606
Ann Hanna, Xiaopeng Sun, Quanhu Sheng, Paula I Gonzalez-Ericsson, Elizabeth C Wescott, Brandie C Taylor, Jacey L Marshall, Susan R Opalenik, Abigail L Toren, Violeta Sanchez, Carly M Fielder, Melinda E Sanders, Justin M Balko

Background: Immune checkpoint inhibitors (ICI) have improved survival in various cancers, but their success in breast cancer, specifically triple-negative breast cancer, remains limited, benefiting less than 10% of patients.

Methods: We modeled ICI response in vivo to unravel the mechanisms underlying immunotherapy efficacy, identify mechanisms of resistance in non-responsive tumors, and ascertain the therapeutic benefits of different chemotherapeutic combinations with ICI in breast cancer. We investigated the impact of ICI as monotherapy and in combination with other therapeutics in mouse models of mammary cancer, which we found robustly suppressed primary tumor growth and extended survival.

Results: Interestingly, even within a single model, responses to ICI were highly variable. Resistance was not reliably retained by transplantation into syngeneic hosts, suggesting a role for systemic host immunity rather than tumor-autonomous mechanisms. Transcriptomic analysis of the primary tumor landscape by fine-needle aspiration revealed that upregulated cytotoxic T-cell response and inflammatory interferon signaling (both at baseline and post anti-programmed death-ligand 1 administration) corresponded to favorable response to ICI. Longitudinal analysis of the peripheral blood uncovered enhanced myeloid cell recruitment in resistant mice, prior to therapy initiation. Similar effects were observed through longitudinal assessment of peripheral blood in patients with ICI-treated human breast cancer. Blocking myeloid cell recruitment with navaraxin (CXCR1/2 inhibitor) improved ICI responses, further suppressing tumor growth and improving survival.

Conclusions: These findings provide insight into resistance mechanisms and suggest the potential for minimally invasive strategies (sampling of systemic immune cells from peripheral blood) to identify patients likely to respond to ICI. This approach may help inform de-escalation strategies to mitigate therapeutic toxicities and limit unnecessary treatments.

背景:免疫检查点抑制剂(ICI)提高了各种癌症的生存率,但它们在乳腺癌,特别是三阴性乳腺癌中的成功仍然有限,只有不到10%的患者受益。方法:我们在体内模拟了ICI反应,以揭示免疫治疗疗效的机制,确定无反应肿瘤的耐药机制,并确定不同化疗联合ICI治疗乳腺癌的治疗效果。我们在乳腺癌小鼠模型中研究了ICI作为单一疗法和与其他疗法联合使用的影响,我们发现它可以抑制原发肿瘤的生长并延长生存期。结果:有趣的是,即使在单一模型中,对ICI的反应也是高度可变的。通过移植到同基因宿主中,抗性不能可靠地保留,这表明这是系统性宿主免疫的作用,而不是肿瘤自主机制。通过细针穿刺对原发肿瘤的转录组学分析显示,细胞毒性t细胞反应和炎症干扰素信号的上调(在基线和抗程序性死亡配体1给药后)与ICI的有利反应相对应。在治疗开始前,外周血的纵向分析揭示了耐药小鼠骨髓细胞募集的增强。通过对ici治疗的人乳腺癌患者外周血的纵向评估,也观察到类似的效果。用navaraxin (CXCR1/2抑制剂)阻断髓细胞募集可改善ICI反应,进一步抑制肿瘤生长并提高生存率。结论:这些发现提供了对耐药机制的深入了解,并建议采用微创策略(从外周血中采集全身免疫细胞)来识别可能对ICI有反应的患者。这种方法可能有助于制定降低风险的策略,以减轻治疗毒性和限制不必要的治疗。
{"title":"Monitoring systemic immune responses to checkpoint inhibition in breast cancer reveals host responses and mechanisms of resistance.","authors":"Ann Hanna, Xiaopeng Sun, Quanhu Sheng, Paula I Gonzalez-Ericsson, Elizabeth C Wescott, Brandie C Taylor, Jacey L Marshall, Susan R Opalenik, Abigail L Toren, Violeta Sanchez, Carly M Fielder, Melinda E Sanders, Justin M Balko","doi":"10.1136/jitc-2025-012606","DOIUrl":"10.1136/jitc-2025-012606","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICI) have improved survival in various cancers, but their success in breast cancer, specifically triple-negative breast cancer, remains limited, benefiting less than 10% of patients.</p><p><strong>Methods: </strong>We modeled ICI response in vivo to unravel the mechanisms underlying immunotherapy efficacy, identify mechanisms of resistance in non-responsive tumors, and ascertain the therapeutic benefits of different chemotherapeutic combinations with ICI in breast cancer. We investigated the impact of ICI as monotherapy and in combination with other therapeutics in mouse models of mammary cancer, which we found robustly suppressed primary tumor growth and extended survival.</p><p><strong>Results: </strong>Interestingly, even within a single model, responses to ICI were highly variable. Resistance was not reliably retained by transplantation into syngeneic hosts, suggesting a role for systemic host immunity rather than tumor-autonomous mechanisms. Transcriptomic analysis of the primary tumor landscape by fine-needle aspiration revealed that upregulated cytotoxic T-cell response and inflammatory interferon signaling (both at baseline and post anti-programmed death-ligand 1 administration) corresponded to favorable response to ICI. Longitudinal analysis of the peripheral blood uncovered enhanced myeloid cell recruitment in resistant mice, prior to therapy initiation. Similar effects were observed through longitudinal assessment of peripheral blood in patients with ICI-treated human breast cancer. Blocking myeloid cell recruitment with navaraxin (CXCR1/2 inhibitor) improved ICI responses, further suppressing tumor growth and improving survival.</p><p><strong>Conclusions: </strong>These findings provide insight into resistance mechanisms and suggest the potential for minimally invasive strategies (sampling of systemic immune cells from peripheral blood) to identify patients likely to respond to ICI. This approach may help inform de-escalation strategies to mitigate therapeutic toxicities and limit unnecessary treatments.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12699608/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145723026","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