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.
{"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}
Pub Date : 2025-12-21DOI: 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.
{"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}
Pub Date : 2025-12-21DOI: 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.
{"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}
Pub Date : 2025-12-21DOI: 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.
{"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}
Pub Date : 2025-12-21DOI: 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}
Pub Date : 2025-12-21DOI: 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.
{"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}
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.
{"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}
Pub Date : 2025-12-21DOI: 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.
{"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}
Pub Date : 2025-12-21DOI: 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}
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.
{"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}