OncoimmunologyPub Date : 2025-12-01Epub Date: 2025-05-27DOI: 10.1080/2162402X.2025.2508050
Willemijn van der Wulp, Dennis F G Remst, Carli S Koster, Anne K Wouters, Maaike E Ressing, Janine Schuurman, Sander I van Kasteren, Boris Bleijlevens, Rob C Hoeben, Lars Guelen, Mirjam H M Heemskerk
{"title":"Increasing the odds: antibody-mediated delivery of two distinct immunogenic T-cell epitopes with one antibody.","authors":"Willemijn van der Wulp, Dennis F G Remst, Carli S Koster, Anne K Wouters, Maaike E Ressing, Janine Schuurman, Sander I van Kasteren, Boris Bleijlevens, Rob C Hoeben, Lars Guelen, Mirjam H M Heemskerk","doi":"10.1080/2162402X.2025.2508050","DOIUrl":"10.1080/2162402X.2025.2508050","url":null,"abstract":"<p><p>Antibody-epitope conjugates (AECs) proved to be a promising new therapeutic strategy to redirect virus-specific CD8<sup>+</sup> T cells toward cancer cells by delivering T-cell epitopes. To be able to redirect a larger fraction of the virus-specific T-cell population, it is beneficial to deliver a broader selection of T-cell epitopes. We investigated two different methods to generate AECs with two distinct virus-specific T-cell epitopes fused to one antibody. Epitopes were either placed in a tandem-like fashion at the C-terminus of the AEC (t-AEC) or bispecific-AECs (bs-AECs) were generated via controlled Fab-arm exchange to generate bs-AECs with two identical antigen binding domains, but two distinct epitopes on each Fab-arm. Our study revealed that maintaining a free epitope terminus was required for efficient delivery of the virus-specific T-cell epitopes. Consequently, viral-epitope delivery using t-AECs was suboptimal as the concatenated epitopes were less effectively delivered to the target cells. However, well-defined bs-AECs containing both CMV and EBV epitopes were successfully generated and both <i>in vitro</i> and <i>in vivo</i> efficacy was evaluated. Our results demonstrate that bispecific-AECs can efficiently deliver EBV and CMV epitopes simultaneously to multiple cancer cell lines from different origins, thereby redirecting and activating two distinct populations of virus-specific T cells. Furthermore, our <i>in vivo</i> findings indicate that when both virus-specific T-cell populations are present and tumor cells express the proteases required for efficient epitope delivery, bs-AECs exhibit similar efficacy in reducing tumor burden compared to AECs. To conclude, our study demonstrates the feasibility of redirecting two groups of virus-specific T cells using a single antibody and highlights the potential of bs-AECs both <i>in vitro</i> and <i>in vivo</i>.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2508050"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12118402/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144163150","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncoimmunologyPub Date : 2025-12-01Epub Date: 2025-04-15DOI: 10.1080/2162402X.2025.2492932
Fabrice Barlesi, Adrien Dixmier, Didier Debieuvre, Christophe Raspaud, Jean-Bernard Auliac, Nicolas Benoit, Pierre Bombaron, Denis Moro-Sibilot, Bernard Asselain, Clarisse Audigier-Valette, Florence Brellier, François-Emery Cotté, Yaacoub Khalife, Maurice Pérol
{"title":"Final 3-year results from the EVIDENS study, an observational study of nivolumab in non-small cell lung cancer.","authors":"Fabrice Barlesi, Adrien Dixmier, Didier Debieuvre, Christophe Raspaud, Jean-Bernard Auliac, Nicolas Benoit, Pierre Bombaron, Denis Moro-Sibilot, Bernard Asselain, Clarisse Audigier-Valette, Florence Brellier, François-Emery Cotté, Yaacoub Khalife, Maurice Pérol","doi":"10.1080/2162402X.2025.2492932","DOIUrl":"https://doi.org/10.1080/2162402X.2025.2492932","url":null,"abstract":"<p><p>EVIDENS was a prospective, non-interventional, longitudinal study conducted in non-small cell lung cancer (NSCLC) patients receiving nivolumab in France. It recruited adults with pathologically confirmed NSCLC who initiated nivolumab between October 2016 and November 2017; the final results are reported here. Primary outcomes included baseline characteristics and 36-month overall survival (OS). Secondary outcomes included progression free survival (PFS), objective response rate (ORR), safety and health-related quality of life (HRQoL; assessed regardless of nivolumab continuation or interruption). Overall, 1423 patients were included in the analysis population (median age 66 years; non-squamous histology 69.1%; stage IV disease 91.5%; brain metastases 19.9%). Almost all patients (99.7%) had received prior chemotherapy, and most patients received nivolumab as second-line (73.5%) or later (26.1%) therapy. The 36-month OS rate was 19.7% (95% confidence interval [CI] 17.5-22.0); OS was significantly shorter in patients with squamous versus non-squamous tumors (9.8 [95% CI 8.6-11.2] months vs 11.8 [95% CI 10.2-13.2] months; <i>p</i> = 0.005). The 36-month PFS rate was 8.8% (95% CI 7.3-10.4). The 12-month investigator-assessed best ORR in the overall population was 20.4%. Eastern Cooperative Oncology Group performance status, smoking status, tumor histology, disease stage and liver metastasis independently predicted survival. Grade 3 and 4 treatment-related adverse events were reported in 8.0% and 0.8% of patients, respectively; eight treatment-related deaths occurred (0.005%). HRQoL was maintained with slight improvement throughout the study, without statistical significance. These results confirm that the real-world effectiveness and safety of nivolumab in these patients is similar to that observed in clinical trials.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2492932"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12001549/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144053980","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Calpain 2 regulates IL-1α secretion and inhibits tumor development via modulating calpain 1 expression in the tumor microenvironment.","authors":"Zuhairah Binte Hanafi, Yu Mei, Huey Yee Teo, Ying Zhu, Chew Chin Yong Lionel, Jing Wen Chiu, Jinhua Lu, Haiyan Liu","doi":"10.1080/2162402X.2025.2451444","DOIUrl":"10.1080/2162402X.2025.2451444","url":null,"abstract":"<p><p>Tumor-promoting inflammation significantly impacts cancer progression, and targeting inflammatory cytokines has emerged as a promising therapeutic approach in clinical trials. Interleukin (IL)-1α, a member of the IL-1 cytokine family, plays a crucial role in both inflammation and carcinogenesis. How IL-1α is secreted in the tumor microenvironment has been poorly understood, and we previously showed that calpain 1 cleaves pro-IL-1α for mature IL-1α secretion, which exacerbates hepatocellular carcinoma by recruiting myeloid-derived suppressor cells. In this study, we report that calpain 2 also modulates IL-1α secretion. Notably, a deficiency in calpain 2 resulted in enhanced hepatocellular carcinoma development within an IL-1α-enriched tumor microenvironment. Further investigations revealed that calpain 2 deficiency increased calpain 1 expression, implying a compensatory mechanism between the two calpains. Mechanistically, calpain 2 deficiency led to increased expression of FoxO3, which is a forkhead transcription factor that promotes calpain 1 expression. Collectively, these results suggest that calpain 2 modulates calpain 1 expression, and therefore IL-1α secretion through the induction of FoxO3, offering novel potential therapeutic targets for cancer treatment.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2451444"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730618/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncoimmunologyPub Date : 2025-12-01Epub Date: 2025-05-12DOI: 10.1080/2162402X.2025.2502354
Carolina Abrate, Fernando P Canale, Sabrina N Bossio, Jimena Tosello Boari, María C Ramello, Nicolas Nuñez, Wilfrid Richer, Christine Sedlik, Jordan Denizeau, Anne Vincent-Salomon, Edith Borcoman, Andres Del Castillo, Adriana Gruppi, Eva V Acosta Rodríguez, Eliane Piaggio, Carolina L Montes
{"title":"CD8<sup>+</sup> T cells in breast cancer tumors and draining lymph nodes: PD-1 levels, effector functions and prognostic relevance.","authors":"Carolina Abrate, Fernando P Canale, Sabrina N Bossio, Jimena Tosello Boari, María C Ramello, Nicolas Nuñez, Wilfrid Richer, Christine Sedlik, Jordan Denizeau, Anne Vincent-Salomon, Edith Borcoman, Andres Del Castillo, Adriana Gruppi, Eva V Acosta Rodríguez, Eliane Piaggio, Carolina L Montes","doi":"10.1080/2162402X.2025.2502354","DOIUrl":"10.1080/2162402X.2025.2502354","url":null,"abstract":"<p><p>CD8<sup>+</sup> T cells shape the antitumor immune response. Here, we evaluated CD8<sup>+</sup> T cells expressing different levels of PD-1, their functional status, and distribution in different tissues of luminal breast cancer (BC) patients. We characterized the exhaustion stages of CD8<sup>+</sup> T cells in tumors, juxtatumoral tissues (JTs), and tumor-draining lymph nodes (TDLNs). Terminal exhausted CD8<sup>+</sup> T cells (PD-1<sup>High</sup> CD8<sup>+</sup>) were predominant in tumors and nearly absent in other tissues. However, in all tissues evaluated, most CD8<sup>+</sup> T cells exhibited a pre-exhausted phenotype (PD-1<sup>Int</sup> CD8<sup>+</sup>) or did not express PD-1. PD-1<sup>High</sup> and PD-1<sup>Int</sup> CD8<sup>+</sup> T cells from tumors and JTs presented central and effector memory phenotypes, while in TDLNs were primarily central memory. TCR-β sequencing revealed higher clonality among CD8<sup>+</sup> T cells from tumor than TDLNs, with tumor-enriched clones also detected in TDLNs. Analysis of scRNA-seq datasets from tumors and JTs of colorectal and non-small cell lung cancer patients, identified a CD8<sup>+</sup> terminal exhaustion and a CD8<sup>+</sup> pre-exhausted signatures. High expression of exhaustion-associated genes in BC tumors correlated with improved overall survival. Overall, PD-1 expression effectively distinguishes exhaustion stages in CD8<sup>+</sup> T cells. PD-1<sup>Int</sup> cells found in tumors, JTs, and TDLNs represent a promising therapeutic target for cancer immunotherapy.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2502354"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12077459/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144057818","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncoimmunologyPub Date : 2025-12-01Epub Date: 2024-12-26DOI: 10.1080/2162402X.2024.2444701
Miriam Velasco-Sidro, Javier Arroyo-Ródenas, Laura Díez-Alonso, Ángel Ramírez-Fernández, Luis Álvarez-Vallina
{"title":"Dual-targeted STAb-T cells secreting BCMA and CD19 T cell engagers for improved control of haematological cancers.","authors":"Miriam Velasco-Sidro, Javier Arroyo-Ródenas, Laura Díez-Alonso, Ángel Ramírez-Fernández, Luis Álvarez-Vallina","doi":"10.1080/2162402X.2024.2444701","DOIUrl":"https://doi.org/10.1080/2162402X.2024.2444701","url":null,"abstract":"<p><p>Despite recent advances in immunotherapy against B cell malignancies such as BCMA (B cell maturation antigen) and CD19-targeted treatments using soluble T cell-engaging (TCE) antibodies or chimeric antigen receptor T cells (CAR-T), there is still an important number of patients experiencing refractory/relapsed (R/R) disease. Approaches to avoid tumor-intrinsic mechanisms of resistance such as immune pressure-mediated antigen downmodulation, are being broadly investigated. These strategies include BCMA/CD19 dual-targeting therapies, which may be of particular interest to patients with B cell lymphoma and multiple myeloma, where a specific double-positive immature subpopulation is commonly associated with poor prognosis and poor response to current treatments. In fact, several clinical trials targeting both antigens through different strategies are currently underway. Here, based on the previously validated STAb (in situ secretion of T cell-redirecting bispecific antibodies) concept, we used two different engineering strategies (pool and co-transduction) to generate dual-targeted STAb-T cells simultaneously secreting BCMA TCE and CD19 TCE that outperformed single-targeted STAb-T cells in different <i>in vitro</i> models. These promising results encourage further preclinical clinical testing of dual STAb-T cells in R/R B-cell malignancies.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2444701"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142899531","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A targeted MAVS fusion protein for controlled innate immune activation and antitumor therapy.","authors":"Muhan Wang, Zhijie Zhang, YouYou Yang, Xiaoyi Peng, Hongping Yin","doi":"10.1080/2162402X.2025.2478850","DOIUrl":"10.1080/2162402X.2025.2478850","url":null,"abstract":"<p><p>Targeted therapies leveraging the innate immune system are emerging as promising cancer treatments. The mitochondrial antiviral signaling protein (MAVS) plays a crucial role in initiating innate immune responses, but its clinical use is limited by the risk of uncontrolled activation and systemic toxicity. To address this, we developed a novel therapeutic agent, the truncated interferon activation switch (TRIAS), combining MAVS truncates with a tumor antigen-targeting single-chain variable fragment (scFv). This design ensures antigen-dependent, controlled activation. Lentiviral delivery of TRIAS induced significant antitumor responses, including complete tumor regression in some cases. Flow cytometry (FCM) analysis further confirmed that tumor cells were the predominant population expressing the transgene. TRIAS-expressing tumor cells exhibited enhanced antitumor activity, likely due to increased cytokine release and upregulated major histocompatibility complex (MHC) expression, enabling tumor cells to function as antigen-presenting cells. This activated other immune cells, driving adaptive immune responses. Additionally, TRIAS promoted a proinflammatory shift in the tumor microenvironment (TME). In conclusion, TRIAS was validated as an innovative immunotherapeutic agent with MAVS-like immune-activating properties and tightly controlled mechanisms, offering a safer and more effective approach for clinical cancer immunotherapy.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2478850"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11913393/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143630951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncoimmunologyPub Date : 2025-12-01Epub Date: 2025-03-17DOI: 10.1080/2162402X.2025.2477872
Felipe F Lamenza, Peyton Roth, Puja Upadhaya, Suvekshya Shrestha, Sushmitha Jagadeesha, Natalie Kazmierowicz, Natalie Horn, Hasan Pracha, Sonali Dasari, Steve Oghumu
{"title":"Efficacy of anti-LAG3 and anti-PD-1 combination checkpoint inhibitor therapy against head and neck squamous cell carcinoma in a genetically engineered mouse model.","authors":"Felipe F Lamenza, Peyton Roth, Puja Upadhaya, Suvekshya Shrestha, Sushmitha Jagadeesha, Natalie Kazmierowicz, Natalie Horn, Hasan Pracha, Sonali Dasari, Steve Oghumu","doi":"10.1080/2162402X.2025.2477872","DOIUrl":"10.1080/2162402X.2025.2477872","url":null,"abstract":"<p><p>Head and neck squamous cell carcinoma (HNSCC) continues to be among the most common malignancies worldwide with limited treatment options for patients. Targeting the PD-1/PDL-1 axis is currently the only FDA approved immune checkpoint inhibitor treatment for HNSCC. Novel therapies targeting other pathways are needed along with testing a combinational approach to find new and more efficient ways to treat this disease. We utilized a tamoxifen inducible <i>TgfβR1/Pten</i> deletion mouse model to explore the efficacy of combined anti-LAG-3 and anti-PD-1 therapy against tongue HNSCC and determine underlying immunological mechanisms. Combined anti-LAG-3/anti-PD-1 therapy was effective at decreasing the tumor burden and lymphatic metastasis compared to anti-LAG-3 treatment but not when compared to the anti-PD-1 treatment alone. Anti-tumoral effects of anti-PD1 and anti-LAG-3/anti-PD-1 combined therapy were associated with increased CD4+ and CD8+ T-cell proliferative responses in secondary lymphoid organs along with increased CD8+ T-cell tumor infiltration. Anti-LAG-3 treatment potentiated the anti-tumoral properties of CD4+ T-cells treated with anti-PD-1, including enhanced systemic IFN-γ production and TNF-α production in the tumor microenvironment. Further, anti-tumoral cytotoxic CD8+ T-cell effector function and granzyme B production were enhanced by anti-PD-1 and combinatorial anti-LAG-3/anti-PD-1 immunotherapy, resulting in greater tumor cell death. Our results demonstrate that anti-LAG-3 has the potential to enhance the efficacy of anti-PD-1 therapy; however, humanized mouse models that better recapitulate the human disease with FDA approved antibodies are needed to further characterize the efficacy of this treatment as a viable treatment option for HNSCC patients.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2477872"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143651566","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncoimmunologyPub Date : 2025-12-01Epub Date: 2025-02-18DOI: 10.1080/2162402X.2025.2466308
Daniel A Ruiz-Torres, Michael E Bryan, Shun Hirayama, Ross D Merkin, Evelyn Luciani, Thomas J Roberts, Manisha Patel, Jong C Park, Lori J Wirth, Peter M Sadow, Moshe Sade-Feldman, Shannon L Stott, Daniel L Faden
{"title":"Spatial characterization of tertiary lymphoid structures as predictive biomarkers for immune checkpoint blockade in head and neck squamous cell carcinoma.","authors":"Daniel A Ruiz-Torres, Michael E Bryan, Shun Hirayama, Ross D Merkin, Evelyn Luciani, Thomas J Roberts, Manisha Patel, Jong C Park, Lori J Wirth, Peter M Sadow, Moshe Sade-Feldman, Shannon L Stott, Daniel L Faden","doi":"10.1080/2162402X.2025.2466308","DOIUrl":"10.1080/2162402X.2025.2466308","url":null,"abstract":"<p><p>Immune checkpoint blockade (ICB) is the standard of care for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), yet efficacy remains low. The combined positive score (CPS) for PD-L1 is the only biomarker approved to predict response to ICB and has limited performance. Tertiary Lymphoid Structures (TLS) have shown promising potential for predicting response to ICB. However, their exact composition, size, and spatial biology in HNSCC remain understudied. To elucidate the impact of TLS spatial biology in response to ICB, we utilized pre-ICB tumor tissue sections from 9 responders (complete response, partial response, or stable disease) and 11 non-responders (progressive disease) classified via RECISTv1.1. A custom multi-immunofluorescence (mIF) staining assay was applied to characterize tumor cells (pan-cytokeratin), T cells (CD4, CD8), B cells (CD19, CD20), myeloid cells (CD16, CD56, CD163), dendritic cells (LAMP3), fibroblasts (α Smooth Muscle Actin), proliferative status (Ki67) and immunoregulatory molecules (PD1). A machine learning model was employed to measure the effect of spatial metrics on achieving a response to ICB. A higher density of B cells (CD20+) was found in responders compared to non-responders to ICB (<i>p</i> = 0.022). The presence of TLS within 100 µm of the tumor was associated with improved overall (<i>p</i> = 0.04) and progression-free survival (<i>p</i> = 0.03). A multivariate machine learning model identified TLS density as a leading predictor of response to ICB with 80% accuracy. Immune cell densities and TLS spatial location play a critical role in the response to ICB in HNSCC and may potentially outperform CPS as a predictor of response.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2466308"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11845054/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143442459","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncoimmunologyPub Date : 2025-12-01Epub Date: 2025-05-26DOI: 10.1080/2162402X.2025.2510998
Leire Arrizabalaga, Ignacio Melero, Pedro Berraondo, Fernando Aranda
{"title":"Routing immunomodulatory cytokine-encoded mRNAs to the omentum: turning an enemy into an ally in peritoneal metastasis.","authors":"Leire Arrizabalaga, Ignacio Melero, Pedro Berraondo, Fernando Aranda","doi":"10.1080/2162402X.2025.2510998","DOIUrl":"10.1080/2162402X.2025.2510998","url":null,"abstract":"<p><p>Peritoneal metastases remain a significant clinical challenge owing to the immunosuppressive nature of the omentum, which serves as a protective niche for disseminated tumor cells. Our recent study explored the targeted delivery of immunomodulatory cytokine-encoded mRNAs to the omentum, aiming to shift its role from a tumor-supportive site to an immune-activating ally. Our findings highlight the potential of cytokine mRNA delivery as a novel and safe immunotherapeutic strategy for peritoneal metastases.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2510998"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12118438/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144152626","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
OncoimmunologyPub Date : 2025-12-01Epub Date: 2024-12-18DOI: 10.1080/2162402X.2024.2432726
Pierre-Antoine Laurent, Fabrice André, Alexandre Bobard, Desiree Deandreis, Sandra Demaria, Stephane Depil, Stefan B Eichmüller, Cristian Fernandez-Palomo, Floris Foijer, Lorenzo Galluzzi, Jérôme Galon, Matthias Guckenberger, Kevin J Harrington, Fernanda G Herrera, Peter E Huber, Antoine Italiano, Sana D Karam, Guido Kroemer, Philippe Lambin, Carola Leuschner, Alberto Mantovani, Etienne Meylan, Michele Mondini, Mikael J Pittet, Jean-Pierre Pouget, Jordi Remon, Claus S Sørensen, Christos Sotiriou, Claire Vanpouille-Box, Ralph R Weichselbaum, James W Welsh, Laurence Zitvogel, Silvia C Formenti, Eric Deutsch
{"title":"Pushing the boundaries of radiotherapy-immunotherapy combinations: highlights from the 7<sup>th</sup> immunorad conference.","authors":"Pierre-Antoine Laurent, Fabrice André, Alexandre Bobard, Desiree Deandreis, Sandra Demaria, Stephane Depil, Stefan B Eichmüller, Cristian Fernandez-Palomo, Floris Foijer, Lorenzo Galluzzi, Jérôme Galon, Matthias Guckenberger, Kevin J Harrington, Fernanda G Herrera, Peter E Huber, Antoine Italiano, Sana D Karam, Guido Kroemer, Philippe Lambin, Carola Leuschner, Alberto Mantovani, Etienne Meylan, Michele Mondini, Mikael J Pittet, Jean-Pierre Pouget, Jordi Remon, Claus S Sørensen, Christos Sotiriou, Claire Vanpouille-Box, Ralph R Weichselbaum, James W Welsh, Laurence Zitvogel, Silvia C Formenti, Eric Deutsch","doi":"10.1080/2162402X.2024.2432726","DOIUrl":"10.1080/2162402X.2024.2432726","url":null,"abstract":"<p><p>Over the last decade, the annual Immunorad Conference, held under the joint auspicies of Gustave Roussy (Villejuif, France) and the Weill Cornell Medical College (New-York, USA) has aimed at exploring the latest advancements in the fields of tumor immunology and radiotherapy-immunotherapy combinations for the treatment of cancer. Gathering medical oncologists, radiation oncologists, physicians and researchers with esteemed expertise in these fields, the Immunorad Conference bridges the gap between preclinical outcomes and clinical opportunities. Thus, it paves a promising way toward optimizing radiotherapy-immunotherapy combinations and, from a broader perspective, improving therapeutic strategies for patients with cancer. Herein, we report on the topics developed by key-opinion leaders during the 7<sup>th</sup> Immunorad Conference held in Paris-Les Cordeliers (France) from September 27th to 29th 2023, and set the stage for the 8<sup>th</sup> edition of Immunorad which will be held at Weill Cornell Medical College (New-York, USA) in October 2024.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2432726"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11660406/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142856327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}