OncoimmunologyPub Date : 2023-11-27DOI: 10.1080/2162402x.2023.2284486
Killian Carnet Le Provost, Oliver Kepp, Guido Kroemer, Lucillia Bezu
{"title":"Trial watch: beta-blockers in cancer therapy","authors":"Killian Carnet Le Provost, Oliver Kepp, Guido Kroemer, Lucillia Bezu","doi":"10.1080/2162402x.2023.2284486","DOIUrl":"https://doi.org/10.1080/2162402x.2023.2284486","url":null,"abstract":"Compelling evidence supports the hypothesis that stress negatively impacts cancer development and prognosis. Irrespective of its physical, biological or psychological source, stress triggers a phys...","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138516200","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 : 2023-11-22DOI: 10.1080/2162402x.2023.2281179
muta abiff, Mohammad Alshebremi, Melissa Bonner, Jay T. Myers, Byung-Gyu Kim, Suzanne L. Tomchuck, Alicia Santin, Daniel Kingsley, Sung Hee Choi, Alex Y. Huang
{"title":"Piezo1 facilitates optimal T cell activation during tumor challenge","authors":"muta abiff, Mohammad Alshebremi, Melissa Bonner, Jay T. Myers, Byung-Gyu Kim, Suzanne L. Tomchuck, Alicia Santin, Daniel Kingsley, Sung Hee Choi, Alex Y. Huang","doi":"10.1080/2162402x.2023.2281179","DOIUrl":"https://doi.org/10.1080/2162402x.2023.2281179","url":null,"abstract":"Functional effector T cells in the tumor microenvironment (TME) are critical for successful anti-tumor responses. T cell anti-tumor function is dependent on their ability to differentiate from a na...","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138516184","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 : 2023-11-22DOI: 10.1080/2162402x.2023.2283353
Fei Gao, Mauricio Campos Mora, Michael Constantinides, Loïs Coenon, Caroline Multrier, Loïc Vaillant, Tianxiang Zhang, Martin Villalba
{"title":"g-NK cells from umbilical cord blood are phenotypically and functionally different than g-NK cells from peripheral blood","authors":"Fei Gao, Mauricio Campos Mora, Michael Constantinides, Loïs Coenon, Caroline Multrier, Loïc Vaillant, Tianxiang Zhang, Martin Villalba","doi":"10.1080/2162402x.2023.2283353","DOIUrl":"https://doi.org/10.1080/2162402x.2023.2283353","url":null,"abstract":"FcRγ-deficient natural killer (NK) cells, designated as g-NK cells, exhibit enhanced antibody-dependent cellular cytotoxicity (ADCC) capacity and increased IFN-γ and TNF-α production, rendering the...","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138516185","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 : 2023-11-20DOI: 10.1080/2162402x.2023.2279800
Baojun Wang, Bo Jiang, Lin Du, Wenyuan Chen, Qing Zhang, Wei Chen, Meng Ding, Wenmin Cao, Jie Gao, Yongming Deng, Yao Fu, Yan Li, Yonglong Xiao, Wenli Diao, Hongqian Guo
{"title":"Tumor-intrinsic RGS1 potentiates checkpoint blockade response via ATF3-IFNGR1 axis","authors":"Baojun Wang, Bo Jiang, Lin Du, Wenyuan Chen, Qing Zhang, Wei Chen, Meng Ding, Wenmin Cao, Jie Gao, Yongming Deng, Yao Fu, Yan Li, Yonglong Xiao, Wenli Diao, Hongqian Guo","doi":"10.1080/2162402x.2023.2279800","DOIUrl":"https://doi.org/10.1080/2162402x.2023.2279800","url":null,"abstract":"Non-responsiveness is a major barrier in current cancer immune checkpoint blockade therapies, and the mechanism has not been elucidated yet. Therefore, it is necessary to discover the mechanism and...","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138516199","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 : 2023-11-20DOI: 10.1080/2162402x.2023.2282252
Jiao Liu, Jingbo Li, Rui Kang, Daolin Tang
{"title":"Cell type-specific induction of ferroptosis to boost antitumor immunity","authors":"Jiao Liu, Jingbo Li, Rui Kang, Daolin Tang","doi":"10.1080/2162402x.2023.2282252","DOIUrl":"https://doi.org/10.1080/2162402x.2023.2282252","url":null,"abstract":"Traditional ferroptosis activators typically suppress antitumor immunity. Our discovery shows that N6F11, a small molecule compound, can selectively induce ferroptosis by targeting TRIM25-mediated ...","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138543694","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 : 2023-11-20DOI: 10.1080/2162402x.2023.2285106
Ángela Bella, Ignacio Melero, Pedro Berraondo, Fernando Aranda
{"title":"The significance of the omentum in locoregional immunotherapy for peritoneal carcinomatosis","authors":"Ángela Bella, Ignacio Melero, Pedro Berraondo, Fernando Aranda","doi":"10.1080/2162402x.2023.2285106","DOIUrl":"https://doi.org/10.1080/2162402x.2023.2285106","url":null,"abstract":"Our recent research has unveiled the potential of locoregional immunotherapy. Cytokine-armored viral vectors, such as modified vaccinia virus Ankara vector encoding single-chain interleukin-12 (MVA...","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138516201","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 : 2023-10-05eCollection Date: 2023-01-01DOI: 10.1080/2162402X.2023.2261326
Constanza Rodriguez, Cintia L Araujo Furlan, Jimena Tosello Boari, Sabrina N Bossio, Santiago Boccardo, Laura Fozzatti, Fernando P Canale, Cristian G Beccaria, Nicolás G Nuñez, Danilo G Ceschin, Eliane Piaggio, Adriana Gruppi, Carolina L Montes, Eva V Acosta Rodríguez
{"title":"Interleukin-17 signaling influences CD8<sup>+</sup> T cell immunity and tumor progression according to the IL-17 receptor subunit expression pattern in cancer cells.","authors":"Constanza Rodriguez, Cintia L Araujo Furlan, Jimena Tosello Boari, Sabrina N Bossio, Santiago Boccardo, Laura Fozzatti, Fernando P Canale, Cristian G Beccaria, Nicolás G Nuñez, Danilo G Ceschin, Eliane Piaggio, Adriana Gruppi, Carolina L Montes, Eva V Acosta Rodríguez","doi":"10.1080/2162402X.2023.2261326","DOIUrl":"10.1080/2162402X.2023.2261326","url":null,"abstract":"<p><p>IL-17 immune responses in cancer are controversial, with both tumor-promoting and tumor-repressing effects observed. To clarify the role of IL-17 signaling in cancer progression, we used syngeneic tumor models from different tissue origins. We found that deficiencies in host IL-17RA or IL-17A/F expression had varying effects on the <i>in vivo</i> growth of different solid tumors including melanoma, sarcoma, lymphoma, and leukemia. In each tumor type, the absence of IL-17 led to changes in the expression of mediators associated with inflammation and metastasis in the tumor microenvironment. Furthermore, IL-17 signaling deficiencies in the hosts resulted in decreased anti-tumor CD8<sup>+</sup> T cell immunity and caused tumor-specific changes in several lymphoid cell populations. Our findings were associated with distinct patterns of IL-17A/F cytokine and receptor subunit expression in the injected tumor cell lines. These patterns affected tumor cell responsiveness to IL-17 and downstream intracellular signaling, leading to divergent effects on cancer progression. Additionally, we identified IL-17RC as a critical determinant of the IL-17-mediated response in tumor cells and a potential biomarker for IL-17 signaling effects in tumor progression. Our study offers insight into the molecular mechanisms underlying IL-17 activities in cancer and lays the groundwork for developing personalized immunotherapies.</p>","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/fc/70/KONI_12_2261326.PMC10557545.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41133221","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":"Enhancing T cell anti-tumor efficacy with a PD1-TIGIT chimeric immune-checkpoint switch receptor.","authors":"Jingjing Zhao, Jiebin Dong, Changwen Deng, Qianjing Zhang, Shicheng Sun, Honggang Li, Yun Bai, Hongkui Deng","doi":"10.1080/2162402X.2023.2265703","DOIUrl":"10.1080/2162402X.2023.2265703","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T cell immunotherapy has demonstrated success in the treatment of hematological malignancies; however, its efficacy and applications in solid tumors remain limited. Immunosuppressive factors, particularly inhibitory checkpoint molecules, restrict CAR T cell activity inside solid tumors. The modulation of checkpoint pathways has emerged as a promising approach to promote anti-tumor responses in CAR T cells. Programmed cell death protein 1 (PD1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) are two critical immune-checkpoint molecules that suppress anti-tumor activity in T cells. Simultaneous targeting of these two inhibitory molecules could be an efficient checkpoint modulation strategy. Here, we developed a PD1-TIGIT chimeric immune-checkpoint switch receptor (CISR) that enhances the efficacy of CAR T cell immunotherapy by reversing the inhibitory checkpoint signals of PD1/PDL1 and/or TIGIT/CD155. In addition to neutralizing PDL1 and CD155, this chimeric receptor is engineered with the transmembrane region and intracellular domain of CD28, thereby effectively enhancing T cell survival and tumor-targeting functions. Notably, under simultaneous stimulation of PDL1 and CD155, CISR-CAR T cells demonstrate superior performance in terms of cell survival, proliferation, cytokine release, and cytotoxicity <i>in vitro</i>, compared with conventional CAR T cells. Experiments utilizing both cell line- and patient-derived xenotransplantation tumor models showed that CISR-CAR T cells exhibit robust infiltration and anti-tumor efficiency <i>in vivo</i>. Our results highlight the potential for the CISR strategy to enhance T cell anti-tumor efficacy and provide an alternative approach for T cell-based immunotherapies.</p>","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/5b/67/KONI_12_2265703.PMC10557556.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41158875","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 : 2023-10-04eCollection Date: 2023-01-01DOI: 10.1080/2162402X.2023.2261248
Tuba N Gide, Elizabeth C Paver, Zarwa Yaseen, Nigel Maher, Nurudeen Adegoke, Alexander M Menzies, Ines Pires da Silva, James S Wilmott, Georgina V Long, Richard A Scolyer
{"title":"Lag-3 expression and clinical outcomes in metastatic melanoma patients treated with combination anti-lag-3 + anti-PD-1-based immunotherapies.","authors":"Tuba N Gide, Elizabeth C Paver, Zarwa Yaseen, Nigel Maher, Nurudeen Adegoke, Alexander M Menzies, Ines Pires da Silva, James S Wilmott, Georgina V Long, Richard A Scolyer","doi":"10.1080/2162402X.2023.2261248","DOIUrl":"10.1080/2162402X.2023.2261248","url":null,"abstract":"<p><p>Lymphocyte-activation gene-3 (LAG-3), an immune checkpoint receptor, negatively regulates T-cell function and facilitates immune escape of tumors. Dual inhibition of LAG-3 and programmed cell death receptor-1 (PD-1) significantly improved progression-free survival (PFS) in metastatic melanoma patients compared to anti-PD-1 therapy alone. Investigating the utility of LAG-3 expression as a biomarker of response to anti-LAG-3 + anti-PD-1 immunotherapy is of great clinical relevance. This study sought to evaluate the association between baseline LAG-3 expression and clinical outcomes following anti-LAG-3 and anti-PD-1-based immunotherapy in metastatic melanoma. LAG-3 immunohistochemistry (clone D2G4O) was performed on pre-treatment formalin-fixed, paraffin-embedded metastatic melanoma specimens from 53 patients treated with combination anti-LAG-3 + anti-PD-1-based therapies. Eleven patients had received prior anti-PD-1-based treatment. Patients were categorized as responders (complete/partial response; <i>n</i> = 36) or non-responders (stable/progressive disease; <i>n</i> = 17) based on the Response Evaluation Criteria in Solid Tumours (RECIST). Tumor-infiltrating lymphocytes (TILs) were scored on hematoxylin and eosin-stained sections. LAG-3 expression was observed in 81% of patients, with staining in TILs and dendritic cells. Responders displayed significantly higher proportions of LAG-3+ cells compared to non-responders (<i>P</i> = .0210). LAG-3 expression positively correlated with TIL score (<i>P</i> < .01). There were no significant differences in LAG-3 expression between different sites of metastases (<i>P</i> > .05). Patients with ≥ 1% LAG-3+ cells in their tumors had significantly longer PFS compared to patients with < 1% LAG-3 expression (<i>P</i> = .0037). No significant difference was observed in overall survival between the two groups (<i>P</i> = .1417). Therefore, the assessment of LAG-3 expression via IHC warrants further evaluation to determine its role as a predictive marker of response and survival in metastatic melanoma.</p>","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10558007/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41137484","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 : 2023-09-30eCollection Date: 2023-01-01DOI: 10.1080/2162402X.2023.2261242
Chen Zhang, Kankan Cao, Moran Yang, Yiying Wang, Mengdi He, Jiaqi Lu, Yan Huang, Guodong Zhang, Haiou Liu
{"title":"C5aR1 blockade reshapes immunosuppressive tumor microenvironment and synergizes with immune checkpoint blockade therapy in high-grade serous ovarian cancer.","authors":"Chen Zhang, Kankan Cao, Moran Yang, Yiying Wang, Mengdi He, Jiaqi Lu, Yan Huang, Guodong Zhang, Haiou Liu","doi":"10.1080/2162402X.2023.2261242","DOIUrl":"https://doi.org/10.1080/2162402X.2023.2261242","url":null,"abstract":"<p><p>High-grade serous ovarian cancer (HGSC), with a modest response to immune checkpoint blockade (ICB) targeting PD-1/PD-L1 monotherapy, is densely infiltrated by M2-polarized tumor-associated macrophages (TAMs) and regulatory T (Treg) cells. The complement C5a/C5aR1 axis contributes to the programming of the immunosuppressive phenotype of TAMs in solid tumors and represents a promising immunomodulatory target for treating HGSCs. Here, we aimed to identify the relevance of C5aR1 in prognosis, immune microenvironment, and immunotherapy response in HGSCs. The expression and relationship of C5aR1 with tumor-infiltrating immune cells were assessed by immunohistochemistry and flow cytometry in the training cohort (<i>n</i> = 120) and fresh HGSC tissues (<i>n</i> = 36). Transcriptomic analyses of the xenografts delineated the mechanisms driving the immunomodulatory activity of PMX53, an orally bioavailable C5aR1 inhibitor. Therapeutic relevance was confirmed in ex vivo tumor cultures and The Cancer Genome Atlas (TCGA) datasets. C5aR1 expression independently predicted dismal prognosis and was linked to the immunoevasive subtype of HGSC, characterized by increased infiltration of pro-tumor cells (Treg cells, M2-polarized macrophages, and neutrophils) and impaired CD8<sup>+</sup>T functions. PMX53 antagonized subcutaneous tumor growth, modulated immunosuppressive mechanisms and synergized with aPD-1 in several tumor types. Single-cell RNA-seq analysis revealed predominant C5aR1 expression in TAMs, with an immunosuppressive-related expression signature in C5aR1<sup>+</sup>TAMs. Furthermore, the combination of C5aR1 and PD-L1 was associated with specific molecular characteristics and matched clinical response annotations. Therefore, the abundance of C5aR1 could predict an inferior prognosis in HGSCs, and incorporating PD-L1 may serve as a novel predictive biomarker to guide therapeutic options.</p>","PeriodicalId":19683,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2023-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/72/97/KONI_12_2261242.PMC10543342.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41148043","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}