首页 > 最新文献

Oncoimmunology最新文献

英文 中文
CyTOF profiling of bone marrow immune dynamics across myeloma stages. 骨髓免疫动力学在骨髓瘤分期的CyTOF分析。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-08-18 DOI: 10.1080/2162402X.2025.2542333
Dana Cholujova, Gabor Beke, Lubos Klucar, Lubos Drgona, Zuzana Valuskova, Merav Leiba, Efstathios Kastritis, David M Dorfman, Kenneth C Anderson, Jana Jakubikova

Multiple myeloma (MM) orchestrates a profound disruption of immune balance within the bone marrow (BM) microenvironment, driving disease progression and therapeutic resistance. To better understand these complex immune dynamics, we used high-dimensional mass cytometry (CyTOF) profiling to comprehensively characterize the immune landscape of the BM across different stages of myeloma progression, including MGUS (n = 16), smoldering MM (SMM; n = 25), and active MM, both newly diagnosed (n = 43) and relapsed/refractory (n = 104). Our analysis revealed substantial immune remodeling during disease progression, characterized by adaptive immune suppression and extensive infiltration of innate immune populations. Transformation from MGUS to SMM was marked by significant alterations in central and effector memory T helper cells, effector cytotoxic T cells, and an enrichment of monocytic and neutrophil subsets. Active MM stages were further distinguished by increased expansion of myeloid and monocytic lineages, alongside a pronounced reduction in progenitor and transitional B cells. Correspondence analysis demonstrated that specific immune profiles were significantly associated with clinical outcomes, including progression-free survival and overall survival. This study highlights the potential of CyTOF-based molecular profiling to unravel the intricate immune dynamics of the BM microenvironment across MM disease stages, enhancing our understanding of MM pathogenesis and providing a foundation for identifying prognostic biomarkers and tailoring precision immunotherapeutic strategies.

多发性骨髓瘤(MM)协调骨髓(BM)微环境中免疫平衡的深刻破坏,驱动疾病进展和治疗耐药性。为了更好地理解这些复杂的免疫动力学,我们使用高维质量细胞术(CyTOF)分析来全面表征骨髓瘤在不同进展阶段的免疫景观,包括MGUS (n = 16)、阴发性MM (SMM; n = 25)和活动性MM,包括新诊断的(n = 43)和复发/难治性MM (n = 104)。我们的分析揭示了疾病进展过程中大量的免疫重塑,其特征是适应性免疫抑制和先天免疫群体的广泛浸润。从MGUS到SMM的转化标志着中枢和效应记忆T辅助细胞、效应细胞毒性T细胞的显著改变,以及单核细胞和中性粒细胞亚群的富集。髓系和单核细胞谱系增加,祖细胞和移行B细胞明显减少,进一步区分了活动性MM分期。对应分析表明,特异性免疫谱与临床结果显著相关,包括无进展生存期和总生存期。这项研究强调了基于细胞f的分子谱分析在揭示MM疾病阶段BM微环境复杂的免疫动力学方面的潜力,增强了我们对MM发病机制的理解,并为确定预后生物标志物和定制精确的免疫治疗策略提供了基础。
{"title":"CyTOF profiling of bone marrow immune dynamics across myeloma stages.","authors":"Dana Cholujova, Gabor Beke, Lubos Klucar, Lubos Drgona, Zuzana Valuskova, Merav Leiba, Efstathios Kastritis, David M Dorfman, Kenneth C Anderson, Jana Jakubikova","doi":"10.1080/2162402X.2025.2542333","DOIUrl":"10.1080/2162402X.2025.2542333","url":null,"abstract":"<p><p>Multiple myeloma (MM) orchestrates a profound disruption of immune balance within the bone marrow (BM) microenvironment, driving disease progression and therapeutic resistance. To better understand these complex immune dynamics, we used high-dimensional mass cytometry (CyTOF) profiling to comprehensively characterize the immune landscape of the BM across different stages of myeloma progression, including MGUS (<i>n</i> = 16), smoldering MM (SMM; <i>n</i> = 25), and active MM, both newly diagnosed (<i>n</i> = 43) and relapsed/refractory (<i>n</i> = 104). Our analysis revealed substantial immune remodeling during disease progression, characterized by adaptive immune suppression and extensive infiltration of innate immune populations. Transformation from MGUS to SMM was marked by significant alterations in central and effector memory T helper cells, effector cytotoxic T cells, and an enrichment of monocytic and neutrophil subsets. Active MM stages were further distinguished by increased expansion of myeloid and monocytic lineages, alongside a pronounced reduction in progenitor and transitional B cells. Correspondence analysis demonstrated that specific immune profiles were significantly associated with clinical outcomes, including progression-free survival and overall survival. This study highlights the potential of CyTOF-based molecular profiling to unravel the intricate immune dynamics of the BM microenvironment across MM disease stages, enhancing our understanding of MM pathogenesis and providing a foundation for identifying prognostic biomarkers and tailoring precision immunotherapeutic strategies.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2542333"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12363513/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144876219","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}
引用次数: 0
Divergent on-target off-tumor effects by CAR T and CAR NK cells suggest different efficacy and safety of cell therapies. CAR - T和CAR - NK细胞不同的靶外肿瘤效应表明细胞疗法的疗效和安全性不同。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-09-05 DOI: 10.1080/2162402X.2025.2546443
Katharina Schindler-Wnek, Anika Stahringer, Nadine Heimer, Ulrike Koehl, Stephan Fricke, Dominik Schmiedel

CAR-based cell therapies have shown clinical success in treating various cancers, with CAR T cell therapies entering the clinical route and CAR NK cell therapies being evaluated in early-stage clinical trials. A key challenge is the presence of tumor-associated antigens on healthy cells, risking on-target off-tumor toxicities. Our comparative analysis of CAR T and CAR NK cells targeting the multiple myeloma-associated antigens BCMA, SLAMF7, and CD38 revealed that antigen density on target cells significantly modulates CAR NK cell activation and cytotoxicity. The cytotoxic potential of CAR NK cells was comparable to that of CAR T cells when targeting BCMA and CD38, but notable differences were observed in SLAMF7-directed CAR cells. While CAR sensitivity was similar in both cell types, CAR NK cell activity was balanced by inhibitory receptors like KIRs and NKG2A. This balance allows effective tumor control while potentially reducing on-target off-tumor effects on healthy cells with low antigen expression. Consequently, CAR NK cells offer greater flexibility in target antigen selection, potentially expanding the range of targetable antigens for cancer immunotherapy.

基于CAR的细胞疗法在治疗各种癌症方面取得了临床成功,CAR- T细胞疗法进入临床途径,CAR- NK细胞疗法正在早期临床试验中进行评估。一个关键的挑战是肿瘤相关抗原在健康细胞上的存在,有可能产生靶外肿瘤毒性。我们对靶向多发性骨髓瘤相关抗原BCMA、SLAMF7和CD38的CAR - T和CAR - NK细胞进行比较分析,发现靶细胞上的抗原密度显著调节CAR - NK细胞的活化和细胞毒性。当靶向BCMA和CD38时,CAR NK细胞的细胞毒性潜能与CAR T细胞相当,但在靶向slamf7的CAR细胞中观察到显著差异。虽然两种细胞类型的CAR敏感性相似,但CAR NK细胞的活性由抑制受体如KIRs和NKG2A来平衡。这种平衡允许有效的肿瘤控制,同时潜在地减少对低抗原表达的健康细胞的靶外肿瘤效应。因此,CAR - NK细胞在靶抗原选择方面提供了更大的灵活性,潜在地扩大了癌症免疫治疗的可靶向抗原范围。
{"title":"Divergent on-target off-tumor effects by CAR T and CAR NK cells suggest different efficacy and safety of cell therapies.","authors":"Katharina Schindler-Wnek, Anika Stahringer, Nadine Heimer, Ulrike Koehl, Stephan Fricke, Dominik Schmiedel","doi":"10.1080/2162402X.2025.2546443","DOIUrl":"10.1080/2162402X.2025.2546443","url":null,"abstract":"<p><p>CAR-based cell therapies have shown clinical success in treating various cancers, with CAR T cell therapies entering the clinical route and CAR NK cell therapies being evaluated in early-stage clinical trials. A key challenge is the presence of tumor-associated antigens on healthy cells, risking on-target off-tumor toxicities. Our comparative analysis of CAR T and CAR NK cells targeting the multiple myeloma-associated antigens BCMA, SLAMF7, and CD38 revealed that antigen density on target cells significantly modulates CAR NK cell activation and cytotoxicity. The cytotoxic potential of CAR NK cells was comparable to that of CAR T cells when targeting BCMA and CD38, but notable differences were observed in SLAMF7-directed CAR cells. While CAR sensitivity was similar in both cell types, CAR NK cell activity was balanced by inhibitory receptors like KIRs and NKG2A. This balance allows effective tumor control while potentially reducing on-target off-tumor effects on healthy cells with low antigen expression. Consequently, CAR NK cells offer greater flexibility in target antigen selection, potentially expanding the range of targetable antigens for cancer immunotherapy.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2546443"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12416173/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145001706","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}
引用次数: 0
An SNP-dependent cancer-testis antigenic epitope serves as a promising immunotherapeutic target for cancer. 一个snp依赖的癌睾丸抗原表位作为一个有希望的癌症免疫治疗靶点。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-07-09 DOI: 10.1080/2162402X.2025.2528110
Kenji Murata, Tomoyuki Minowa, Tomohide Tsukahara, Taku Yoshida, Akiko Minami, Munehide Nakatsugawa, Yuka Mizue, Aiko Murai, Serina Tokita, Kenta Sasaki, Hisashi Uhara, Terufumi Kubo, Takayuki Kanaseki, Toshihiko Torigoe, Yoshihiko Hirohashi

T cells recognize peptides presented by human leukocyte antigen molecules on the cell surface, enabling the immune surveillance of pathological conditions such as cancer. Cancer-testis (CT) antigens are promising targets for cancer immunotherapy because of their restricted expression in normal tissues. In this study, we performed antigen screening of T cell receptors isolated from tumor-infiltrating lymphocytes (TILs) in acral melanoma, using cDNA expression cloning and identified a novel CT antigenic epitope encoded by MAGE-A6 with a single nucleotide polymorphism (SNP). This SNP conferred immunogenicity to the epitope, eliciting a robust immune response against tumor cells. While antigen identification has increasingly relied on reverse immunology approaches using reference sequences that do not contain SNPs, forward immunology approaches, such as cDNA expression cloning, directly identify antigens recognized by T cells exhibiting immune responses, enabling the detection of SNP-derived epitopes. Furthermore, in hot tumors such as acral melanoma that are characterized by a low tumor mutational burden, but high TIL infiltration, TILs predominantly respond to shared antigens with high immunogenicity. These findings underscore the utility of forward immunology in antigen discovery and highlight the potential of SNP-dependent tumor antigens in cancer immunotherapy.

T细胞识别人类白细胞抗原分子在细胞表面呈现的肽,从而实现对病理状况(如癌症)的免疫监视。癌睾丸(CT)抗原在正常组织中表达受限,是肿瘤免疫治疗的重要靶点。在这项研究中,我们利用cDNA表达克隆技术对肢端黑色素瘤肿瘤浸润淋巴细胞(TILs)中分离的T细胞受体进行抗原筛选,并鉴定出一个由MAGE-A6编码的具有单核苷酸多态性(SNP)的新型CT抗原表位。这种SNP赋予表位免疫原性,引发针对肿瘤细胞的强大免疫反应。虽然抗原鉴定越来越依赖于使用不含snp的参考序列的反向免疫学方法,但正向免疫学方法,如cDNA表达克隆,直接鉴定具有免疫应答的T细胞识别的抗原,从而能够检测snp衍生的表位。此外,在肢端黑色素瘤等肿瘤中,其特点是肿瘤突变负担低,但TIL浸润率高,TIL主要对具有高免疫原性的共享抗原产生反应。这些发现强调了正向免疫学在抗原发现中的作用,并强调了snp依赖性肿瘤抗原在癌症免疫治疗中的潜力。
{"title":"An SNP-dependent cancer-testis antigenic epitope serves as a promising immunotherapeutic target for cancer.","authors":"Kenji Murata, Tomoyuki Minowa, Tomohide Tsukahara, Taku Yoshida, Akiko Minami, Munehide Nakatsugawa, Yuka Mizue, Aiko Murai, Serina Tokita, Kenta Sasaki, Hisashi Uhara, Terufumi Kubo, Takayuki Kanaseki, Toshihiko Torigoe, Yoshihiko Hirohashi","doi":"10.1080/2162402X.2025.2528110","DOIUrl":"10.1080/2162402X.2025.2528110","url":null,"abstract":"<p><p>T cells recognize peptides presented by human leukocyte antigen molecules on the cell surface, enabling the immune surveillance of pathological conditions such as cancer. Cancer-testis (CT) antigens are promising targets for cancer immunotherapy because of their restricted expression in normal tissues. In this study, we performed antigen screening of T cell receptors isolated from tumor-infiltrating lymphocytes (TILs) in acral melanoma, using cDNA expression cloning and identified a novel CT antigenic epitope encoded by <i>MAGE-A6</i> with a single nucleotide polymorphism (SNP). This SNP conferred immunogenicity to the epitope, eliciting a robust immune response against tumor cells. While antigen identification has increasingly relied on reverse immunology approaches using reference sequences that do not contain SNPs, forward immunology approaches, such as cDNA expression cloning, directly identify antigens recognized by T cells exhibiting immune responses, enabling the detection of SNP-derived epitopes. Furthermore, in hot tumors such as acral melanoma that are characterized by a low tumor mutational burden, but high TIL infiltration, TILs predominantly respond to shared antigens with high immunogenicity. These findings underscore the utility of forward immunology in antigen discovery and highlight the potential of SNP-dependent tumor antigens in cancer immunotherapy.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2528110"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12243901/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144592698","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}
引用次数: 0
Neutrophil extracellular trap inhibition revitalizes PDAC immunotherapy responsiveness via reduced fibrosis and TCF1+CD8+ progenitor T-cell expansion. 中性粒细胞胞外陷阱抑制通过减少纤维化和TCF1+CD8+祖t细胞扩增来恢复PDAC免疫治疗反应性。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-12-18 DOI: 10.1080/2162402X.2025.2601394
Britney Niemann, Abby Ivey, Quinn Hopen, Duaa Dakhlallah, Kathleen Brundage, Nicole Mihalik, Timothy Eubank, Brian A Boone

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor survival. The immunosuppressive tumor microenvironment (TME) drives resistance to therapy, including immunotherapy. This may be, in part, mediated by the formation of neutrophil extracellular traps (NETs), formed when neutrophils release their intracellular contents. NETs are elevated in PDAC and are associated with nearly every stage of tumor progression. We investigated the influence of NETs on the PDAC TME and immunotherapy response. An orthotopic PDAC model was utilized in C57BJ6 or PAD4-/- mice receiving one of the following: control, DNase, anti-PD-1 therapy, or DNase and anti-PD-1. NET markers, fibrosis, and the TME immune profile were evaluated. Human PDAC patients were also evaluated for levels of NETs and tumor-infiltrating T cells. Circulating NET markers correlated with intra-tumoral CD8+ cells in PDAC patients. Patients with high NET levels also experienced more post-operative complications. NET inhibition in mice reduced tumor growth and enhanced survival. Decreased expression of collagen and matrix metalloproteinase (MMP) genes, as well as reduced intra-tumoral collagen deposition were found in NET deficient mice. Additionally, an increase in TCF1+PD-1+CD44+CD8+ progenitor T cells, a subpopulation of T cells responsive to immunotherapy, were identified. These changes resulted in further tumor burden reduction and prolonged survival when anti-PD-1 therapy was given in conjunction with NET inhibition. NETs influence extracellular matrix remodeling and the T cell response to PDAC, allowing for a significant response to anti-PD-1 therapy. These findings support the combination therapy of immunotherapy and NET inhibition in patients with PDAC.

胰腺导管腺癌(PDAC)是一种侵袭性疾病,生存率低。免疫抑制肿瘤微环境(TME)驱动对包括免疫治疗在内的治疗的耐药性。这可能部分是由中性粒细胞释放其细胞内内容物时形成的中性粒细胞胞外陷阱(NETs)介导的。NETs在PDAC中升高,并且几乎与肿瘤进展的每个阶段有关。我们研究了NETs对PDAC、TME和免疫治疗反应的影响。C57BJ6或PAD4-/-小鼠接受对照、DNase、抗pd -1治疗或DNase和抗pd -1治疗,建立原位PDAC模型。评估NET标记物、纤维化和TME免疫谱。人类PDAC患者也被评估NETs和肿瘤浸润T细胞的水平。循环NET标记物与PDAC患者肿瘤内CD8+细胞相关。NET水平高的患者也经历了更多的术后并发症。NET抑制小鼠肿瘤生长,提高生存率。NET缺陷小鼠的胶原和基质金属蛋白酶(MMP)基因表达降低,肿瘤内胶原沉积减少。此外,发现TCF1+PD-1+CD44+CD8+祖T细胞(对免疫治疗有反应的T细胞亚群)增加。当抗pd -1治疗与NET抑制联合使用时,这些变化导致肿瘤负荷进一步减轻和生存期延长。NETs影响细胞外基质重塑和T细胞对PDAC的反应,从而对抗pd -1治疗产生显著反应。这些发现支持免疫治疗和NET抑制联合治疗PDAC患者。
{"title":"<b>Neutrophil extracellular trap inhibition revitalizes PDAC immunotherapy responsiveness via reduced fibrosis and TCF1</b><sup><b>+</b></sup><b>CD8</b><sup><b>+</b></sup> <b>progenitor T-cell expansion</b>.","authors":"Britney Niemann, Abby Ivey, Quinn Hopen, Duaa Dakhlallah, Kathleen Brundage, Nicole Mihalik, Timothy Eubank, Brian A Boone","doi":"10.1080/2162402X.2025.2601394","DOIUrl":"https://doi.org/10.1080/2162402X.2025.2601394","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor survival. The immunosuppressive tumor microenvironment (TME) drives resistance to therapy, including immunotherapy. This may be, in part, mediated by the formation of neutrophil extracellular traps (NETs), formed when neutrophils release their intracellular contents. NETs are elevated in PDAC and are associated with nearly every stage of tumor progression. We investigated the influence of NETs on the PDAC TME and immunotherapy response. An orthotopic PDAC model was utilized in C57BJ6 or PAD4<sup>-/-</sup> mice receiving one of the following: control, DNase, anti-PD-1 therapy, or DNase and anti-PD-1. NET markers, fibrosis, and the TME immune profile were evaluated. Human PDAC patients were also evaluated for levels of NETs and tumor-infiltrating T cells. Circulating NET markers correlated with intra-tumoral CD8<sup>+</sup> cells in PDAC patients. Patients with high NET levels also experienced more post-operative complications. NET inhibition in mice reduced tumor growth and enhanced survival. Decreased expression of collagen and matrix metalloproteinase (MMP) genes, as well as reduced intra-tumoral collagen deposition were found in NET deficient mice. Additionally, an increase in TCF1<sup>+</sup>PD-1<sup>+</sup>CD44<sup>+</sup>CD8<sup>+</sup> progenitor T cells, a subpopulation of T cells responsive to immunotherapy, were identified. These changes resulted in further tumor burden reduction and prolonged survival when anti-PD-1 therapy was given in conjunction with NET inhibition. NETs influence extracellular matrix remodeling and the T cell response to PDAC, allowing for a significant response to anti-PD-1 therapy. These findings support the combination therapy of immunotherapy and NET inhibition in patients with PDAC.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2601394"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145783467","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}
引用次数: 0
Anti-FAP CAR-NK cells as a novel targeted therapy against cervical cancer and cancer-associated fibroblasts. 抗fap CAR-NK细胞作为一种新的靶向治疗宫颈癌和癌症相关成纤维细胞。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-09-17 DOI: 10.1080/2162402X.2025.2556714
Robert Polten, Ivana Kutle, Jan Lennart Stalp, Jens Hachenberg, Ann-Kathrin Seyda, Lavinia Neubert, Jan C Kamp, Constantin von Kaisenberg, Dirk Schaudien, Peter Hillemanns, Rüdiger Klapdor, Michael Morgan, Axel Schambach

The tumor microenvironment (TME) has a central role in many cancers, particularly by fostering an immunosuppressive milieu. Chimeric antigen receptor (CAR)-based immunotherapy displays a promising strategy to re-direct immune cells toward specific antigens, thereby inducing targeted cytotoxicity. The fibroblast activation protein (FAP) is overexpressed in various cancer types and has shown promise in CAR-based therapies. However, its application in gynecological cancers remains unexplored. This study evaluates the efficacy of anti-FAP CAR-NK cells as a targeted immunotherapy for cervical cancer and cancer-associated fibroblasts (CAFs). FAP expression was quantified on cervical cancer cell lines, primary cervical cancer tissues, and cells isolated from these tissues. Alpharetroviral SIN vectors were used to transduce NK-92 cells and primary cord blood-derived NK cells with 3rd-generation anti-FAP CARs. Immunohistochemistry and flow cytometry revealed high FAP expression on CaSki cells, cervical cancer tissues, and primary cervical CAFs. In 2D co-cultures with FAP-positive target cells, anti-FAP CAR-NK cells exhibited significantly enhanced cytotoxicity and elevated degranulation compared to control NK cells, with no observed effects against FAP-negative target cells. Primary NK cells revealed high cytotoxicity against cervical cancer cells with a high release of cytolytic enzymes. Anti-FAP CAR-NK cells also showed efficient elimination of cervical cancer cells and CAFs in 3D tumor spheroid models. These findings underscore the potential of anti-FAP CAR-NK cells as a potent therapeutic approach for cervical cancer and suggest broader applicability in diseases characterized by high FAP expression.

肿瘤微环境(TME)在许多癌症中具有中心作用,特别是通过培养免疫抑制环境。基于嵌合抗原受体(CAR)的免疫疗法显示出一种很有前途的策略,可以将免疫细胞重新导向特定抗原,从而诱导靶向细胞毒性。成纤维细胞激活蛋白(FAP)在各种癌症类型中过度表达,并在基于car的治疗中显示出前景。然而,它在妇科癌症中的应用仍未被探索。本研究评估了抗fap CAR-NK细胞作为宫颈癌和癌症相关成纤维细胞(CAFs)靶向免疫治疗的疗效。FAP在宫颈癌细胞系、原发性宫颈癌组织和从这些组织中分离的细胞中的表达进行了量化。用α -逆转录病毒SIN载体转染NK-92细胞和原代脐带血NK细胞,转染第3代抗fap CARs。免疫组织化学和流式细胞术显示FAP在CaSki细胞、宫颈癌组织和原发性宫颈CAFs中高表达。在与fap阳性靶细胞共培养的2D中,与对照NK细胞相比,抗fap CAR-NK细胞表现出显著增强的细胞毒性和更高的脱颗粒水平,而对fap阴性靶细胞没有观察到任何影响。原代NK细胞对宫颈癌细胞表现出高的细胞毒性,细胞溶解酶释放量高。在三维肿瘤球体模型中,抗fap CAR-NK细胞也显示出对宫颈癌细胞和caf的有效清除。这些发现强调了抗FAP CAR-NK细胞作为一种有效的宫颈癌治疗方法的潜力,并表明在FAP高表达的疾病中具有更广泛的适用性。
{"title":"Anti-FAP CAR-NK cells as a novel targeted therapy against cervical cancer and cancer-associated fibroblasts.","authors":"Robert Polten, Ivana Kutle, Jan Lennart Stalp, Jens Hachenberg, Ann-Kathrin Seyda, Lavinia Neubert, Jan C Kamp, Constantin von Kaisenberg, Dirk Schaudien, Peter Hillemanns, Rüdiger Klapdor, Michael Morgan, Axel Schambach","doi":"10.1080/2162402X.2025.2556714","DOIUrl":"10.1080/2162402X.2025.2556714","url":null,"abstract":"<p><p>The tumor microenvironment (TME) has a central role in many cancers, particularly by fostering an immunosuppressive milieu. Chimeric antigen receptor (CAR)-based immunotherapy displays a promising strategy to re-direct immune cells toward specific antigens, thereby inducing targeted cytotoxicity. The fibroblast activation protein (FAP) is overexpressed in various cancer types and has shown promise in CAR-based therapies. However, its application in gynecological cancers remains unexplored. This study evaluates the efficacy of anti-FAP CAR-NK cells as a targeted immunotherapy for cervical cancer and cancer-associated fibroblasts (CAFs). FAP expression was quantified on cervical cancer cell lines, primary cervical cancer tissues, and cells isolated from these tissues. Alpharetroviral SIN vectors were used to transduce NK-92 cells and primary cord blood-derived NK cells with 3<sup>rd</sup>-generation anti-FAP CARs. Immunohistochemistry and flow cytometry revealed high FAP expression on CaSki cells, cervical cancer tissues, and primary cervical CAFs. In 2D co-cultures with FAP-positive target cells, anti-FAP CAR-NK cells exhibited significantly enhanced cytotoxicity and elevated degranulation compared to control NK cells, with no observed effects against FAP-negative target cells. Primary NK cells revealed high cytotoxicity against cervical cancer cells with a high release of cytolytic enzymes. Anti-FAP CAR-NK cells also showed efficient elimination of cervical cancer cells and CAFs in 3D tumor spheroid models. These findings underscore the potential of anti-FAP CAR-NK cells as a potent therapeutic approach for cervical cancer and suggest broader applicability in diseases characterized by high FAP expression.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2556714"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12716053/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145076333","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}
引用次数: 0
The human STING agonist E7766 induces immunogenic tumor clearance, independent of tumor-intrinsic STING expression in the KRASG12D/+ Trp53-/- murine model of sarcoma. 在KRASG12D/+ Trp53-/-小鼠肉瘤模型中,人STING激动剂E7766诱导免疫原性肿瘤清除,独立于肿瘤固有的STING表达。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-07-22 DOI: 10.1080/2162402X.2025.2534912
Karys M Hildebrand, Kurt N Hildebrand, Carolina Salazar Arcila, Kayla Marritt, Jahanara Rajwani, Golpira Elmi Assadzadeh, Antoine Dufour, Frank R Jirik, Michael J Monument

Soft tissue sarcomas (STS) are aggressive high-fatality cancers that affect children and adults. Most STS subtypes harbor an immunosuppressive tumor microenvironment (TME) and respond poorly to immunotherapy. Therapies capable of dismantling the immunosuppressive TME are needed to improve sensitivity to emerging immunotherapies. Activation of the Stimulator of INterferon Genes (STING) pathway has shown promising anti-tumor effects in preclinical models of carcinoma, but evaluations in sarcoma are lacking. Herein, we sought to examine the immune modulation and therapeutic efficacy of three translational small molecule STING agonists in an immunologically cold model of STS. Three classes of STING agonists, ML RR-S2 CDA, MSA-2, and E7766 were evaluated in an orthotopic KrasG12D/+ Trp53-/- model of STS. Dose titration survival studies, cytokine serology, and tumor immune phenotyping were used to examine STING agonist efficacy following intra-tumoral treatment. All STING agonists significantly increased survival time, however, only E7766 resulted in durable tumor clearance, inducing CD8+ T-cell infiltration and activated lymphocyte transcriptomic signatures in the TME. Antibody depletion was used to assess the dependency of treatment responses on CD8+ T-cells, showing that in their absence, tumor clearance did not occur following E7766 therapy. Using STING deficient mice, and CRISPR/Cas9 gene editing, we demonstrated that STS clearance following STING therapy was dependent on host STING and not tumor-intrinsic STING pathway functionality. E7766 represents a promising candidate able to remodel the TME of murine STS tumors toward an inflamed phenotype independent of tumor-intrinsic STING functionality, and should be considered for potential translation in STS treatment.

软组织肉瘤(STS)是侵袭性高致死率的癌症,影响儿童和成人。大多数STS亚型具有免疫抑制肿瘤微环境(TME),对免疫治疗反应较差。需要能够拆除免疫抑制TME的疗法来提高对新兴免疫疗法的敏感性。干扰素基因刺激因子(STING)通路的激活在临床前肿瘤模型中显示出有希望的抗肿瘤作用,但在肉瘤中的评估缺乏。在此,我们试图在STS免疫冷模型中检测三种翻译小分子STING激动剂的免疫调节和治疗效果。在原位KrasG12D/+ Trp53-/-模型中对3类STING激动剂ML RR-S2 CDA、MSA-2和E7766进行评价。使用剂量滴定生存研究、细胞因子血清学和肿瘤免疫表型来检查肿瘤内治疗后STING激动剂的疗效。所有的STING激动剂都能显著延长生存时间,然而,只有E7766能持久清除肿瘤,诱导CD8+ t细胞浸润,激活TME中的淋巴细胞转录组特征。抗体消耗被用来评估治疗反应对CD8+ t细胞的依赖性,显示在没有CD8+ t细胞的情况下,E7766治疗后没有发生肿瘤清除。通过使用STING缺陷小鼠和CRISPR/Cas9基因编辑,我们证明了STING治疗后的STS清除依赖于宿主STING,而不是肿瘤固有的STING通路功能。E7766是一种有希望的候选基因,能够将小鼠STS肿瘤的TME重塑为一种独立于肿瘤固有STING功能的炎症表型,应该考虑在STS治疗中进行潜在的翻译。
{"title":"The human STING agonist E7766 induces immunogenic tumor clearance, independent of tumor-intrinsic STING expression in the <i>KRAS<sup>G12D/+</sup> Trp53<sup>-/-</sup></i> murine model of sarcoma.","authors":"Karys M Hildebrand, Kurt N Hildebrand, Carolina Salazar Arcila, Kayla Marritt, Jahanara Rajwani, Golpira Elmi Assadzadeh, Antoine Dufour, Frank R Jirik, Michael J Monument","doi":"10.1080/2162402X.2025.2534912","DOIUrl":"10.1080/2162402X.2025.2534912","url":null,"abstract":"<p><p>Soft tissue sarcomas (STS) are aggressive high-fatality cancers that affect children and adults. Most STS subtypes harbor an immunosuppressive tumor microenvironment (TME) and respond poorly to immunotherapy. Therapies capable of dismantling the immunosuppressive TME are needed to improve sensitivity to emerging immunotherapies. Activation of the Stimulator of INterferon Genes (STING) pathway has shown promising anti-tumor effects in preclinical models of carcinoma, but evaluations in sarcoma are lacking. Herein, we sought to examine the immune modulation and therapeutic efficacy of three translational small molecule STING agonists in an immunologically cold model of STS. Three classes of STING agonists, ML RR-S2 CDA, MSA-2, and E7766 were evaluated in an orthotopic KrasG12D/+ Trp53-/- model of STS. Dose titration survival studies, cytokine serology, and tumor immune phenotyping were used to examine STING agonist efficacy following intra-tumoral treatment. All STING agonists significantly increased survival time, however, only E7766 resulted in durable tumor clearance, inducing CD8+ T-cell infiltration and activated lymphocyte transcriptomic signatures in the TME. Antibody depletion was used to assess the dependency of treatment responses on CD8+ T-cells, showing that in their absence, tumor clearance did not occur following E7766 therapy. Using STING deficient mice, and CRISPR/Cas9 gene editing, we demonstrated that STS clearance following STING therapy was dependent on host STING and not tumor-intrinsic STING pathway functionality. E7766 represents a promising candidate able to remodel the TME of murine STS tumors toward an inflamed phenotype independent of tumor-intrinsic STING functionality, and should be considered for potential translation in STS treatment.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2534912"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12296111/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144683485","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}
引用次数: 0
A versatile silica nanoparticle platform for induction of T cell responses - applied for therapeutic vaccination against HPV16 E6/E7-positive tumors in MHC-humanized mice. 用于诱导T细胞反应的多功能二氧化硅纳米颗粒平台-应用于mhc人源化小鼠中针对HPV16 E6/ e7阳性肿瘤的治疗性疫苗接种。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-08-25 DOI: 10.1080/2162402X.2025.2548002
Sebastian Kruse, Lia T Fricke, Samantha Zottnick, Ann-Katrin Schlosser, Agnieszka K Grabowska, Eva Feidt, Philipp Uhl, Ellen Junglas, Jonas D Förster, Josephine Blersch, Philip Denner, Manina Günter, Stella E Autenrieth, Eugenio Fava, Walter Mier, Armin Kübelbeck, Angelika B Riemer

Therapeutic vaccines represent a promising treatment option for (pre)cancerous lesions, such as human papillomavirus-induced malignancies. They act via administration of tumor-specific antigens, leading to induction of antigen-specific cytotoxic T cell responses. However, vaccination efficiency is often limited when the antigen is administered alone, due to antigen instability and inefficient uptake by antigen-presenting cells (APCs). To address these limitations, nanoparticle-based vaccine delivery systems are currently under investigation. Here, we present a novel silica nanoparticle (SiNP)-based vaccine delivery platform that can be applied for the treatment of various diseases and cancer types. We show that surface-functionalized SiNPs are non-cytotoxic and quickly taken up by APCs. Incorporation of a linker/solubilizer sequence N-terminal of the epitope allows attachment of peptides regardless of their solubility as well as efficient processing and surface presentation by APCs. Whole-body distribution studies confirmed retention of the antigen at the injection site and decelerated excretion when connected to SiNPs. Furthermore, treatment with SiNPs, especially when combined with the adjuvant poly(I:C), resulted in activation of dendritic cells capable of priming CD8+ T cells. In C57BL/6 and MHC-humanized A2.DR1 mice, the SiNP-based vaccinations induced epitope-specific CD8+ T cells. Moreover, they exhibited anti-tumor activity and provided a survival benefit in a tumor model using HPV16 E6/E7-expressing PAP-A2 cells. Thus, the novel SiNP platform represents a promising new vehicle for therapeutic vaccine delivery.

治疗性疫苗对于(癌前)病变(如人乳头瘤病毒诱导的恶性肿瘤)是一种很有希望的治疗选择。它们通过给药肿瘤特异性抗原起作用,导致抗原特异性细胞毒性T细胞反应的诱导。然而,由于抗原不稳定和抗原呈递细胞(apc)摄取效率低下,单独给药时疫苗接种效率往往受到限制。为了解决这些限制,目前正在研究基于纳米颗粒的疫苗递送系统。在这里,我们提出了一种新的基于二氧化硅纳米颗粒(SiNP)的疫苗递送平台,可用于治疗各种疾病和癌症类型。我们发现,表面功能化的sinp是无细胞毒性的,并能迅速被apc吸收。结合连接/增溶剂序列的表位n端允许肽的附着,而不考虑其溶解度以及apc的有效处理和表面呈现。全身分布研究证实了抗原在注射部位的保留和与SiNPs连接时的排泄减慢。此外,用SiNPs处理,特别是当与佐剂poly(I:C)结合时,导致能够启动CD8+ T细胞的树突状细胞的激活。在C57BL/6和mhc人源化A2中。在DR1小鼠中,基于sinp的疫苗可诱导表位特异性CD8+ T细胞。此外,它们表现出抗肿瘤活性,并在使用表达HPV16 E6/ e7的PAP-A2细胞的肿瘤模型中提供生存益处。因此,新的SiNP平台代表了一种有希望的治疗性疫苗递送新载体。
{"title":"A versatile silica nanoparticle platform for induction of T cell responses - applied for therapeutic vaccination against HPV16 E6/E7-positive tumors in MHC-humanized mice.","authors":"Sebastian Kruse, Lia T Fricke, Samantha Zottnick, Ann-Katrin Schlosser, Agnieszka K Grabowska, Eva Feidt, Philipp Uhl, Ellen Junglas, Jonas D Förster, Josephine Blersch, Philip Denner, Manina Günter, Stella E Autenrieth, Eugenio Fava, Walter Mier, Armin Kübelbeck, Angelika B Riemer","doi":"10.1080/2162402X.2025.2548002","DOIUrl":"10.1080/2162402X.2025.2548002","url":null,"abstract":"<p><p>Therapeutic vaccines represent a promising treatment option for (pre)cancerous lesions, such as human papillomavirus-induced malignancies. They act via administration of tumor-specific antigens, leading to induction of antigen-specific cytotoxic T cell responses. However, vaccination efficiency is often limited when the antigen is administered alone, due to antigen instability and inefficient uptake by antigen-presenting cells (APCs). To address these limitations, nanoparticle-based vaccine delivery systems are currently under investigation. Here, we present a novel silica nanoparticle (SiNP)-based vaccine delivery platform that can be applied for the treatment of various diseases and cancer types. We show that surface-functionalized SiNPs are non-cytotoxic and quickly taken up by APCs. Incorporation of a linker/solubilizer sequence N-terminal of the epitope allows attachment of peptides regardless of their solubility as well as efficient processing and surface presentation by APCs. Whole-body distribution studies confirmed retention of the antigen at the injection site and decelerated excretion when connected to SiNPs. Furthermore, treatment with SiNPs, especially when combined with the adjuvant poly(I:C), resulted in activation of dendritic cells capable of priming CD8<sup>+</sup> T cells. In C57BL/6 and MHC-humanized A2.DR1 mice, the SiNP-based vaccinations induced epitope-specific CD8<sup>+</sup> T cells. Moreover, they exhibited anti-tumor activity and provided a survival benefit in a tumor model using HPV16 E6/E7-expressing PAP-A2 cells. Thus, the novel SiNP platform represents a promising new vehicle for therapeutic vaccine delivery.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2548002"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12382477/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145114577","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}
引用次数: 0
The pancreatic tumor microenvironment of treatment-naïve patients causes a functional shift in γδ T cells, impairing their anti-tumoral defense. treatment-naïve患者的胰腺肿瘤微环境导致γδ T细胞的功能改变,损害其抗肿瘤防御功能。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-02-13 DOI: 10.1080/2162402X.2025.2466301
Elena Lo Presti, Francesca Cupaioli, Daniela Scimeca, Elettra Unti, Vincenzo Di Martino, Rossella Daidone, Michele Amata, Nunzia Scibetta, Erinn Soucie, Serena Meraviglia, Juan Iovanna, Nelson Dusetti, Andrea De Gaetano, Ivan Merelli, Roberto Di Mitri

Pancreatic ductal adenocarcinoma (PDAC) presents a unique challenge for researchers due to its late diagnosis caused by vague symptoms and lack of early detection markers. Additionally, PDAC is characterized by an immunosuppressive microenvironment (TME), making it a difficult tumor to treat. While γδ T cells have shown potential for anti-tumor activity, conflicting studies exist regarding their effectiveness in pancreatic cancer. This study aims to explore the hypothesis that the PDAC TME hinders the anti-tumor capabilities of γδ T cells through blockade of cytotoxic functions. For this reason, we chose to enroll PDAC treatment-naive patients to avoid the possibility of therapy modifying the TME. By flow cytometry, our research findings indicate that the presence of γδ T cells among CD45+ cells in tumor tissue is lower compared to CD66+ cells, but higher than in blood. Circulating Vδ1 T cells exhibit a terminal effector memory phenotype (TEMRA) more than Vδ2 T cells. Interestingly, Vδ1 and Vδ2 T cells appear to be more prevalent at different stages of tumor development. In our in vitro culture using conditioned medium derived from Patient-derived organoids ;(PDOs), we observed a shift in expression markers in γδ T cells of healthy individuals toward an activation and exhaustion phenotype, as confirmed by scRNA-seq analysis extracted from a public database. A deeper understanding of γδ T cells in PDAC could be valuable for developing novel therapies aimed at mitigating the impact of the pancreatic tumor microenvironment on this cell population.

胰腺导管腺癌(PDAC)由于其症状模糊和缺乏早期检测标志物而导致诊断较晚,对研究人员提出了独特的挑战。此外,PDAC的特点是免疫抑制微环境(TME),使其成为一种难以治疗的肿瘤。虽然γδ T细胞已显示出潜在的抗肿瘤活性,但关于其在胰腺癌中的有效性存在相互矛盾的研究。本研究旨在探讨PDAC TME通过阻断细胞毒性功能来阻碍γδ T细胞抗肿瘤能力的假设。出于这个原因,我们选择招募PDAC治疗初期患者,以避免治疗改变TME的可能性。通过流式细胞术,我们的研究结果表明,肿瘤组织中CD45+细胞中γδ T细胞的存在低于CD66+细胞,但高于血液中。循环Vδ1 T细胞比Vδ2 T细胞表现出终端效应记忆表型(TEMRA)。有趣的是,Vδ1和Vδ2 T细胞似乎在肿瘤发展的不同阶段更为普遍。在我们的体外培养中,我们观察到健康个体的γδ T细胞中表达标记物向激活和耗竭表型转变,这一点得到了从公共数据库中提取的scRNA-seq分析的证实。对PDAC中γδ T细胞的深入了解可能对开发旨在减轻胰腺肿瘤微环境对该细胞群影响的新疗法有价值。
{"title":"The pancreatic tumor microenvironment of treatment-naïve patients causes a functional shift in γδ T cells, impairing their anti-tumoral defense.","authors":"Elena Lo Presti, Francesca Cupaioli, Daniela Scimeca, Elettra Unti, Vincenzo Di Martino, Rossella Daidone, Michele Amata, Nunzia Scibetta, Erinn Soucie, Serena Meraviglia, Juan Iovanna, Nelson Dusetti, Andrea De Gaetano, Ivan Merelli, Roberto Di Mitri","doi":"10.1080/2162402X.2025.2466301","DOIUrl":"10.1080/2162402X.2025.2466301","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) presents a unique challenge for researchers due to its late diagnosis caused by vague symptoms and lack of early detection markers. Additionally, PDAC is characterized by an immunosuppressive microenvironment (TME), making it a difficult tumor to treat. While γδ T cells have shown potential for anti-tumor activity, conflicting studies exist regarding their effectiveness in pancreatic cancer. This study aims to explore the hypothesis that the PDAC TME hinders the anti-tumor capabilities of γδ T cells through blockade of cytotoxic functions. For this reason, we chose to enroll PDAC treatment-naive patients to avoid the possibility of therapy modifying the TME. By flow cytometry, our research findings indicate that the presence of γδ T cells among CD45+ cells in tumor tissue is lower compared to CD66+ cells, but higher than in blood. Circulating Vδ1 T cells exhibit a terminal effector memory phenotype (TEMRA) more than Vδ2 T cells. Interestingly, Vδ1 and Vδ2 T cells appear to be more prevalent at different stages of tumor development. In our <i>in vitro</i> culture using conditioned medium derived from Patient-derived organoids ;(PDOs), we observed a shift in expression markers in γδ T cells of healthy individuals toward an activation and exhaustion phenotype, as confirmed by scRNA-seq analysis extracted from a public database. A deeper understanding of γδ T cells in PDAC could be valuable for developing novel therapies aimed at mitigating the impact of the pancreatic tumor microenvironment on this cell population.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2466301"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11834455/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143411294","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}
引用次数: 0
Correction. 修正。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2025-07-25 DOI: 10.1080/2162402X.2025.2538308
{"title":"Correction.","authors":"","doi":"10.1080/2162402X.2025.2538308","DOIUrl":"10.1080/2162402X.2025.2538308","url":null,"abstract":"","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2538308"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12309527/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144715162","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}
引用次数: 0
Hyperdifferentiated murine melanoma cells promote adaptive anti-tumor immunity but activate the immune checkpoint system. 高分化小鼠黑色素瘤细胞促进适应性抗肿瘤免疫,但激活免疫检查点系统。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-01 Epub Date: 2024-12-08 DOI: 10.1080/2162402X.2024.2437211
Yukie Ando, Yutaka Horiuchi, Sara Hatazawa, Momo Mataki, Akihiro Nakamura, Takashi Murakami

Accumulating evidence suggests that phenotype switching of cancer cells is essential for therapeutic resistance. However, the immunological characteristics of drug-induced phenotype-switching melanoma cells (PSMCs) are unknown. We investigated PSMC elimination by host immunity using hyperdifferentiated melanoma model cells derived from murine B16F10 melanoma cells. Exposure of B16F10 cells to staurosporine induced a hyperdifferentiated phenotype associated with transient drug tolerance. Staurosporine-induced hyperdifferentiated B16F10 (sB16F10) cells expressed calreticulin on their surface and were phagocytosed efficiently. Furthermore, the inoculation of mice with sB16F10 cells induced immune responses against tumor-derived antigens. Despite the immunogenicity of sB16F10 cells, they activated the PD-1/PD-L1 immune checkpoint system and strongly resisted T cell-mediated tumor destruction. However, in vivo treatment with immune checkpoint inhibitors successfully eliminated the tumor. Thus, hyperdifferentiated melanoma cells have conflicting immunological properties - enhanced immunogenicity and immune evasion. Inhibiting the ability of PSMCs to evade T cell-mediated elimination might lead to complete melanoma eradication.

越来越多的证据表明,癌细胞的表型转换对治疗耐药性至关重要。然而,药物诱导的表型转换黑色素瘤细胞(PSMCs)的免疫学特性尚不清楚。我们利用来源于小鼠B16F10黑色素瘤细胞的高分化黑色素瘤模型细胞,研究宿主免疫对PSMC的消除作用。B16F10细胞暴露于staurosporine诱导了与短暂药物耐受相关的高分化表型。staurosporine诱导的高分化B16F10 (sB16F10)细胞表面表达钙网蛋白,并被高效吞噬。此外,用sB16F10细胞接种小鼠可诱导针对肿瘤源性抗原的免疫应答。尽管sB16F10细胞具有免疫原性,但它们激活了PD-1/PD-L1免疫检查点系统,并强烈抵抗T细胞介导的肿瘤破坏。然而,免疫检查点抑制剂在体内治疗成功地消除了肿瘤。因此,高分化黑色素瘤细胞具有相互冲突的免疫特性-增强的免疫原性和免疫逃避。抑制PSMCs逃避T细胞介导消除的能力可能导致黑色素瘤的完全根除。
{"title":"Hyperdifferentiated murine melanoma cells promote adaptive anti-tumor immunity but activate the immune checkpoint system.","authors":"Yukie Ando, Yutaka Horiuchi, Sara Hatazawa, Momo Mataki, Akihiro Nakamura, Takashi Murakami","doi":"10.1080/2162402X.2024.2437211","DOIUrl":"10.1080/2162402X.2024.2437211","url":null,"abstract":"<p><p>Accumulating evidence suggests that phenotype switching of cancer cells is essential for therapeutic resistance. However, the immunological characteristics of drug-induced phenotype-switching melanoma cells (PSMCs) are unknown. We investigated PSMC elimination by host immunity using hyperdifferentiated melanoma model cells derived from murine B16F10 melanoma cells. Exposure of B16F10 cells to staurosporine induced a hyperdifferentiated phenotype associated with transient drug tolerance. Staurosporine-induced hyperdifferentiated B16F10 (sB16F10) cells expressed calreticulin on their surface and were phagocytosed efficiently. Furthermore, the inoculation of mice with sB16F10 cells induced immune responses against tumor-derived antigens. Despite the immunogenicity of sB16F10 cells, they activated the PD-1/PD-L1 immune checkpoint system and strongly resisted T cell-mediated tumor destruction. However, <i>in vivo</i> treatment with immune checkpoint inhibitors successfully eliminated the tumor. Thus, hyperdifferentiated melanoma cells have conflicting immunological properties - enhanced immunogenicity and immune evasion. Inhibiting the ability of PSMCs to evade T cell-mediated elimination might lead to complete melanoma eradication.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"14 1","pages":"2437211"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11633153/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142796568","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}
引用次数: 0
期刊
Oncoimmunology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1