首页 > 最新文献

Cancer Immunology, Immunotherapy最新文献

英文 中文
Impaired TGF-β signaling via AHNAK family mutations elicits an esophageal cancer subtype with sensitivities to genotoxic therapy and immunotherapy. 通过AHNAK家族突变导致的TGF-β信号传导受损会诱发一种对基因毒性疗法和免疫疗法敏感的食管癌亚型。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-09-05 DOI: 10.1007/s00262-024-03798-z
Zihang Mai, Luo Kongjia, Xinye Wang, Xiuying Xie, Lanlan Pang, Hong Yang, Jing Wen, Jianhua Fu

Background: Genome instability (GI) is a hallmark of esophageal squamous cell carcinoma (ESCC) while factors affecting GI remain unclear.

Methods: Here, we aimed to characterize genomic events representing specific mechanisms of GI based on 201 ESCC samples and validated our findings at the patient, single-cell and cancer cell-line levels, including a newly generated multi-omics dataset of the trial NCT04006041.

Results: A two-gene (AHNAK and AHNAK2) mutation signature was identified to define the "AHNAK1/2-mutant" cancer subtype. Single-cell-assisted multi-omics analysis showed that this subtype had a higher neoantigen load, active antigen presentation, and proficient CD8 + T cell infiltrations, which were validated at pan-cancer levels. Mechanistically, AHNAK1/2-mutant ESCC was characterized by impaired response of TGF-β and the inefficient alternative end-join repair (Alt-EJ) that might promote GI. Knockdown of AHNAK in ESCC cell lines resulted in more Alt-EJ events and increased sensitivities to cisplatin. Furthermore, this two-gene signature accurately predicted better responses to DNA-damaging therapy in various clinical settings (HR ≈ 0.25). The two-gene signature predicted higher pCR rates in ESCCs receiving neoadjuvant immunotherapy-involved treatment. Finally, a molecular classification scheme was built and outperformed established molecular typing models in the prognosis stratification of ESCC patients.

Conclusion: Our study extended our understanding of the AHNAK family in promoting GI and selecting treatment responders of ESCC.

背景:基因组不稳定性(GI)是食管鳞状细胞癌(ESCC)的一个特征,而影响GI的因素仍不清楚。方法:在此,我们以201份ESCC样本为基础,旨在描述代表GI特定机制的基因组事件的特征,并在患者、单细胞和癌细胞系水平验证我们的发现,包括NCT04006041试验中新生成的多组学数据集:结果:确定了一个双基因(AHNAK和AHNAK2)突变特征,从而定义了 "AHNAK1/2突变 "癌症亚型。单细胞辅助多组学分析表明,该亚型具有较高的新抗原负荷、活跃的抗原呈递和熟练的CD8 + T细胞浸润,这在泛癌水平上得到了验证。从机理上讲,AHNAK1/2突变型ESCC的特点是对TGF-β的反应受损和替代性末端连接修复(Alt-EJ)效率低下,这可能会促进GI。在ESCC细胞系中敲除AHNAK会导致更多的Alt-EJ事件,并增加对顺铂的敏感性。此外,这两个基因特征还能准确预测各种临床环境中对DNA损伤疗法的更好反应(HR≈0.25)。在接受新辅助免疫疗法治疗的 ESCC 中,该双基因特征预测了更高的 pCR 率。最后,我们建立了一个分子分类方案,该方案在ESCC患者预后分层方面优于已有的分子分型模型:我们的研究拓展了我们对AHNAK家族在促进GI和选择ESCC治疗应答者方面的认识。
{"title":"Impaired TGF-β signaling via AHNAK family mutations elicits an esophageal cancer subtype with sensitivities to genotoxic therapy and immunotherapy.","authors":"Zihang Mai, Luo Kongjia, Xinye Wang, Xiuying Xie, Lanlan Pang, Hong Yang, Jing Wen, Jianhua Fu","doi":"10.1007/s00262-024-03798-z","DOIUrl":"10.1007/s00262-024-03798-z","url":null,"abstract":"<p><strong>Background: </strong>Genome instability (GI) is a hallmark of esophageal squamous cell carcinoma (ESCC) while factors affecting GI remain unclear.</p><p><strong>Methods: </strong>Here, we aimed to characterize genomic events representing specific mechanisms of GI based on 201 ESCC samples and validated our findings at the patient, single-cell and cancer cell-line levels, including a newly generated multi-omics dataset of the trial NCT04006041.</p><p><strong>Results: </strong>A two-gene (AHNAK and AHNAK2) mutation signature was identified to define the \"AHNAK1/2-mutant\" cancer subtype. Single-cell-assisted multi-omics analysis showed that this subtype had a higher neoantigen load, active antigen presentation, and proficient CD8 + T cell infiltrations, which were validated at pan-cancer levels. Mechanistically, AHNAK1/2-mutant ESCC was characterized by impaired response of TGF-β and the inefficient alternative end-join repair (Alt-EJ) that might promote GI. Knockdown of AHNAK in ESCC cell lines resulted in more Alt-EJ events and increased sensitivities to cisplatin. Furthermore, this two-gene signature accurately predicted better responses to DNA-damaging therapy in various clinical settings (HR ≈ 0.25). The two-gene signature predicted higher pCR rates in ESCCs receiving neoadjuvant immunotherapy-involved treatment. Finally, a molecular classification scheme was built and outperformed established molecular typing models in the prognosis stratification of ESCC patients.</p><p><strong>Conclusion: </strong>Our study extended our understanding of the AHNAK family in promoting GI and selecting treatment responders of ESCC.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11377381/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131971","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
Distribution characteristics of immune infiltration and lymphovascular invasion in patients with breast cancer skin recurrence. 乳腺癌皮肤复发患者免疫浸润和淋巴管侵犯的分布特征
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-09-05 DOI: 10.1007/s00262-024-03783-6
Danyang Zhou, Mei Li, Wei Wu, Ying Wu, Qiaohong Nong, Shusen Wang, Ruoxi Hong

Background: To assess the distribution characteristics of immune infiltration and lymphovascular invasion in breast cancer skin recurrence patients.

Methods: We retrospectively analyzed the clinicopathological data of patients who underwent radical surgery for primary breast cancer and experienced skin recurrence between January 2001 and April 2019. Immune and lymphovascular biomarkers were quantified in primary breast cancers, skin lesions and visceral metastatic lesions. Differences in biomarkers distribution between matched tissues were statistically analyzed using the Wilcoxon signed-rank test and Kruskal-Wallis one-way ANOVA.

Results: A total of 71 female breast cancer patients were reviewed in this study. Our study found that the expression levels of various lymphocyte immune markers in primary tumor specimens were higher than those in skin recurrences. The expression of CD8, CD57 and CD31 in primary breast cancer was higher than those in the skin. Compared to visceral metastatic lesions, D2-40 was highly expressed in the skin, while CD8 tended to decrease. In the skin specimens, the expression of CD8 (P < 0.001), FOXP3 (P = 0.006) and CD68 (P < 0.001) in the intratumoral area was higher, while the expression of CD57 (P < 0.001) was higher in the peritumoral area. Analyzing specimens from the same patient at different time points of skin progression, it was found that the expression of peritumoral CD4 decreased (P = 0.044) as the disease progressed. The low expression of D2-40 and CD163 in the skin lesions suggested a decrease in DFS.

Conclusion: The immune microenvironment of breast cancer skin recurrence may be in a state of suppression, and this suppression may intensify with disease progression. The pattern of skin recurrence may be more inclined toward lymphatic invasion. Our study provides new insights into the biological behaviors of this disease and its response to immunotherapy.

背景:评估乳腺癌皮肤复发患者免疫浸润和淋巴管侵犯的分布特征:评估乳腺癌皮肤复发患者免疫浸润和淋巴管侵犯的分布特征:我们回顾性分析了 2001 年 1 月至 2019 年 4 月间接受原发性乳腺癌根治术并出现皮肤复发的患者的临床病理数据。对原发性乳腺癌、皮肤病变和内脏转移病变中的免疫和淋巴管生物标志物进行了量化。采用Wilcoxon符号秩检验和Kruskal-Wallis单因素方差分析对匹配组织间的生物标志物分布差异进行统计分析:本研究共回顾性分析了71例女性乳腺癌患者。研究发现,原发肿瘤标本中各种淋巴细胞免疫标志物的表达水平高于皮肤复发标本。CD8、CD57和CD31在原发性乳腺癌中的表达高于皮肤中的表达。与内脏转移病灶相比,D2-40在皮肤中的表达量较高,而CD8则呈下降趋势。在皮肤标本中,CD8(P 结论:CD8在皮肤标本中的表达高于CD31在皮肤标本中的表达:乳腺癌皮肤复发的免疫微环境可能处于抑制状态,这种抑制可能会随着疾病的进展而加剧。皮肤复发的模式可能更倾向于淋巴侵袭。我们的研究为了解这种疾病的生物学行为及其对免疫疗法的反应提供了新的视角。
{"title":"Distribution characteristics of immune infiltration and lymphovascular invasion in patients with breast cancer skin recurrence.","authors":"Danyang Zhou, Mei Li, Wei Wu, Ying Wu, Qiaohong Nong, Shusen Wang, Ruoxi Hong","doi":"10.1007/s00262-024-03783-6","DOIUrl":"10.1007/s00262-024-03783-6","url":null,"abstract":"<p><strong>Background: </strong>To assess the distribution characteristics of immune infiltration and lymphovascular invasion in breast cancer skin recurrence patients.</p><p><strong>Methods: </strong>We retrospectively analyzed the clinicopathological data of patients who underwent radical surgery for primary breast cancer and experienced skin recurrence between January 2001 and April 2019. Immune and lymphovascular biomarkers were quantified in primary breast cancers, skin lesions and visceral metastatic lesions. Differences in biomarkers distribution between matched tissues were statistically analyzed using the Wilcoxon signed-rank test and Kruskal-Wallis one-way ANOVA.</p><p><strong>Results: </strong>A total of 71 female breast cancer patients were reviewed in this study. Our study found that the expression levels of various lymphocyte immune markers in primary tumor specimens were higher than those in skin recurrences. The expression of CD8, CD57 and CD31 in primary breast cancer was higher than those in the skin. Compared to visceral metastatic lesions, D2-40 was highly expressed in the skin, while CD8 tended to decrease. In the skin specimens, the expression of CD8 (P < 0.001), FOXP3 (P = 0.006) and CD68 (P < 0.001) in the intratumoral area was higher, while the expression of CD57 (P < 0.001) was higher in the peritumoral area. Analyzing specimens from the same patient at different time points of skin progression, it was found that the expression of peritumoral CD4 decreased (P = 0.044) as the disease progressed. The low expression of D2-40 and CD163 in the skin lesions suggested a decrease in DFS.</p><p><strong>Conclusion: </strong>The immune microenvironment of breast cancer skin recurrence may be in a state of suppression, and this suppression may intensify with disease progression. The pattern of skin recurrence may be more inclined toward lymphatic invasion. Our study provides new insights into the biological behaviors of this disease and its response to immunotherapy.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11377393/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131966","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 short-term efficacy of neoadjuvant SOX versus SOX plus immune checkpoint inhibitor following laparoscopic gastrectomy for locally advanced gastric cancer: a multicenter retrospective cohort study in China. 中国一项多中心回顾性队列研究:局部晚期胃癌腹腔镜胃切除术后新辅助SOX与SOX加免疫检查点抑制剂的短期疗效比较。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-09-05 DOI: 10.1007/s00262-024-03802-6
Hao Cui, Yongpu Yang, Liqiang Song, Zhen Yuan, Linde Sun, Jiajun Du, Yuyuan Lu, Ning Ning, Jianxin Cui, Yan Shi, Lin Chen, Bo Wei

Background: This study aims to evaluate the short-term efficacy for locally advanced gastric cancer (LAGC) who accepted laparoscopic gastrectomy (LG) after neoadjuvant SOX versus SOX plus immune checkpoint inhibitors (ICIs).

Methods: LAGC patients who accepted LG after neoadjuvant SOX (SOX-LG, n = 169) and SOX plus ICIs (SOX + ICIs-LG, n = 140) in three medical centers between Jan 2020 and Mar 2024 were analyzed. We compared the tumor regression, treatment-related adverse events (TRAEs), perioperative safety between two groups, and explored the risk factors of postoperative complications (POCs) for LG after neoadjuvant therapy.

Results: The baseline characteristics were comparable between two groups (P > 0.05). SOX + ICIs-LG group acquired a higher proportion of objective response (63.6% vs. 46.7%, P = 0.003), major pathological response (43.6% vs. 31.4%, P = 0.001), and pathological complete response (17.9% vs. 9.5%, P = 0.030). There were no significant differences in the TRAEs rates, operation time, R0 resection, retrieved lymph nodes, postoperative first flatus, and hospitalized days, overall and severe POCs between two groups (P > 0.05). Patients in the SOX-ICIs-LG group had lower estimated blood loss (EBL) compared with SOX-LG (P = 0.001). Multivariate analysis showed that more EBL (P = 0.003) and prognostic nutritional index (PNI) < 40 (P = 0.005) were independent risk factors of POCs for LG after neoadjuvant therapy.

Conclusion: Neoadjuvant SOX plus ICIs brings better tumor regression and similar TRAEs compared with SOX alone for LAGC. SOX + ICIs-LG is safe and feasible to conduct with less EBL. Surgeons should focus on the perioperative management to control POCs for patients with PNI < 40 and more EBL.

研究背景本研究旨在评估接受腹腔镜胃切除术(LG)的局部晚期胃癌(LAGC)患者在新辅助SOX与SOX加免疫检查点抑制剂(ICIs)治疗后的短期疗效:分析了2020年1月至2024年3月期间在三家医疗中心接受新辅助SOX(SOX-LG,n = 169)和SOX加ICIs(SOX + ICIs-LG,n = 140)后腹腔镜胃切除术(LG)的LAGC患者。我们比较了两组患者的肿瘤消退情况、治疗相关不良事件(TRAEs)、围手术期安全性,并探讨了新辅助治疗后LG术后并发症(POCs)的风险因素:结果:两组患者的基线特征具有可比性(P>0.05)。SOX+ICIs-LG组获得客观反应(63.6% vs. 46.7%,P = 0.003)、主要病理反应(43.6% vs. 31.4%,P = 0.001)和病理完全反应(17.9% vs. 9.5%,P = 0.030)的比例更高。两组患者的TRAEs率、手术时间、R0切除率、淋巴结取回率、术后首次排便率、住院天数、总体POCs和严重POCs无明显差异(P > 0.05)。与SOX-LG组相比,SOX-ICIs-LG组患者的估计失血量(EBL)更低(P = 0.001)。多变量分析显示,EBL(P = 0.003)和预后营养指数(PNI)更高:与单用 SOX 治疗 LAGC 相比,新辅助 SOX 加 ICIs 能带来更好的肿瘤消退效果和相似的 TRAEs。SOX+ICIs-LG安全可行,EBL较低。外科医生应重视围手术期管理,以控制 PNI 患者的 POCs
{"title":"The short-term efficacy of neoadjuvant SOX versus SOX plus immune checkpoint inhibitor following laparoscopic gastrectomy for locally advanced gastric cancer: a multicenter retrospective cohort study in China.","authors":"Hao Cui, Yongpu Yang, Liqiang Song, Zhen Yuan, Linde Sun, Jiajun Du, Yuyuan Lu, Ning Ning, Jianxin Cui, Yan Shi, Lin Chen, Bo Wei","doi":"10.1007/s00262-024-03802-6","DOIUrl":"10.1007/s00262-024-03802-6","url":null,"abstract":"<p><strong>Background: </strong>This study aims to evaluate the short-term efficacy for locally advanced gastric cancer (LAGC) who accepted laparoscopic gastrectomy (LG) after neoadjuvant SOX versus SOX plus immune checkpoint inhibitors (ICIs).</p><p><strong>Methods: </strong>LAGC patients who accepted LG after neoadjuvant SOX (SOX-LG, n = 169) and SOX plus ICIs (SOX + ICIs-LG, n = 140) in three medical centers between Jan 2020 and Mar 2024 were analyzed. We compared the tumor regression, treatment-related adverse events (TRAEs), perioperative safety between two groups, and explored the risk factors of postoperative complications (POCs) for LG after neoadjuvant therapy.</p><p><strong>Results: </strong>The baseline characteristics were comparable between two groups (P > 0.05). SOX + ICIs-LG group acquired a higher proportion of objective response (63.6% vs. 46.7%, P = 0.003), major pathological response (43.6% vs. 31.4%, P = 0.001), and pathological complete response (17.9% vs. 9.5%, P = 0.030). There were no significant differences in the TRAEs rates, operation time, R0 resection, retrieved lymph nodes, postoperative first flatus, and hospitalized days, overall and severe POCs between two groups (P > 0.05). Patients in the SOX-ICIs-LG group had lower estimated blood loss (EBL) compared with SOX-LG (P = 0.001). Multivariate analysis showed that more EBL (P = 0.003) and prognostic nutritional index (PNI) < 40 (P = 0.005) were independent risk factors of POCs for LG after neoadjuvant therapy.</p><p><strong>Conclusion: </strong>Neoadjuvant SOX plus ICIs brings better tumor regression and similar TRAEs compared with SOX alone for LAGC. SOX + ICIs-LG is safe and feasible to conduct with less EBL. Surgeons should focus on the perioperative management to control POCs for patients with PNI < 40 and more EBL.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11377379/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131977","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
Oncolytic virotherapy augments self-maintaining natural killer cell line cytotoxicity against neuroblastoma. 肿瘤溶解病毒疗法增强了自维持自然杀伤细胞系对神经母细胞瘤的细胞毒性。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-09-05 DOI: 10.1007/s00262-024-03818-y
Colin H Quinn, Janet R Julson, Hooper R Markert, Nazia Nazam, Swatika Butey, Jerry E Stewart, Jennifer C Coleman, James M Markert, Jianmei W Leavenworth, Elizabeth A Beierle

Background: Neuroblastoma is the most common extracranial solid tumor in children and accounts for 15% of pediatric cancer related deaths. Targeting neuroblastoma with immunotherapies has proven challenging due to a paucity of immune cells in the tumor microenvironment and the release of immunosuppressive cytokines by neuroblastoma tumor cells. We hypothesized that combining an oncolytic Herpes Simplex Virus (oHSV) with natural killer (NK) cells might overcome these barriers and incite tumor cell death.

Methods: We utilized MYCN amplified and non-amplified neuroblastoma cell lines, the IL-12 expressing oHSV, M002, and the human NK cell line, NK-92 MI. We assessed the cytotoxicity of NK cells against neuroblastoma with and without M002 infection, the effects of M002 on NK cell priming, and the impact of M002 and priming on the migratory capacity and CD107a expression of NK cells. To test clinical applicability, we then investigated the effects of M002 and NK cells on neuroblastoma in vivo.

Results: NK cells were more attracted to neuroblastoma cells that were infected with M002. There was an increase in neuroblastoma cell death with the combination treatment of M002 and NK cells both in vitro and in vivo. Priming the NK cells enhanced their cytotoxicity, migratory capacity and CD107a expression.

Conclusions: To the best of our knowledge, these investigations are the first to demonstrate the effects of an oncolytic virus combined with self-maintaining NK cells in neuroblastoma and the priming effect of neuroblastoma on NK cells. The current studies provide a deeper understanding of the relation between NK cells and neuroblastoma and these data suggest that oHSV increases NK cell cytotoxicity towards neuroblastoma.

背景:神经母细胞瘤是儿童最常见的颅外实体瘤,占儿童癌症相关死亡人数的 15%。由于肿瘤微环境中免疫细胞稀少以及神经母细胞瘤肿瘤细胞释放免疫抑制细胞因子,用免疫疗法靶向神经母细胞瘤已被证明具有挑战性。我们假设,将溶瘤性单纯疱疹病毒(oHSV)与自然杀伤细胞(NK)相结合,可能会克服这些障碍并诱导肿瘤细胞死亡:我们利用了 MYCN 扩增和非扩增的神经母细胞瘤细胞系、表达 IL-12 的 oHSV M002 和人类 NK 细胞系 NK-92 MI。我们评估了感染和未感染 M002 的 NK 细胞对神经母细胞瘤的细胞毒性、M002 对 NK 细胞引物的影响,以及 M002 和引物对 NK 细胞迁移能力和 CD107a 表达的影响。为了测试临床应用性,我们随后研究了M002和NK细胞对体内神经母细胞瘤的影响:结果:NK细胞更容易被感染了M002的神经母细胞瘤细胞吸引。M002和NK细胞在体外和体内的联合处理增加了神经母细胞瘤细胞的死亡。对NK细胞进行引物处理可增强其细胞毒性、迁移能力和CD107a表达:据我们所知,这些研究首次证明了溶瘤病毒与自我维持的 NK 细胞结合对神经母细胞瘤的影响,以及神经母细胞瘤对 NK 细胞的引诱作用。目前的研究加深了人们对 NK 细胞与神经母细胞瘤之间关系的理解,这些数据表明 oHSV 增加了 NK 细胞对神经母细胞瘤的细胞毒性。
{"title":"Oncolytic virotherapy augments self-maintaining natural killer cell line cytotoxicity against neuroblastoma.","authors":"Colin H Quinn, Janet R Julson, Hooper R Markert, Nazia Nazam, Swatika Butey, Jerry E Stewart, Jennifer C Coleman, James M Markert, Jianmei W Leavenworth, Elizabeth A Beierle","doi":"10.1007/s00262-024-03818-y","DOIUrl":"10.1007/s00262-024-03818-y","url":null,"abstract":"<p><strong>Background: </strong>Neuroblastoma is the most common extracranial solid tumor in children and accounts for 15% of pediatric cancer related deaths. Targeting neuroblastoma with immunotherapies has proven challenging due to a paucity of immune cells in the tumor microenvironment and the release of immunosuppressive cytokines by neuroblastoma tumor cells. We hypothesized that combining an oncolytic Herpes Simplex Virus (oHSV) with natural killer (NK) cells might overcome these barriers and incite tumor cell death.</p><p><strong>Methods: </strong>We utilized MYCN amplified and non-amplified neuroblastoma cell lines, the IL-12 expressing oHSV, M002, and the human NK cell line, NK-92 MI. We assessed the cytotoxicity of NK cells against neuroblastoma with and without M002 infection, the effects of M002 on NK cell priming, and the impact of M002 and priming on the migratory capacity and CD107a expression of NK cells. To test clinical applicability, we then investigated the effects of M002 and NK cells on neuroblastoma in vivo.</p><p><strong>Results: </strong>NK cells were more attracted to neuroblastoma cells that were infected with M002. There was an increase in neuroblastoma cell death with the combination treatment of M002 and NK cells both in vitro and in vivo. Priming the NK cells enhanced their cytotoxicity, migratory capacity and CD107a expression.</p><p><strong>Conclusions: </strong>To the best of our knowledge, these investigations are the first to demonstrate the effects of an oncolytic virus combined with self-maintaining NK cells in neuroblastoma and the priming effect of neuroblastoma on NK cells. The current studies provide a deeper understanding of the relation between NK cells and neuroblastoma and these data suggest that oHSV increases NK cell cytotoxicity towards neuroblastoma.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11377387/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131973","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
Immune microenvironmental heterogeneity according to tumor DNA methylation phenotypes in microsatellite instability-high colorectal cancers. 根据微卫星不稳定性高的结直肠癌中肿瘤 DNA 甲基化表型的免疫微环境异质性。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-09-05 DOI: 10.1007/s00262-024-03805-3
Jung Ho Kim, Jiyun Hong, Ji Ae Lee, Minsun Jung, Eunwoo Choi, Nam-Yun Cho, Gyeong Hoon Kang, Sangwoo Kim

The detailed association between tumor DNA methylation, including CpG island methylation, and tumor immunity is poorly understood. CpG island methylator phenotype (CIMP) is observed typically in sporadic colorectal cancers (CRCs) with microsatellite instability-high (MSI-H). Here, we investigated the differential features of the tumor immune microenvironment according to CIMP status in MSI-H CRCs. CIMP-high (CIMP-H) or CIMP-low/negative (CIMP-L/0) status was determined using MethyLight assay in 133 MSI-H CRCs. All MSI-H CRCs were subjected to digital pathology-based quantification of CD3 + /CD8 + /CD4 + /FoxP3 + /CD68 + /CD204 + /CD177 + tumor-infiltrating immune cells using whole-slide immunohistochemistry. Programmed death-ligand 1 (PD-L1) immunohistochemistry was evaluated using the tumor proportion score (TPS) and combined positive score (CPS). Representative cases were analyzed using whole-exome and RNA-sequencing. In 133 MSI-H CRCs, significantly higher densities of CD8 + tumor-infiltrating lymphocytes (TILs) were observed in CIMP-H tumors compared with CIMP-L/0 tumors. PD-L1 TPS and CPS in CIMP-H tumors were higher than in CIMP-L/0 tumors. Next-generation sequencing revealed that, compared with CIMP-L/0 tumors, CIMP-H tumors had higher fractions of CD8 + T cells/cytotoxic lymphocytes, higher cytolytic activity scores, and activated immune-mediated cell killing pathways. In contrast to CIMP-L/0 tumors, most CIMP-H tumors were identified as consensus molecular subtype 1, an immunogenic transcriptomic subtype of CRC. However, there were no differences in tumor mutational burden (TMB) between CIMP-H and CIMP-L/0 tumors in MSI-H CRCs. In conclusion, CIMP-H is associated with abundant cytotoxic CD8 + TILs and PD-L1 overexpression independent of TMB in MSI-H CRCs, suggesting that CIMP-H tumors represent a typical immune-hot subtype and are optimal candidates for immunotherapy in MSI-H tumors.

人们对肿瘤 DNA 甲基化(包括 CpG 岛甲基化)与肿瘤免疫之间的详细关系知之甚少。CpG岛甲基化表型(CIMP)通常出现在微卫星不稳定性高(MSI-H)的散发性结直肠癌(CRC)中。在此,我们研究了微卫星不稳定性高(MSI-H)结直肠癌中根据 CIMP 状态不同的肿瘤免疫微环境特征。在133例MSI-H型CRC中使用MethyLight检测法确定CIMP-高(CIMP-H)或CIMP-低/阴性(CIMP-L/0)状态。所有 MSI-H CRC 均采用全切片免疫组织化学方法,对 CD3 + /CD8 + /CD4 + /FoxP3 + /CD68 + /CD204 + /CD177 + 肿瘤浸润免疫细胞进行基于数字病理学的定量分析。程序性死亡配体 1(PD-L1)免疫组化采用肿瘤比例评分(TPS)和联合阳性评分(CPS)进行评估。采用全外显子组和 RNA 序列对代表性病例进行了分析。在133例MSI-H型CRC中,与CIMP-L/0型肿瘤相比,CIMP-H型肿瘤中CD8 +肿瘤浸润淋巴细胞(TIL)的密度明显更高。CIMP-H肿瘤中的PD-L1 TPS和CPS高于CIMP-L/0肿瘤。下一代测序显示,与CIMP-L/0肿瘤相比,CIMP-H肿瘤的CD8 + T细胞/细胞毒性淋巴细胞比例更高,细胞溶解活性评分更高,免疫介导的细胞杀伤通路被激活。与 CIMP-L/0 肿瘤相比,大多数 CIMP-H 肿瘤被鉴定为共识分子亚型 1,即 CRC 的免疫原性转录组亚型。然而,在MSI-H型CRC中,CIMP-H和CIMP-L/0肿瘤的肿瘤突变负荷(TMB)并无差异。总之,在MSI-H型CRC中,CIMP-H与丰富的细胞毒性CD8 + TILs和PD-L1过表达相关,与TMB无关,这表明CIMP-H肿瘤代表了一种典型的免疫热亚型,是MSI-H肿瘤免疫疗法的最佳候选者。
{"title":"Immune microenvironmental heterogeneity according to tumor DNA methylation phenotypes in microsatellite instability-high colorectal cancers.","authors":"Jung Ho Kim, Jiyun Hong, Ji Ae Lee, Minsun Jung, Eunwoo Choi, Nam-Yun Cho, Gyeong Hoon Kang, Sangwoo Kim","doi":"10.1007/s00262-024-03805-3","DOIUrl":"10.1007/s00262-024-03805-3","url":null,"abstract":"<p><p>The detailed association between tumor DNA methylation, including CpG island methylation, and tumor immunity is poorly understood. CpG island methylator phenotype (CIMP) is observed typically in sporadic colorectal cancers (CRCs) with microsatellite instability-high (MSI-H). Here, we investigated the differential features of the tumor immune microenvironment according to CIMP status in MSI-H CRCs. CIMP-high (CIMP-H) or CIMP-low/negative (CIMP-L/0) status was determined using MethyLight assay in 133 MSI-H CRCs. All MSI-H CRCs were subjected to digital pathology-based quantification of CD3 + /CD8 + /CD4 + /FoxP3 + /CD68 + /CD204 + /CD177 + tumor-infiltrating immune cells using whole-slide immunohistochemistry. Programmed death-ligand 1 (PD-L1) immunohistochemistry was evaluated using the tumor proportion score (TPS) and combined positive score (CPS). Representative cases were analyzed using whole-exome and RNA-sequencing. In 133 MSI-H CRCs, significantly higher densities of CD8 + tumor-infiltrating lymphocytes (TILs) were observed in CIMP-H tumors compared with CIMP-L/0 tumors. PD-L1 TPS and CPS in CIMP-H tumors were higher than in CIMP-L/0 tumors. Next-generation sequencing revealed that, compared with CIMP-L/0 tumors, CIMP-H tumors had higher fractions of CD8 + T cells/cytotoxic lymphocytes, higher cytolytic activity scores, and activated immune-mediated cell killing pathways. In contrast to CIMP-L/0 tumors, most CIMP-H tumors were identified as consensus molecular subtype 1, an immunogenic transcriptomic subtype of CRC. However, there were no differences in tumor mutational burden (TMB) between CIMP-H and CIMP-L/0 tumors in MSI-H CRCs. In conclusion, CIMP-H is associated with abundant cytotoxic CD8 + TILs and PD-L1 overexpression independent of TMB in MSI-H CRCs, suggesting that CIMP-H tumors represent a typical immune-hot subtype and are optimal candidates for immunotherapy in MSI-H tumors.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11377388/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131969","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
Stat3-mediated Atg7 expression regulates anti-tumor immunity in mouse melanoma. Stat3 介导的 Atg7 表达调节小鼠黑色素瘤的抗肿瘤免疫。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-09-05 DOI: 10.1007/s00262-024-03804-4
Sarah M Zimmerman, Erin Suh, Sofia R Smith, George P Souroullas

Epigenetic modifications to DNA and chromatin control oncogenic and tumor-suppressive mechanisms in melanoma. Ezh2, the catalytic component of the Polycomb Repressive Complex 2 (PRC2), which mediates methylation of lysine 27 on histone 3 (H3K27me3), can regulate both melanoma initiation and progression. We previously found that mutant Ezh2Y641F interacts with the immune regulator Stat3 and together they affect anti-tumor immunity. However, given the numerous downstream targets and pathways affected by Ezh2, many mechanisms that determine its oncogenic activity remain largely unexplored. Using genetically engineered mouse models, we further investigated the role of pathways downstream of Ezh2 in melanoma carcinogenesis and identified significant enrichment in several autophagy signatures, along with increased expression of autophagy regulators, such as Atg7. In this study, we investigated the effect of Atg7 on melanoma growth and tumor immunity within the context of a wild-type or Ezh2Y641F epigenetic state. We found that the Atg7 locus is controlled by multiple Ezh2 and Stat3 binding sites, Atg7 expression is dependent on Stat3 expression, and that deletion of Atg7 slows down melanoma cell growth in vivo, but not in vitro. Atg7 deletion also results in increased CD8 + T cells in Ezh2Y641F melanomas and reduced myelosuppressive cell infiltration in the tumor microenvironment, particularly in Ezh2WT melanomas, suggesting a strong immune system contribution in the role of Atg7 in melanoma progression. These findings highlight the complex interplay between genetic mutations, epigenetic regulators, and autophagy in shaping tumor immunity in melanoma.

DNA和染色质的表观遗传修饰控制着黑色素瘤的致癌和抑瘤机制。Ezh2是多聚核抑制复合体2(PRC2)的催化元件,它介导组蛋白3上赖氨酸27的甲基化(H3K27me3),可调控黑色素瘤的发生和发展。我们以前曾发现,突变体Ezh2Y641F与免疫调节剂Stat3相互作用,共同影响抗肿瘤免疫。然而,由于受Ezh2影响的下游靶点和通路众多,决定其致癌活性的许多机制在很大程度上仍未被探索。利用基因工程小鼠模型,我们进一步研究了 Ezh2 下游通路在黑色素瘤癌变中的作用,发现了多个自噬特征的显著富集,以及 Atg7 等自噬调节因子的表达增加。在本研究中,我们研究了野生型或 Ezh2Y641F 表观遗传学状态下 Atg7 对黑色素瘤生长和肿瘤免疫的影响。我们发现,Atg7基因座受多个Ezh2和Stat3结合位点控制,Atg7的表达依赖于Stat3的表达。Atg7缺失还导致Ezh2Y641F黑色素瘤中CD8 + T细胞增加,肿瘤微环境中骨髓抑制细胞浸润减少,尤其是在Ezh2WT黑色素瘤中,这表明Atg7在黑色素瘤进展过程中的作用与免疫系统密切相关。这些发现突显了基因突变、表观遗传调控因子和自噬在黑色素瘤中影响肿瘤免疫的复杂相互作用。
{"title":"Stat3-mediated Atg7 expression regulates anti-tumor immunity in mouse melanoma.","authors":"Sarah M Zimmerman, Erin Suh, Sofia R Smith, George P Souroullas","doi":"10.1007/s00262-024-03804-4","DOIUrl":"10.1007/s00262-024-03804-4","url":null,"abstract":"<p><p>Epigenetic modifications to DNA and chromatin control oncogenic and tumor-suppressive mechanisms in melanoma. Ezh2, the catalytic component of the Polycomb Repressive Complex 2 (PRC2), which mediates methylation of lysine 27 on histone 3 (H3K27me3), can regulate both melanoma initiation and progression. We previously found that mutant Ezh2<sup>Y641F</sup> interacts with the immune regulator Stat3 and together they affect anti-tumor immunity. However, given the numerous downstream targets and pathways affected by Ezh2, many mechanisms that determine its oncogenic activity remain largely unexplored. Using genetically engineered mouse models, we further investigated the role of pathways downstream of Ezh2 in melanoma carcinogenesis and identified significant enrichment in several autophagy signatures, along with increased expression of autophagy regulators, such as Atg7. In this study, we investigated the effect of Atg7 on melanoma growth and tumor immunity within the context of a wild-type or Ezh2<sup>Y641F</sup> epigenetic state. We found that the Atg7 locus is controlled by multiple Ezh2 and Stat3 binding sites, Atg7 expression is dependent on Stat3 expression, and that deletion of Atg7 slows down melanoma cell growth in vivo, but not in vitro. Atg7 deletion also results in increased CD8 + T cells in Ezh2<sup>Y641F</sup> melanomas and reduced myelosuppressive cell infiltration in the tumor microenvironment, particularly in Ezh2<sup>WT</sup> melanomas, suggesting a strong immune system contribution in the role of Atg7 in melanoma progression. These findings highlight the complex interplay between genetic mutations, epigenetic regulators, and autophagy in shaping tumor immunity in melanoma.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11377374/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131976","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
Preclinical development of a novel CCR8/CTLA-4 bispecific antibody for cancer treatment by disrupting CTLA-4 signaling on CD8 T cells and specifically depleting tumor-resident Tregs. 新型 CCR8/CTLA-4 双特异性抗体的临床前开发,通过破坏 CD8 T 细胞上的 CTLA-4 信号传导和特异性消耗肿瘤驻留的 Tregs 来治疗癌症。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-08-09 DOI: 10.1007/s00262-024-03794-3
Cuicui Guo, Xiaodong Dai, Yulei Du, Xiumei Xiong, Xun Gui

Anti-CTLA-4 antibodies faced challenges due to frequent adverse events and limited efficacy, which spurred the exploration of next-generation CTLA-4 therapeutics to balance regulatory T cells (Tregs) depletion and CD8 T cells activation. CCR8, identified primarily on tumor-infiltrating Tregs, has become a target of interest due to the anti-tumor effects demonstrated by CCR8 antibody-mediated Tregs depletion. Single-cell RNA sequencing analysis reveals that CCR8-positive Tregs constitute a small subset, with concurrent expression of CCR8 and CTLA-4. Consequently, we proposed a novel bispecific antibody targeting CCR8 and CTLA-4 that had the potential to enhance T cell activation while selectively depleting intratumor Tregs. The candidate molecule 2MW4691 was developed in a tetravalent symmetric format, maintaining a strong binding affinity for CCR8 while exhibiting relatively weaker CTLA-4 binding. This selective binding ability allowed 2MW4691 to target and deplete tumor-infiltrating Tregs with higher specificity. In vitro assays verified the antibody's capacity for antibody-dependent cellular cytotoxicity (ADCC) to Tregs with high level of CTLA-4 expression, but not CD8 T cells with relatively low level of CTLA-4 on cell surface. Also, 2MW4691 inhibited the CTLA-4 pathway and enhanced T cell activation. The in vivo therapeutic efficacy of 2MW4691 was further demonstrated using hCCR8 or hCTLA-4 humanized mouse models and hCCR8/hCTLA-4 double knock-in mouse models. In cynomolgus monkeys, 2MW4691 was well-tolerated, exhibited the anticipated pharmacokinetic profile, and had a minimal impact on the peripheral T cell population. The promising preclinical results supported the further evaluation of 2MW4691 as a next-generation Treg-based therapeutics in clinical trials.

抗CTLA-4抗体因不良反应频发和疗效有限而面临挑战,这促使人们探索下一代CTLA-4疗法,以平衡调节性T细胞(Tregs)消耗和CD8 T细胞激活。CCR8主要在肿瘤浸润性Tregs上被发现,由于CCR8抗体介导的Tregs耗竭具有抗肿瘤作用,CCR8已成为人们关注的靶点。单细胞 RNA 测序分析表明,CCR8 阳性的 Tregs 构成了一个小的亚群,同时表达 CCR8 和 CTLA-4。因此,我们提出了一种靶向CCR8和CTLA-4的新型双特异性抗体,它有可能在增强T细胞活化的同时选择性地消耗肿瘤内的Tregs。候选分子 2MW4691 采用四价对称形式开发,与 CCR8 的结合亲和力很强,而与 CTLA-4 的结合力相对较弱。这种选择性结合能力使 2MW4691 能够以更高的特异性靶向并清除肿瘤浸润性集落细胞。体外实验验证了该抗体对高水平 CTLA-4 表达的 Tregs 的抗体依赖性细胞毒性(ADCC)能力,但对细胞表面 CTLA-4 水平相对较低的 CD8 T 细胞的抗体依赖性细胞毒性(ADCC)能力却没有验证。此外,2MW4691还能抑制CTLA-4通路,增强T细胞活化。使用 hCCR8 或 hCTLA-4 人源化小鼠模型和 hCCR8/hCTLA-4 双基因敲入小鼠模型进一步证实了 2MW4691 的体内疗效。在猴体内,2MW4691的耐受性良好,表现出预期的药代动力学特征,并且对外周T细胞群的影响极小。充满希望的临床前研究结果支持在临床试验中进一步评估 2MW4691 作为基于 Treg 的下一代疗法。
{"title":"Preclinical development of a novel CCR8/CTLA-4 bispecific antibody for cancer treatment by disrupting CTLA-4 signaling on CD8 T cells and specifically depleting tumor-resident Tregs.","authors":"Cuicui Guo, Xiaodong Dai, Yulei Du, Xiumei Xiong, Xun Gui","doi":"10.1007/s00262-024-03794-3","DOIUrl":"10.1007/s00262-024-03794-3","url":null,"abstract":"<p><p>Anti-CTLA-4 antibodies faced challenges due to frequent adverse events and limited efficacy, which spurred the exploration of next-generation CTLA-4 therapeutics to balance regulatory T cells (Tregs) depletion and CD8 T cells activation. CCR8, identified primarily on tumor-infiltrating Tregs, has become a target of interest due to the anti-tumor effects demonstrated by CCR8 antibody-mediated Tregs depletion. Single-cell RNA sequencing analysis reveals that CCR8-positive Tregs constitute a small subset, with concurrent expression of CCR8 and CTLA-4. Consequently, we proposed a novel bispecific antibody targeting CCR8 and CTLA-4 that had the potential to enhance T cell activation while selectively depleting intratumor Tregs. The candidate molecule 2MW4691 was developed in a tetravalent symmetric format, maintaining a strong binding affinity for CCR8 while exhibiting relatively weaker CTLA-4 binding. This selective binding ability allowed 2MW4691 to target and deplete tumor-infiltrating Tregs with higher specificity. In vitro assays verified the antibody's capacity for antibody-dependent cellular cytotoxicity (ADCC) to Tregs with high level of CTLA-4 expression, but not CD8 T cells with relatively low level of CTLA-4 on cell surface. Also, 2MW4691 inhibited the CTLA-4 pathway and enhanced T cell activation. The in vivo therapeutic efficacy of 2MW4691 was further demonstrated using hCCR8 or hCTLA-4 humanized mouse models and hCCR8/hCTLA-4 double knock-in mouse models. In cynomolgus monkeys, 2MW4691 was well-tolerated, exhibited the anticipated pharmacokinetic profile, and had a minimal impact on the peripheral T cell population. The promising preclinical results supported the further evaluation of 2MW4691 as a next-generation Treg-based therapeutics in clinical trials.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11315865/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911869","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
NKG2D-bispecific enhances NK and CD8+ T cell antitumor immunity. NKG2D 双特异性可增强 NK 和 CD8+ T 细胞的抗肿瘤免疫力。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-08-08 DOI: 10.1007/s00262-024-03795-2
Aurelie Herault, Judy Mak, Josefa de la Cruz-Chuh, Michael A Dillon, Diego Ellerman, MaryAnn Go, Ely Cosino, Robyn Clark, Emily Carson, Stacey Yeung, Melanie Pichery, Mylène Gador, Eugene Y Chiang, Jia Wu, Yuxin Liang, Zora Modrusan, Gautham Gampa, Jawahar Sudhamsu, Christopher C Kemball, Victoria Cheung, Thi Thu Thao Nguyen, Dhaya Seshasayee, Robert Piskol, Klara Totpal, Shang-Fan Yu, Genee Lee, Katherine R Kozak, Christoph Spiess, Kevin B Walsh

Background: Cancer immunotherapy approaches that elicit immune cell responses, including T and NK cells, have revolutionized the field of oncology. However, immunosuppressive mechanisms restrain immune cell activation within solid tumors so additional strategies to augment activity are required.

Methods: We identified the co-stimulatory receptor NKG2D as a target based on its expression on a large proportion of CD8+ tumor infiltrating lymphocytes (TILs) from breast cancer patient samples. Human and murine surrogate NKG2D co-stimulatory receptor-bispecifics (CRB) that bind NKG2D on NK and CD8+ T cells as well as HER2 on breast cancer cells (HER2-CRB) were developed as a proof of concept for targeting this signaling axis in vitro and in vivo.

Results: HER2-CRB enhanced NK cell activation and cytokine production when co-cultured with HER2 expressing breast cancer cell lines. HER2-CRB when combined with a T cell-dependent-bispecific (TDB) antibody that synthetically activates T cells by crosslinking CD3 to HER2 (HER2-TDB), enhanced T cell cytotoxicity, cytokine production and in vivo antitumor activity. A mouse surrogate HER2-CRB (mHER2-CRB) improved in vivo efficacy of HER2-TDB and augmented NK as well as T cell activation, cytokine production and effector CD8+ T cell differentiation.

Conclusion: We demonstrate that targeting NKG2D with bispecific antibodies (BsAbs) is an effective approach to augment NK and CD8+ T cell antitumor immune responses. Given the large number of ongoing clinical trials leveraging NK and T cells for cancer immunotherapy, NKG2D-bispecifics have broad combinatorial potential.

背景:激发免疫细胞(包括 T 细胞和 NK 细胞)反应的癌症免疫疗法为肿瘤学领域带来了革命性的变化。然而,免疫抑制机制抑制了实体瘤内免疫细胞的活化,因此需要更多增强活性的策略:方法:根据乳腺癌患者样本中大量 CD8+ 肿瘤浸润淋巴细胞(TILs)上 NKG2D 的表达,我们确定了共刺激受体 NKG2D 作为靶点。作为体外和体内靶向这一信号轴的概念验证,我们开发了与 NK 和 CD8+ T 细胞上的 NKG2D 以及乳腺癌细胞上的 HER2 结合的人类和小鼠代用 NKG2D 协同刺激受体双特异性药物(CRB):结果:与表达 HER2 的乳腺癌细胞系共同培养时,HER2-CRB 可增强 NK 细胞的活化和细胞因子的产生。HER2-CRB与T细胞依赖性双特异性(TDB)抗体(通过将CD3与HER2交联合成激活T细胞的抗体)结合,可增强T细胞的细胞毒性、细胞因子生成和体内抗肿瘤活性。小鼠代用 HER2-CRB(mHER2-CRB)提高了 HER2-TDB 的体内疗效,并增强了 NK 以及 T 细胞的活化、细胞因子的产生和效应 CD8+ T 细胞的分化:我们证明了用双特异性抗体(BsAbs)靶向 NKG2D 是增强 NK 和 CD8+ T 细胞抗肿瘤免疫反应的有效方法。鉴于目前正在进行大量利用 NK 和 T 细胞进行癌症免疫治疗的临床试验,NKG2D 双特异性抗体具有广泛的组合潜力。
{"title":"NKG2D-bispecific enhances NK and CD8+ T cell antitumor immunity.","authors":"Aurelie Herault, Judy Mak, Josefa de la Cruz-Chuh, Michael A Dillon, Diego Ellerman, MaryAnn Go, Ely Cosino, Robyn Clark, Emily Carson, Stacey Yeung, Melanie Pichery, Mylène Gador, Eugene Y Chiang, Jia Wu, Yuxin Liang, Zora Modrusan, Gautham Gampa, Jawahar Sudhamsu, Christopher C Kemball, Victoria Cheung, Thi Thu Thao Nguyen, Dhaya Seshasayee, Robert Piskol, Klara Totpal, Shang-Fan Yu, Genee Lee, Katherine R Kozak, Christoph Spiess, Kevin B Walsh","doi":"10.1007/s00262-024-03795-2","DOIUrl":"10.1007/s00262-024-03795-2","url":null,"abstract":"<p><strong>Background: </strong>Cancer immunotherapy approaches that elicit immune cell responses, including T and NK cells, have revolutionized the field of oncology. However, immunosuppressive mechanisms restrain immune cell activation within solid tumors so additional strategies to augment activity are required.</p><p><strong>Methods: </strong>We identified the co-stimulatory receptor NKG2D as a target based on its expression on a large proportion of CD8+ tumor infiltrating lymphocytes (TILs) from breast cancer patient samples. Human and murine surrogate NKG2D co-stimulatory receptor-bispecifics (CRB) that bind NKG2D on NK and CD8+ T cells as well as HER2 on breast cancer cells (HER2-CRB) were developed as a proof of concept for targeting this signaling axis in vitro and in vivo.</p><p><strong>Results: </strong>HER2-CRB enhanced NK cell activation and cytokine production when co-cultured with HER2 expressing breast cancer cell lines. HER2-CRB when combined with a T cell-dependent-bispecific (TDB) antibody that synthetically activates T cells by crosslinking CD3 to HER2 (HER2-TDB), enhanced T cell cytotoxicity, cytokine production and in vivo antitumor activity. A mouse surrogate HER2-CRB (mHER2-CRB) improved in vivo efficacy of HER2-TDB and augmented NK as well as T cell activation, cytokine production and effector CD8+ T cell differentiation.</p><p><strong>Conclusion: </strong>We demonstrate that targeting NKG2D with bispecific antibodies (BsAbs) is an effective approach to augment NK and CD8+ T cell antitumor immune responses. Given the large number of ongoing clinical trials leveraging NK and T cells for cancer immunotherapy, NKG2D-bispecifics have broad combinatorial potential.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11306676/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141901084","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
DNA hypo-methylation and expression of GBP4 induces T cell exhaustion in pancreatic cancer. DNA 低甲基化和 GBP4 的表达可诱导胰腺癌 T 细胞衰竭。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-08-07 DOI: 10.1007/s00262-024-03786-3
Yesiboli Tasiheng, Xuan Lin, Xu Wang, Xuan Zou, Yusheng Chen, Yu Yan, Mingjian Ma, Zhengjie Dai, Xu Wang, Xianjun Yu, He Cheng, Chen Liu

Immunotherapy for pancreatic ductal carcinoma (PDAC) remains disappointing due to the repressive tumor microenvironment and T cell exhaustion, in which the roles of interferon-stimulated genes were largely unknown. Here, we focused on a typical interferon-stimulated gene, GBP4, and investigated its potential diagnostic and therapeutic value in pancreatic cancer. Expression analysis on both local samples and public databases indicated that GBP4 was one of the most dominant GBP family members present in the PDAC microenvironment, and the expression level of GBP4 was negatively associated with patient survival. We then identified DNA hypo-methylation in regulatory regions of GBP4 in PDAC, and validated its regulatory role on GBP4 expression via performing targeted methylation using dCas9-SunTag-DNMAT3A-sgRNA-targeted methylation system on selected DNA locus. After that, we investigated the downstream functions of GBP4, and chemotaxis assays indicated that GBP4 overexpression significantly improved the infiltration of CD8+T cells, but also induced upregulation of immune checkpoint genes and T cell exhaustion. Lastly, in vitro T cell killing assays using primary organoids suggested that the PDAC samples with high level of GBP4 expression displayed significantly higher sensitivity to anti-PD-1 treatment. Taken together, our studies revealed the expression patterns and epigenetic regulatory mechanisms of GBP4 in pancreatic cancer and clarified the effects of GBP4 on T cell exhaustion and antitumor immunology.

胰腺导管癌(PDAC)的免疫疗法由于肿瘤微环境的抑制和T细胞的衰竭而仍然令人失望,其中干扰素刺激基因的作用在很大程度上是未知的。在此,我们聚焦于一个典型的干扰素刺激基因--GBP4,研究其在胰腺癌中的潜在诊断和治疗价值。对本地样本和公共数据库进行的表达分析表明,GBP4是PDAC微环境中最主要的GBP家族成员之一,而且GBP4的表达水平与患者生存期呈负相关。随后,我们发现了PDAC中GBP4调控区的DNA低甲基化,并通过使用dCas9-SunTag-DNMAT3A-sgRNA靶向甲基化系统对选定的DNA位点进行靶向甲基化,验证了其对GBP4表达的调控作用。随后,我们研究了 GBP4 的下游功能,趋化实验表明,GBP4 的过表达能显著改善 CD8+T 细胞的浸润,同时还能诱导免疫检查点基因的上调和 T 细胞衰竭。最后,利用原代器官组织进行的体外T细胞杀伤试验表明,GBP4高表达的PDAC样本对抗PD-1治疗的敏感性明显更高。综上所述,我们的研究揭示了GBP4在胰腺癌中的表达模式和表观遗传调控机制,并阐明了GBP4对T细胞衰竭和抗肿瘤免疫学的影响。
{"title":"DNA hypo-methylation and expression of GBP4 induces T cell exhaustion in pancreatic cancer.","authors":"Yesiboli Tasiheng, Xuan Lin, Xu Wang, Xuan Zou, Yusheng Chen, Yu Yan, Mingjian Ma, Zhengjie Dai, Xu Wang, Xianjun Yu, He Cheng, Chen Liu","doi":"10.1007/s00262-024-03786-3","DOIUrl":"10.1007/s00262-024-03786-3","url":null,"abstract":"<p><p>Immunotherapy for pancreatic ductal carcinoma (PDAC) remains disappointing due to the repressive tumor microenvironment and T cell exhaustion, in which the roles of interferon-stimulated genes were largely unknown. Here, we focused on a typical interferon-stimulated gene, GBP4, and investigated its potential diagnostic and therapeutic value in pancreatic cancer. Expression analysis on both local samples and public databases indicated that GBP4 was one of the most dominant GBP family members present in the PDAC microenvironment, and the expression level of GBP4 was negatively associated with patient survival. We then identified DNA hypo-methylation in regulatory regions of GBP4 in PDAC, and validated its regulatory role on GBP4 expression via performing targeted methylation using dCas9-SunTag-DNMAT3A-sgRNA-targeted methylation system on selected DNA locus. After that, we investigated the downstream functions of GBP4, and chemotaxis assays indicated that GBP4 overexpression significantly improved the infiltration of CD8<sup>+</sup>T cells, but also induced upregulation of immune checkpoint genes and T cell exhaustion. Lastly, in vitro T cell killing assays using primary organoids suggested that the PDAC samples with high level of GBP4 expression displayed significantly higher sensitivity to anti-PD-1 treatment. Taken together, our studies revealed the expression patterns and epigenetic regulatory mechanisms of GBP4 in pancreatic cancer and clarified the effects of GBP4 on T cell exhaustion and antitumor immunology.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11306721/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896862","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
Targeting PI3K-gamma in myeloid driven tumour immune suppression: a systematic review and meta-analysis of the preclinical literature. 在骨髓驱动的肿瘤免疫抑制中靶向 PI3K-gamma:临床前文献的系统回顾和荟萃分析。
IF 4.6 2区 医学 Q2 IMMUNOLOGY Pub Date : 2024-08-06 DOI: 10.1007/s00262-024-03779-2
Haonan Xu, Shannon Nicole Russell, Katherine Steiner, Eric O'Neill, Keaton Ian Jones

The intricate interplay between immune and stromal cells within the tumour microenvironment (TME) significantly influences tumour progression. Myeloid cells, including tumour-associated macrophages (TAMs), neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs), contribute to immune suppression in the TME (Nakamura and Smyth in Cell Mol Immunol 17(1):1-12 (2020). https://doi.org/10.1038/s41423-019-0306-1 ; DeNardo and Ruffell in Nat Rev Immunol 19(6):369-382 (2019). https://doi.org/10.1038/s41577-019-0127-6 ). This poses a significant challenge for novel immunotherapeutics that rely on host immunity to exert their effect. This systematic review explores the preclinical evidence surrounding the inhibition of phosphoinositide 3-kinase gamma (PI3Kγ) as a strategy to reverse myeloid-driven immune suppression in solid tumours. EMBASE, MEDLINE, and PubMed databases were searched on 6 October 2022 using keyword and subject heading terms to capture relevant studies. The studies, focusing on PI3Kγ inhibition in animal models, were subjected to predefined inclusion and exclusion criteria. Extracted data included tumour growth kinetics, survival endpoints, and immunological responses which were meta-analysed. PRISMA and MOOSE guidelines were followed. A total of 36 studies covering 73 animal models were included in the review and meta-analysis. Tumour models covered breast, colorectal, lung, skin, pancreas, brain, liver, prostate, head and neck, soft tissue, gastric, and oral cancer. The predominant PI3Kγ inhibitors were IPI-549 and TG100-115, demonstrating favourable specificity for the gamma isoform. Combination therapies, often involving chemotherapy, radiotherapy, immune checkpoint inhibitors, biological agents, or vaccines, were explored in 81% of studies. Analysis of tumour growth kinetics revealed a statistically significant though heterogeneous response to PI3Kγ monotherapy, whereas the tumour growth in combination treated groups were more consistently reduced. Survival analysis showed a pronounced increase in median overall survival with combination therapy. This systematic review provides a comprehensive analysis of preclinical studies investigating PI3Kγ inhibition in myeloid-driven tumour immune suppression. The identified studies underscore the potential of PI3Kγ inhibition in reshaping the TME by modulating myeloid cell functions. The combination of PI3Kγ inhibition with other therapeutic modalities demonstrated enhanced antitumour effects, suggesting a synergistic approach to overcome immune suppression. These findings support the potential of PI3Kγ-targeted therapies, particularly in combination regimens, as a promising avenue for future clinical exploration in diverse solid tumour types.

肿瘤微环境(TME)中免疫细胞和基质细胞之间错综复杂的相互作用对肿瘤的进展产生了重大影响。髓系细胞,包括肿瘤相关巨噬细胞(TAMs)、中性粒细胞(TANs)和髓源抑制细胞(MDSCs),对肿瘤微环境中的免疫抑制做出了贡献(Nakamura 和 Smyth 在 Cell Mol Immunol 17(1):1-12 (2020). https://doi.org/10.1038/s41423-019-0306-1 ; DeNardo 和 Ruffell 在 Nat Rev Immunol 19(6):369-382 (2019). https://doi.org/10.1038/s41577-019-0127-6 )。这对依赖宿主免疫发挥作用的新型免疫疗法提出了巨大挑战。本系统综述探讨了抑制磷酸肌酸 3- 激酶γ(PI3Kγ)作为逆转实体瘤中髓系驱动的免疫抑制策略的临床前证据。2022 年 10 月 6 日,我们使用关键词和主题词检索了 EMBASE、MEDLINE 和 PubMed 数据库,以获取相关研究。这些研究以动物模型中的PI3Kγ抑制为重点,均符合预先确定的纳入和排除标准。提取的数据包括肿瘤生长动力学、生存终点和免疫反应,并进行了荟萃分析。研究遵循 PRISMA 和 MOOSE 指南。共有 36 项涉及 73 种动物模型的研究被纳入综述和荟萃分析。肿瘤模型包括乳腺癌、结直肠癌、肺癌、皮肤癌、胰腺癌、脑癌、肝癌、前列腺癌、头颈癌、软组织癌、胃癌和口腔癌。主要的PI3Kγ抑制剂是IPI-549和TG100-115,这两种抑制剂对γ异构体具有良好的特异性。81%的研究探讨了联合疗法,通常涉及化疗、放疗、免疫检查点抑制剂、生物制剂或疫苗。对肿瘤生长动力学的分析表明,PI3Kγ单药治疗的反应虽不尽相同,但具有统计学意义,而联合治疗组的肿瘤生长则持续降低。生存期分析表明,联合治疗可明显提高中位总生存期。这篇系统综述全面分析了在骨髓驱动的肿瘤免疫抑制中研究 PI3Kγ 抑制的临床前研究。已确定的研究强调了 PI3Kγ 抑制通过调节髓系细胞功能重塑 TME 的潜力。PI3Kγ抑制与其他治疗方式的结合显示出更强的抗肿瘤效果,表明这是一种克服免疫抑制的协同方法。这些发现支持了PI3Kγ靶向疗法的潜力,尤其是在联合疗法中的潜力,是未来在各种实体瘤类型中进行临床探索的一个很有前景的途径。
{"title":"Targeting PI3K-gamma in myeloid driven tumour immune suppression: a systematic review and meta-analysis of the preclinical literature.","authors":"Haonan Xu, Shannon Nicole Russell, Katherine Steiner, Eric O'Neill, Keaton Ian Jones","doi":"10.1007/s00262-024-03779-2","DOIUrl":"10.1007/s00262-024-03779-2","url":null,"abstract":"<p><p>The intricate interplay between immune and stromal cells within the tumour microenvironment (TME) significantly influences tumour progression. Myeloid cells, including tumour-associated macrophages (TAMs), neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs), contribute to immune suppression in the TME (Nakamura and Smyth in Cell Mol Immunol 17(1):1-12 (2020). https://doi.org/10.1038/s41423-019-0306-1 ; DeNardo and Ruffell in Nat Rev Immunol 19(6):369-382 (2019). https://doi.org/10.1038/s41577-019-0127-6 ). This poses a significant challenge for novel immunotherapeutics that rely on host immunity to exert their effect. This systematic review explores the preclinical evidence surrounding the inhibition of phosphoinositide 3-kinase gamma (PI3Kγ) as a strategy to reverse myeloid-driven immune suppression in solid tumours. EMBASE, MEDLINE, and PubMed databases were searched on 6 October 2022 using keyword and subject heading terms to capture relevant studies. The studies, focusing on PI3Kγ inhibition in animal models, were subjected to predefined inclusion and exclusion criteria. Extracted data included tumour growth kinetics, survival endpoints, and immunological responses which were meta-analysed. PRISMA and MOOSE guidelines were followed. A total of 36 studies covering 73 animal models were included in the review and meta-analysis. Tumour models covered breast, colorectal, lung, skin, pancreas, brain, liver, prostate, head and neck, soft tissue, gastric, and oral cancer. The predominant PI3Kγ inhibitors were IPI-549 and TG100-115, demonstrating favourable specificity for the gamma isoform. Combination therapies, often involving chemotherapy, radiotherapy, immune checkpoint inhibitors, biological agents, or vaccines, were explored in 81% of studies. Analysis of tumour growth kinetics revealed a statistically significant though heterogeneous response to PI3Kγ monotherapy, whereas the tumour growth in combination treated groups were more consistently reduced. Survival analysis showed a pronounced increase in median overall survival with combination therapy. This systematic review provides a comprehensive analysis of preclinical studies investigating PI3Kγ inhibition in myeloid-driven tumour immune suppression. The identified studies underscore the potential of PI3Kγ inhibition in reshaping the TME by modulating myeloid cell functions. The combination of PI3Kγ inhibition with other therapeutic modalities demonstrated enhanced antitumour effects, suggesting a synergistic approach to overcome immune suppression. These findings support the potential of PI3Kγ-targeted therapies, particularly in combination regimens, as a promising avenue for future clinical exploration in diverse solid tumour types.</p>","PeriodicalId":9595,"journal":{"name":"Cancer Immunology, Immunotherapy","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11303654/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892921","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
Cancer Immunology, Immunotherapy
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:481959085
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