首页 > 最新文献

Cancer Communications最新文献

英文 中文
Tryptophan 2,3-dioxygenase-positive matrix fibroblasts fuel breast cancer lung metastasis via kynurenine-mediated ferroptosis resistance of metastatic cells and T cell dysfunction. 色氨酸-2,3-二氧合酶阳性基质成纤维细胞通过犬尿氨酸介导的转移细胞铁蛋白沉积抗性和 T 细胞功能障碍助长乳腺癌肺转移。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-02 DOI: 10.1002/cac2.12608
Yongcan Liu, Shanchun Chen, Xueying Wan, Rui Wang, Haojun Luo, Chao Chang, Peijin Dai, Yubi Gan, Yuetong Guo, Yixuan Hou, Yan Sun, Yong Teng, Xiaojiang Cui, Manran Liu

Background: Tumor metastasis is a major threat to cancer patient survival. The organ-specific niche plays a pivotal role in tumor organotropic metastasis. Fibroblasts serve as a vital component of the metastatic microenvironment, but how heterogeneous metastasis-associated fibroblasts (MAFs) promote organotropic metastasis is poorly characterized. Here, we aimed to decipher the heterogeneity of MAFs and elucidate the distinct roles of these fibroblasts in pulmonary metastasis formation in breast cancer.

Methods: Mouse models of breast cancer pulmonary metastasis were established using an in vivo selection method of repeated injections of metastatic cells purified from the mouse lung. Single-cell RNA-sequencing (scRNA-seq) was employed to investigate the heterogeneity of MAFs. Transgenic mice were used to examine the contribution of tryptophan 2,3-dioxygenase-positive matrix fibroblasts (TDO2+ MFs) in lung metastasis.

Results: We uncovered 3 subtypes of MAFs in the lung metastatic microenvironment, and their transcriptome profiles changed dynamically as lung metastasis evolved. As the predominant subtype, MFs were exclusively marked by platelet-derived growth factor receptor alpha (PDGFRA) and mainly located on the edge of the metastasis, and T cells were enriched around MFs. Notably, high MF signatures were significantly associated with poor survival in breast cancer patients. Lung metastases were markedly diminished, and the suppression of T cells was dramatically attenuated in MF-depleted experimental metastatic mouse models. We found that TDO2+ MFs controlled pulmonary metastasis by producing kynurenine (KYN), which upregulated ferritin heavy chain 1 (FTH1) level in disseminated tumor cells (DTCs), enabling DTCs to resist ferroptosis. Moreover, TDO2+ MF-secreted chemokines C-C motif chemokine ligand 8 (CCL8) and C-C motif chemokine ligand 11 (CCL11) recruited T cells. TDO2+ MF-derived KYN induced T cell dysfunction. Conditional knockout of Tdo2 in MFs diminished lung metastasis and enhanced immune activation.

Conclusions: Our study reveals crucial roles of TDO2+ MFs in promoting lung metastasis and DTCs' immune evasion in the metastatic niche. It suggests that targeting the metabolism of lung-specific stromal cells may be an effective treatment strategy for breast cancer patients with lung metastasis.

背景:肿瘤转移是癌症患者生存的主要威胁:肿瘤转移是癌症患者生存的主要威胁。器官特异性生态位在肿瘤器官转移中起着关键作用。成纤维细胞是转移微环境的重要组成部分,但异质性转移相关成纤维细胞(MAFs)是如何促进器官转移的还不清楚。在此,我们旨在破译MAFs的异质性,并阐明这些成纤维细胞在乳腺癌肺转移形成中的不同作用:方法:通过反复注射从小鼠肺部纯化的转移细胞的体内选择方法,建立了乳腺癌肺转移小鼠模型。采用单细胞 RNA 测序(scRNA-seq)研究 MAFs 的异质性。利用转基因小鼠研究色氨酸-2,3-二氧合酶阳性基质成纤维细胞(TDO2+ MFs)在肺转移中的贡献:结果:我们在肺转移微环境中发现了三种亚型的基质成纤维细胞,它们的转录组特征随着肺转移的发展而发生动态变化。作为最主要的亚型,MFs完全以血小板衍生生长因子受体α(PDGFRA)为标志,主要位于转移灶边缘,T细胞富集在MFs周围。值得注意的是,高MF特征与乳腺癌患者的低生存率密切相关。在MF缺失的实验性转移小鼠模型中,肺转移明显减少,T细胞的抑制作用也显著减弱。我们发现,TDO2+中频因子通过产生犬尿氨酸(KYN)控制肺转移,而犬尿氨酸能上调播散性肿瘤细胞(DTCs)中铁蛋白重链1(FTH1)的水平,使DTCs能抵抗铁蛋白沉积。此外,TDO2+ MF分泌的趋化因子C-C基团趋化因子配体8(CCL8)和C-C基团趋化因子配体11(CCL11)可招募T细胞。TDO2+ MF衍生的KYN诱导T细胞功能障碍。有条件地敲除MF中的Tdo2可减少肺转移并增强免疫激活:我们的研究揭示了TDO2+ MFs在促进肺转移和DTCs在转移龛中的免疫逃避中的关键作用。结论:我们的研究揭示了 TDO2+ MFs 在促进肺转移和 DTCs 在转移龛中免疫逃避中的关键作用,这表明针对肺特异性基质细胞的代谢可能是乳腺癌肺转移患者的一种有效治疗策略。
{"title":"Tryptophan 2,3-dioxygenase-positive matrix fibroblasts fuel breast cancer lung metastasis via kynurenine-mediated ferroptosis resistance of metastatic cells and T cell dysfunction.","authors":"Yongcan Liu, Shanchun Chen, Xueying Wan, Rui Wang, Haojun Luo, Chao Chang, Peijin Dai, Yubi Gan, Yuetong Guo, Yixuan Hou, Yan Sun, Yong Teng, Xiaojiang Cui, Manran Liu","doi":"10.1002/cac2.12608","DOIUrl":"https://doi.org/10.1002/cac2.12608","url":null,"abstract":"<p><strong>Background: </strong>Tumor metastasis is a major threat to cancer patient survival. The organ-specific niche plays a pivotal role in tumor organotropic metastasis. Fibroblasts serve as a vital component of the metastatic microenvironment, but how heterogeneous metastasis-associated fibroblasts (MAFs) promote organotropic metastasis is poorly characterized. Here, we aimed to decipher the heterogeneity of MAFs and elucidate the distinct roles of these fibroblasts in pulmonary metastasis formation in breast cancer.</p><p><strong>Methods: </strong>Mouse models of breast cancer pulmonary metastasis were established using an in vivo selection method of repeated injections of metastatic cells purified from the mouse lung. Single-cell RNA-sequencing (scRNA-seq) was employed to investigate the heterogeneity of MAFs. Transgenic mice were used to examine the contribution of tryptophan 2,3-dioxygenase-positive matrix fibroblasts (TDO2<sup>+</sup> MFs) in lung metastasis.</p><p><strong>Results: </strong>We uncovered 3 subtypes of MAFs in the lung metastatic microenvironment, and their transcriptome profiles changed dynamically as lung metastasis evolved. As the predominant subtype, MFs were exclusively marked by platelet-derived growth factor receptor alpha (PDGFRA) and mainly located on the edge of the metastasis, and T cells were enriched around MFs. Notably, high MF signatures were significantly associated with poor survival in breast cancer patients. Lung metastases were markedly diminished, and the suppression of T cells was dramatically attenuated in MF-depleted experimental metastatic mouse models. We found that TDO2<sup>+</sup> MFs controlled pulmonary metastasis by producing kynurenine (KYN), which upregulated ferritin heavy chain 1 (FTH1) level in disseminated tumor cells (DTCs), enabling DTCs to resist ferroptosis. Moreover, TDO2<sup>+</sup> MF-secreted chemokines C-C motif chemokine ligand 8 (CCL8) and C-C motif chemokine ligand 11 (CCL11) recruited T cells. TDO2<sup>+</sup> MF-derived KYN induced T cell dysfunction. Conditional knockout of Tdo2 in MFs diminished lung metastasis and enhanced immune activation.</p><p><strong>Conclusions: </strong>Our study reveals crucial roles of TDO2<sup>+</sup> MFs in promoting lung metastasis and DTCs' immune evasion in the metastatic niche. It suggests that targeting the metabolism of lung-specific stromal cells may be an effective treatment strategy for breast cancer patients with lung metastasis.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142104686","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Perioperative toripalimab plus neoadjuvant chemotherapy might improve outcomes in resectable esophageal cancer: an interim analysis of a phase III randomized clinical trial. 围手术期托利帕利单抗加新辅助化疗可改善可切除食管癌的预后:一项III期随机临床试验的中期分析。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-02 DOI: 10.1002/cac2.12604
Yan Zheng, Guanghui Liang, Dongfeng Yuan, Xianben Liu, Yufeng Ba, Zimin Qin, Sining Shen, Zhenxuan Li, Haibo Sun, Baoxing Liu, Quanli Gao, Peng Li, Zongfei Wang, Shilei Liu, Jianping Zhu, Haoran Wang, Haibo Ma, Zhenzhen Liu, Fei Zhao, Jun Zhang, He Zhang, Daoyuan Wu, Jinrong Qu, Jie Ma, Peng Zhang, Wenjie Ma, Ming Yan, Yongkui Yu, Qing Li, Jiangong Zhang, Wenqun Xing

Background: In the era of immunotherapy, neoadjuvant immunochemotherapy (NAIC) for the treatment of locally advanced esophageal squamous cell carcinoma (ESCC) is used clinically but lacks of high-level clinical evidence. This study aimed to compare the safety and long-term efficacy of NAIC followed by minimally invasive esophagectomy (MIE) with those of neoadjuvant chemotherapy (NAC) followed by MIE.

Methods: A prospective, single-center, open-label, randomized phase III clinical trial was conducted at Henan Cancer Hospital, Zhengzhou, China. Patients were randomly assigned to receive either neoadjuvant toripalimab (240 mg) plus paclitaxel (175 mg/m2) + cisplatin (75 mg/m2) (toripalimab group) or paclitaxel + cisplatin alone (chemotherapy group) every 3 weeks for 2 cycles. After surgery, the toripalimab group received toripalimab (240 mg every 3 weeks for up to 6 months). The primary endpoint was event-free survival (EFS). The pathological complete response (pCR) and overall survival (OS) were key secondary endpoints. Adverse events (AEs) and quality of life were also assessed.

Results: Between May 15, 2020 and August 13, 2021, 252 ESCC patients ranging from T1N1-3M0 to T2-3N0-3M0 were enrolled for interim analysis, with 127 in the toripalimab group and 125 in the chemotherapy group. The 1-year EFS rate was 77.9% in the toripalimab group compared to 64.3% in the chemotherapy group (hazard ratio [HR] = 0.62; 95% confidence interval [CI] = 0.39 to 1.00; P = 0.05). The 1-year OS rates were 94.1% and 83.0% in the toripalimab and chemotherapy groups, respectively (HR = 0.48; 95% CI = 0.24 to 0.97; P = 0.037). The patients in the toripalimab group had a higher pCR rate (18.6% vs. 4.6%; P = 0.001). The rates of postoperative Clavien-Dindo grade IIIb or higher morbidity were 9.8% in the toripalimab group and 6.8% in the chemotherapy group, with no significant difference observed (P = 0.460). The rates of grade 3 or 4 treatment-related AEs did not differ between the two groups (12.5% versus 12.4%).

Conclusions: The interim results of this ongoing trial showed that in resectable ESCC, the addition of perioperative toripalimab to NAC is safe, may improve OS and might change the standard treatment in the future.

背景:在免疫疗法时代,新辅助免疫化疗(NAIC)被用于治疗局部晚期食管鳞状细胞癌(ESCC),但缺乏高水平的临床证据。本研究旨在比较新辅助化疗(NAC)后进行微创食管切除术(MIE)与新辅助化疗后进行微创食管切除术(MIE)的安全性和长期疗效:中国郑州市河南省肿瘤医院开展了一项前瞻性、单中心、开放标签、随机III期临床试验。患者被随机分配接受新辅助托瑞帕利单抗(240毫克)+紫杉醇(175毫克/平方米)+顺铂(75毫克/平方米)(托瑞帕利单抗组)或单独紫杉醇+顺铂(化疗组)治疗,每3周1次,共2个周期。手术后,托里帕利单抗组接受托里帕利单抗治疗(每3周240毫克,最长6个月)。主要终点是无事件生存期(EFS)。病理完全反应(pCR)和总生存期(OS)是次要终点。此外,还对不良事件(AEs)和生活质量进行了评估:2020年5月15日至2021年8月13日,252名T1N1-3M0至T2-3N0-3M0的ESCC患者入组进行中期分析,其中托利帕单抗组127人,化疗组125人。托利帕利单抗组的1年EFS率为77.9%,化疗组为64.3%(危险比[HR] = 0.62;95%置信区间[CI] = 0.39至1.00;P = 0.05)。托利帕利单抗组和化疗组的1年OS率分别为94.1%和83.0%(HR = 0.48;95% CI = 0.24至0.97;P = 0.037)。托利帕单抗组患者的 pCR 率更高(18.6% 对 4.6%;P = 0.001)。术后Clavien-Dindo IIIb级或更高的发病率在托利帕利单抗组为9.8%,在化疗组为6.8%,没有观察到显著差异(P = 0.460)。两组的3级或4级治疗相关AEs发生率没有差异(12.5%对12.4%):这项正在进行的试验的中期结果表明,对于可切除的 ESCC,在 NAC 的基础上加用围术期托利帕利单抗是安全的,可改善 OS,并有可能在未来改变标准治疗方法。
{"title":"Perioperative toripalimab plus neoadjuvant chemotherapy might improve outcomes in resectable esophageal cancer: an interim analysis of a phase III randomized clinical trial.","authors":"Yan Zheng, Guanghui Liang, Dongfeng Yuan, Xianben Liu, Yufeng Ba, Zimin Qin, Sining Shen, Zhenxuan Li, Haibo Sun, Baoxing Liu, Quanli Gao, Peng Li, Zongfei Wang, Shilei Liu, Jianping Zhu, Haoran Wang, Haibo Ma, Zhenzhen Liu, Fei Zhao, Jun Zhang, He Zhang, Daoyuan Wu, Jinrong Qu, Jie Ma, Peng Zhang, Wenjie Ma, Ming Yan, Yongkui Yu, Qing Li, Jiangong Zhang, Wenqun Xing","doi":"10.1002/cac2.12604","DOIUrl":"https://doi.org/10.1002/cac2.12604","url":null,"abstract":"<p><strong>Background: </strong>In the era of immunotherapy, neoadjuvant immunochemotherapy (NAIC) for the treatment of locally advanced esophageal squamous cell carcinoma (ESCC) is used clinically but lacks of high-level clinical evidence. This study aimed to compare the safety and long-term efficacy of NAIC followed by minimally invasive esophagectomy (MIE) with those of neoadjuvant chemotherapy (NAC) followed by MIE.</p><p><strong>Methods: </strong>A prospective, single-center, open-label, randomized phase III clinical trial was conducted at Henan Cancer Hospital, Zhengzhou, China. Patients were randomly assigned to receive either neoadjuvant toripalimab (240 mg) plus paclitaxel (175 mg/m<sup>2</sup>) + cisplatin (75 mg/m<sup>2</sup>) (toripalimab group) or paclitaxel + cisplatin alone (chemotherapy group) every 3 weeks for 2 cycles. After surgery, the toripalimab group received toripalimab (240 mg every 3 weeks for up to 6 months). The primary endpoint was event-free survival (EFS). The pathological complete response (pCR) and overall survival (OS) were key secondary endpoints. Adverse events (AEs) and quality of life were also assessed.</p><p><strong>Results: </strong>Between May 15, 2020 and August 13, 2021, 252 ESCC patients ranging from T1N1-3M0 to T2-3N0-3M0 were enrolled for interim analysis, with 127 in the toripalimab group and 125 in the chemotherapy group. The 1-year EFS rate was 77.9% in the toripalimab group compared to 64.3% in the chemotherapy group (hazard ratio [HR] = 0.62; 95% confidence interval [CI] = 0.39 to 1.00; P = 0.05). The 1-year OS rates were 94.1% and 83.0% in the toripalimab and chemotherapy groups, respectively (HR = 0.48; 95% CI = 0.24 to 0.97; P = 0.037). The patients in the toripalimab group had a higher pCR rate (18.6% vs. 4.6%; P = 0.001). The rates of postoperative Clavien-Dindo grade IIIb or higher morbidity were 9.8% in the toripalimab group and 6.8% in the chemotherapy group, with no significant difference observed (P = 0.460). The rates of grade 3 or 4 treatment-related AEs did not differ between the two groups (12.5% versus 12.4%).</p><p><strong>Conclusions: </strong>The interim results of this ongoing trial showed that in resectable ESCC, the addition of perioperative toripalimab to NAC is safe, may improve OS and might change the standard treatment in the future.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142104685","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. 靶向SRSF10可抑制M2巨噬细胞极化,并增强肝细胞癌的抗PD-1疗法。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-02 DOI: 10.1002/cac2.12607
Jialiang Cai, Lina Song, Feng Zhang, Suiyi Wu, Guiqi Zhu, Peiling Zhang, Shiping Chen, Junxian Du, Biao Wang, Yufan Cai, Yi Yang, Jinglei Wan, Jian Zhou, Jia Fan, Zhi Dai

Background: The efficacy of immune checkpoint blockade therapy in patients with hepatocellular carcinoma (HCC) remains poor. Although serine- and arginine-rich splicing factor (SRSF) family members play crucial roles in tumors, their impact on tumor immunology remains unclear. This study aimed to elucidate the role of SRSF10 in HCC immunotherapy.

Methods: To identify the key genes associated with immunotherapy resistance, we conducted single-nuclear RNA sequencing, multiplex immunofluorescence, and The Cancer Genome Atlas and Gene Expression Omnibus database analyses. We investigated the biological functions of SRSF10 in immune evasion using in vitro co-culture systems, flow cytometry, various tumor-bearing mouse models, and patient-derived organotypic tumor spheroids.

Results: SRSF10 was upregulated in various tumors and associated with poor prognosis. Moreover, SRSF10 positively regulated lactate production, and SRSF10/glycolysis/ histone H3 lysine 18 lactylation (H3K18la) formed a positive feedback loop in tumor cells. Increased lactate levels promoted M2 macrophage polarization, thereby inhibiting CD8+ T cell activity. Mechanistically, SRSF10 interacted with the 3'-untranslated region of MYB, enhancing MYB RNA stability, and subsequently upregulating key glycolysis-related enzymes including glucose transporter 1 (GLUT1), hexokinase 1 (HK1), lactate dehydrogenase A (LDHA), resulting in elevated intracellular and extracellular lactate levels. Lactate accumulation induced histone lactylation, which further upregulated SRSF10 expression. Additionally, lactate produced by tumors induced lactylation of the histone H3K18la site upon transport into macrophages, thereby activating transcription and enhancing pro-tumor macrophage activity. M2 macrophages, in turn, inhibited the enrichment of CD8+ T cells and the proportion of interferon-γ+CD8+ T cells in the tumor microenvironment (TME), thus creating an immunosuppressive TME. Clinically, SRSF10 could serve as a biomarker for assessing immunotherapy resistance in various solid tumors. Pharmacological targeting of SRSF10 with a selective inhibitor 1C8 enhanced the efficacy of programmed cell death 1 (PD-1) monoclonal antibodies (mAbs) in both murine and human preclinical models.

Conclusions: The SRSF10/MYB/glycolysis/lactate axis is critical for triggering immune evasion and anti-PD-1 resistance. Inhibiting SRSF10 by 1C8 may overcome anti-PD-1 tolerance in HCC.

背景:免疫检查点阻断疗法对肝细胞癌(HCC)患者的疗效仍然不佳。尽管富含丝氨酸和精氨酸的剪接因子(SRSF)家族成员在肿瘤中发挥着关键作用,但它们对肿瘤免疫学的影响仍不清楚。本研究旨在阐明 SRSF10 在 HCC 免疫治疗中的作用:为了确定与免疫治疗耐药性相关的关键基因,我们进行了单核 RNA 测序、多重免疫荧光以及癌症基因组图谱和基因表达总库数据库分析。我们利用体外共培养系统、流式细胞术、各种肿瘤小鼠模型和患者来源的器官型肿瘤球状体研究了SRSF10在免疫逃避中的生物学功能:结果:SRSF10在多种肿瘤中上调,并与不良预后相关。此外,SRSF10 能正向调节乳酸盐的产生,SRSF10/糖酵解/组蛋白 H3 赖氨酸 18 乳化(H3K18la)在肿瘤细胞中形成正反馈回路。乳酸水平的增加促进了 M2 巨噬细胞的极化,从而抑制了 CD8+ T 细胞的活性。从机制上讲,SRSF10与MYB的3'-非翻译区相互作用,增强了MYB RNA的稳定性,随后上调了关键的糖酵解相关酶,包括葡萄糖转运体1(GLUT1)、己糖激酶1(HK1)、乳酸脱氢酶A(LDHA),导致细胞内和细胞外乳酸水平升高。乳酸积累诱导组蛋白乳化,进一步上调 SRSF10 的表达。此外,肿瘤产生的乳酸在转运到巨噬细胞后会诱导组蛋白 H3K18la 位点的乳化,从而激活转录并增强促肿瘤巨噬细胞的活性。反过来,M2 巨噬细胞又会抑制肿瘤微环境(TME)中 CD8+ T 细胞的富集和干扰素-γ+CD8+ T 细胞的比例,从而形成免疫抑制的 TME。在临床上,SRSF10 可作为评估各种实体瘤免疫疗法耐药性的生物标记物。用选择性抑制剂1C8对SRSF10进行药理靶向治疗,可提高程序性细胞死亡1(PD-1)单克隆抗体(mAbs)在小鼠和人类临床前模型中的疗效:SRSF10/MYB/糖酵解/乳酸轴是引发免疫逃避和抗PD-1耐药性的关键。通过1C8抑制SRSF10可克服HCC的抗PD-1耐受性。
{"title":"Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma.","authors":"Jialiang Cai, Lina Song, Feng Zhang, Suiyi Wu, Guiqi Zhu, Peiling Zhang, Shiping Chen, Junxian Du, Biao Wang, Yufan Cai, Yi Yang, Jinglei Wan, Jian Zhou, Jia Fan, Zhi Dai","doi":"10.1002/cac2.12607","DOIUrl":"https://doi.org/10.1002/cac2.12607","url":null,"abstract":"<p><strong>Background: </strong>The efficacy of immune checkpoint blockade therapy in patients with hepatocellular carcinoma (HCC) remains poor. Although serine- and arginine-rich splicing factor (SRSF) family members play crucial roles in tumors, their impact on tumor immunology remains unclear. This study aimed to elucidate the role of SRSF10 in HCC immunotherapy.</p><p><strong>Methods: </strong>To identify the key genes associated with immunotherapy resistance, we conducted single-nuclear RNA sequencing, multiplex immunofluorescence, and The Cancer Genome Atlas and Gene Expression Omnibus database analyses. We investigated the biological functions of SRSF10 in immune evasion using in vitro co-culture systems, flow cytometry, various tumor-bearing mouse models, and patient-derived organotypic tumor spheroids.</p><p><strong>Results: </strong>SRSF10 was upregulated in various tumors and associated with poor prognosis. Moreover, SRSF10 positively regulated lactate production, and SRSF10/glycolysis/ histone H3 lysine 18 lactylation (H3K18la) formed a positive feedback loop in tumor cells. Increased lactate levels promoted M2 macrophage polarization, thereby inhibiting CD8<sup>+</sup> T cell activity. Mechanistically, SRSF10 interacted with the 3'-untranslated region of MYB, enhancing MYB RNA stability, and subsequently upregulating key glycolysis-related enzymes including glucose transporter 1 (GLUT1), hexokinase 1 (HK1), lactate dehydrogenase A (LDHA), resulting in elevated intracellular and extracellular lactate levels. Lactate accumulation induced histone lactylation, which further upregulated SRSF10 expression. Additionally, lactate produced by tumors induced lactylation of the histone H3K18la site upon transport into macrophages, thereby activating transcription and enhancing pro-tumor macrophage activity. M2 macrophages, in turn, inhibited the enrichment of CD8<sup>+</sup> T cells and the proportion of interferon-γ<sup>+</sup>CD8<sup>+</sup> T cells in the tumor microenvironment (TME), thus creating an immunosuppressive TME. Clinically, SRSF10 could serve as a biomarker for assessing immunotherapy resistance in various solid tumors. Pharmacological targeting of SRSF10 with a selective inhibitor 1C8 enhanced the efficacy of programmed cell death 1 (PD-1) monoclonal antibodies (mAbs) in both murine and human preclinical models.</p><p><strong>Conclusions: </strong>The SRSF10/MYB/glycolysis/lactate axis is critical for triggering immune evasion and anti-PD-1 resistance. Inhibiting SRSF10 by 1C8 may overcome anti-PD-1 tolerance in HCC.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142119068","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Unraveling the routes to distant metastases in colorectal cancer: Tumor deposits and lymph node metastases as the gateway. 揭示结直肠癌远处转移的途径:以肿瘤沉积和淋巴结转移为入口。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-25 DOI: 10.1002/cac2.12598
Nelleke Pietronella Maria Brouwer, Ayse Selcen Oguz Erdogan, Shannon van Vliet, Natasja Rutgers, Nikki Knijn, Gesina van Lijnschoten, Jessica Juliana Tan, Johannes Hendrik Willem de Wilt, Niek Hugen, Gina Brown, Femke Simmer, Iris Dionne Nagtegaal
{"title":"Unraveling the routes to distant metastases in colorectal cancer: Tumor deposits and lymph node metastases as the gateway.","authors":"Nelleke Pietronella Maria Brouwer, Ayse Selcen Oguz Erdogan, Shannon van Vliet, Natasja Rutgers, Nikki Knijn, Gesina van Lijnschoten, Jessica Juliana Tan, Johannes Hendrik Willem de Wilt, Niek Hugen, Gina Brown, Femke Simmer, Iris Dionne Nagtegaal","doi":"10.1002/cac2.12598","DOIUrl":"https://doi.org/10.1002/cac2.12598","url":null,"abstract":"","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142055045","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Concurrent chemoradiotherapyof different radiation doses and different irradiation fields for locally advanced thoracic esophageal squamous cell carcinoma: A randomized, multicenter, phase III clinical trial. 针对局部晚期胸腔食管鳞状细胞癌的不同放射剂量和不同照射野的同期化放疗:一项随机、多中心、III 期临床试验。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-19 DOI: 10.1002/cac2.12601
Jian Zhang, Minghao Li, Kaixian Zhang, Anping Zheng, Guang Li, Wei Huang, Shaoshui Chen, Xiangming Chen, Xiaomin Li, Yanxing Sheng, Xinchen Sun, Liping Liu, Xiaowei Liu, Jie Li, Jun Wang, Hong Ge, Shucheng Ye, Qingsong Pang, Xianwen Zhang, Shengbin Dai, Richard Yu, Wendong Gu, Mingming Dai, Gaowa Siqin, Yunwei Han, Xiaolin Ge, Xin Yuan, Yongjing Yang, Haiwen Zhu, Juan Pu, Lihua Dong, Xiangdong Sun, Jundong Zhou, Weidong Mao, Fei Gao, Haiqun Lin, Heyi Gong, Tao Zhou, Zhenjiang Li, Hongsheng Li, Zhongtang Wang, Baosheng Li

Background: Concurrent chemoradiotherapy (CCRT) is the standard treatment for locally advanced esophageal squamous cell carcinoma (ESCC). However, the optimal radiotherapy regimen, particularly in terms of total dose and planned range of irradiation field, remains unclear. This phase III clinical trial aimed to compare the survival benefits between different radiation doses and different target fields.

Methods: This trial compared two aspects of radiation treatment, total dose and field, using a two-by-two factorial design. The high-dose (HD) group received 59.4 Gy radiation, and the standard-dose (SD) group received 50.4 Gy. The involved field irradiation (IFI) group and elective nodal irradiation (ENI) group adopted different irradiation ranges. The participants were assigned to one of the four groups (HD+ENI, HD+IFI, SD+ENI and SD+IFI). The primary endpoint was overall survival (OS), and the secondary endpoints included progression-free survival (PFS). The synergy indexwas used to measure the interaction effect between dose and field.

Results: The interaction analysis did not reveal significant synergistic effects between the dose and irradiation field. In comparison to the target field, patients in IFI or ENI showed similar OS (hazard ratio [HR] = 0.99, 95% CI: 0.80-1.23, p = 0.930) and PFS (HR = 1.02, 95% CI: 0.82-1.25). The HD treatment did not show significantly prolonged OS compared with SD (HR = 0.90, 95% CI: 0.72-1.11, p = 0.318), but it suggested improved PFS (25.2 months to 18.0 months). Among the four groups, the HD+IFI group presented the best survival, while the SD+IFI group had the worst prognosis. No significant difference in the occurrence of severe adverse events was found in dose or field comparisons.

Conclusions: IFI demonstrated similar treatment efficacy to ENI in CCRT of ESCC. The HD demonstrated improved PFS, but did not significantly improve OS. The dose escalation based on IFI (HD+IFI) showed better therapeutic efficacy than the current recommendation (SD+ENI) and is worth further validation.

背景:同期放化疗(CCRT)是局部晚期食管鳞状细胞癌(ESCC)的标准治疗方法。然而,最佳放疗方案,尤其是总剂量和计划照射野范围,仍不明确。这项III期临床试验旨在比较不同放射剂量和不同靶区对患者生存的益处:该试验采用二乘二的因子设计,比较了放射治疗的两个方面,即总剂量和照射野。高剂量(HD)组接受59.4 Gy的放射治疗,标准剂量(SD)组接受50.4 Gy的放射治疗。介入野照射(IFI)组和选择性结节照射(ENI)组采用不同的照射范围。参与者被分配到四组(HD+ENI组、HD+IFI组、SD+ENI组和SD+IFI组)中的一组。主要终点是总生存期(OS),次要终点包括无进展生存期(PFS)。协同作用指数用于衡量剂量和领域之间的交互作用:结果:交互作用分析并未显示剂量和照射野之间有明显的协同效应。与靶区相比,IFI 或 ENI 患者的 OS(危险比 [HR] = 0.99,95% CI:0.80-1.23,p = 0.930)和 PFS(HR = 1.02,95% CI:0.82-1.25)相似。与 SD 相比,HD 治疗并没有明显延长 OS(HR = 0.90,95% CI:0.72-1.11,p = 0.318),但却改善了 PFS(25.2 个月至 18.0 个月)。在四组患者中,HD+IFI 组的生存率最高,而 SD+IFI 组的预后最差。在剂量或病区比较中,严重不良事件的发生率无明显差异:结论:在ESCC的CCRT治疗中,IFI与ENI的疗效相似。结论:在 ESCC 的 CCRT 治疗中,IFI 的疗效与 ENI 相似,HD 改善了 PFS,但并未显著改善 OS。基于IFI的剂量升级(HD+IFI)比目前的推荐方案(SD+ENI)显示出更好的疗效,值得进一步验证。
{"title":"Concurrent chemoradiotherapyof different radiation doses and different irradiation fields for locally advanced thoracic esophageal squamous cell carcinoma: A randomized, multicenter, phase III clinical trial.","authors":"Jian Zhang, Minghao Li, Kaixian Zhang, Anping Zheng, Guang Li, Wei Huang, Shaoshui Chen, Xiangming Chen, Xiaomin Li, Yanxing Sheng, Xinchen Sun, Liping Liu, Xiaowei Liu, Jie Li, Jun Wang, Hong Ge, Shucheng Ye, Qingsong Pang, Xianwen Zhang, Shengbin Dai, Richard Yu, Wendong Gu, Mingming Dai, Gaowa Siqin, Yunwei Han, Xiaolin Ge, Xin Yuan, Yongjing Yang, Haiwen Zhu, Juan Pu, Lihua Dong, Xiangdong Sun, Jundong Zhou, Weidong Mao, Fei Gao, Haiqun Lin, Heyi Gong, Tao Zhou, Zhenjiang Li, Hongsheng Li, Zhongtang Wang, Baosheng Li","doi":"10.1002/cac2.12601","DOIUrl":"https://doi.org/10.1002/cac2.12601","url":null,"abstract":"<p><strong>Background: </strong>Concurrent chemoradiotherapy (CCRT) is the standard treatment for locally advanced esophageal squamous cell carcinoma (ESCC). However, the optimal radiotherapy regimen, particularly in terms of total dose and planned range of irradiation field, remains unclear. This phase III clinical trial aimed to compare the survival benefits between different radiation doses and different target fields.</p><p><strong>Methods: </strong>This trial compared two aspects of radiation treatment, total dose and field, using a two-by-two factorial design. The high-dose (HD) group received 59.4 Gy radiation, and the standard-dose (SD) group received 50.4 Gy. The involved field irradiation (IFI) group and elective nodal irradiation (ENI) group adopted different irradiation ranges. The participants were assigned to one of the four groups (HD+ENI, HD+IFI, SD+ENI and SD+IFI). The primary endpoint was overall survival (OS), and the secondary endpoints included progression-free survival (PFS). The synergy indexwas used to measure the interaction effect between dose and field.</p><p><strong>Results: </strong>The interaction analysis did not reveal significant synergistic effects between the dose and irradiation field. In comparison to the target field, patients in IFI or ENI showed similar OS (hazard ratio [HR] = 0.99, 95% CI: 0.80-1.23, p = 0.930) and PFS (HR = 1.02, 95% CI: 0.82-1.25). The HD treatment did not show significantly prolonged OS compared with SD (HR = 0.90, 95% CI: 0.72-1.11, p = 0.318), but it suggested improved PFS (25.2 months to 18.0 months). Among the four groups, the HD+IFI group presented the best survival, while the SD+IFI group had the worst prognosis. No significant difference in the occurrence of severe adverse events was found in dose or field comparisons.</p><p><strong>Conclusions: </strong>IFI demonstrated similar treatment efficacy to ENI in CCRT of ESCC. The HD demonstrated improved PFS, but did not significantly improve OS. The dose escalation based on IFI (HD+IFI) showed better therapeutic efficacy than the current recommendation (SD+ENI) and is worth further validation.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142003690","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
FGFR3 alterations in bladder cancer: Sensitivity and resistance to targeted therapies. 膀胱癌中的 FGFR3 改变:靶向疗法的敏感性和耐药性。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-19 DOI: 10.1002/cac2.12602
Maxim Noeraparast, Katarina Krajina, Renate Pichler, Dora Niedersüß-Beke, Shahrokh F Shariat, Viktor Grünwald, Sascha Ahyai, Martin Pichler

In this review, we revisit the pivotal role of fibroblast growth factor receptor 3 (FGFR3) in bladder cancer (BLCA), underscoring its prevalence in both non-muscle-invasive and muscle-invasive forms of the disease. FGFR3 mutations in up to half of BLCAs play a well-established role in tumorigenesis, shaping distinct tumor initiation patterns and impacting the tumor microenvironment (TME). Emphasizing the importance of considering epithelial-mesenchymal transition profile and TME status, we revisit their relevance in predicting responses to immune checkpoint inhibitors in FGFR3-mutated BLCAs. This writing highlights the initially promising yet transient efficacy of the FGFR inhibitor Erdafitinib on FGFR3-mutated BLCA, stressing the pressing need to unravel resistance mechanisms and identify co-targets for future combinatorial studies. A thorough analysis of recent preclinical and clinical evidence reveals resistance mechanisms, including secondary mutations, epigenetic alterations in pathway effectors, phenotypic heterogeneity, and population-specific variations within FGFR3 mutational status. Lastly, we discuss the potential of combinatorial treatments and concepts like synthetic lethality for discovering more effective targeted therapies against FGFR3-mutated BLCA.

在这篇综述中,我们重新审视了成纤维细胞生长因子受体 3 (FGFR3) 在膀胱癌 (BLCA) 中的关键作用,强调了它在非肌肉浸润性和肌肉浸润性膀胱癌中的普遍性。多达半数的膀胱癌患者体内的 FGFR3 基因突变在肿瘤发生过程中发挥了公认的作用,形成了独特的肿瘤起始模式并影响了肿瘤微环境(TME)。我们强调了考虑上皮-间质转化特征和TME状态的重要性,并重新审视了它们在预测FGFR3突变BLCA对免疫检查点抑制剂的反应中的相关性。这篇文章强调了FGFR抑制剂Erdafitinib对FGFR3突变的BLCA最初具有良好但短暂的疗效,强调了揭示耐药机制并为未来的联合研究确定共同靶点的迫切需要。对近期临床前和临床证据的深入分析揭示了耐药机制,包括继发性突变、通路效应因子的表观遗传学改变、表型异质性以及 FGFR3 突变状态下的人群特异性变化。最后,我们讨论了组合疗法和合成致死等概念的潜力,以发现针对 FGFR3 突变的 BLCA 的更有效的靶向疗法。
{"title":"FGFR3 alterations in bladder cancer: Sensitivity and resistance to targeted therapies.","authors":"Maxim Noeraparast, Katarina Krajina, Renate Pichler, Dora Niedersüß-Beke, Shahrokh F Shariat, Viktor Grünwald, Sascha Ahyai, Martin Pichler","doi":"10.1002/cac2.12602","DOIUrl":"https://doi.org/10.1002/cac2.12602","url":null,"abstract":"<p><p>In this review, we revisit the pivotal role of fibroblast growth factor receptor 3 (FGFR3) in bladder cancer (BLCA), underscoring its prevalence in both non-muscle-invasive and muscle-invasive forms of the disease. FGFR3 mutations in up to half of BLCAs play a well-established role in tumorigenesis, shaping distinct tumor initiation patterns and impacting the tumor microenvironment (TME). Emphasizing the importance of considering epithelial-mesenchymal transition profile and TME status, we revisit their relevance in predicting responses to immune checkpoint inhibitors in FGFR3-mutated BLCAs. This writing highlights the initially promising yet transient efficacy of the FGFR inhibitor Erdafitinib on FGFR3-mutated BLCA, stressing the pressing need to unravel resistance mechanisms and identify co-targets for future combinatorial studies. A thorough analysis of recent preclinical and clinical evidence reveals resistance mechanisms, including secondary mutations, epigenetic alterations in pathway effectors, phenotypic heterogeneity, and population-specific variations within FGFR3 mutational status. Lastly, we discuss the potential of combinatorial treatments and concepts like synthetic lethality for discovering more effective targeted therapies against FGFR3-mutated BLCA.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142003691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
4-1BB transcriptomic expression patterns across malignancies: Implications for clinical trials of 4-1BB agonists. 不同恶性肿瘤的 4-1BB 转录组表达模式:4-1BB 激动剂临床试验的意义。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-04 DOI: 10.1002/cac2.12592
Yuji Uehara, Shumei Kato, Daisuke Nishizaki, Hirotaka Miyashita, Suzanna Lee, Mary K Nesline, Sarabjot Pabla, Jeffrey M Conroy, Paul DePietro, Heidi Ko, Jason K Sicklick, Razelle Kurzrock
{"title":"4-1BB transcriptomic expression patterns across malignancies: Implications for clinical trials of 4-1BB agonists.","authors":"Yuji Uehara, Shumei Kato, Daisuke Nishizaki, Hirotaka Miyashita, Suzanna Lee, Mary K Nesline, Sarabjot Pabla, Jeffrey M Conroy, Paul DePietro, Heidi Ko, Jason K Sicklick, Razelle Kurzrock","doi":"10.1002/cac2.12592","DOIUrl":"10.1002/cac2.12592","url":null,"abstract":"","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141888502","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Beyond the Gut: The intratumoral microbiome's influence on tumorigenesis and treatment response. 超越肠道:瘤内微生物组对肿瘤发生和治疗反应的影响
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1002/cac2.12597
Hao Zhang, Li Fu, Xinwen Leiliang, Chunrun Qu, Wantao Wu, Rong Wen, Ning Huang, Qiuguang He, Quan Cheng, Guodong Liu, Yuan Cheng

The intratumoral microbiome (TM) refers to the microorganisms in the tumor tissues, including bacteria, fungi, viruses, and so on, and is distinct from the gut microbiome and circulating microbiota. TM is strongly associated with tumorigenesis, progression, metastasis, and response to therapy. This paper highlights the current status of TM. Tract sources, adjacent normal tissue, circulatory system, and concomitant tumor co-metastasis are the main origin of TM. The advanced techniques in TM analysis are comprehensively summarized. Besides, TM is involved in tumor progression through several mechanisms, including DNA damage, activation of oncogenic signaling pathways (phosphoinositide 3-kinase [PI3K], signal transducer and activator of transcription [STAT], WNT/β-catenin, and extracellular regulated protein kinases [ERK]), influence of cytokines and induce inflammatory responses, and interaction with the tumor microenvironment (anti-tumor immunity, pro-tumor immunity, and microbial-derived metabolites). Moreover, promising directions of TM in tumor therapy include immunotherapy, chemotherapy, radiotherapy, the application of probiotics/prebiotics/synbiotics, fecal microbiome transplantation, engineered microbiota, phage therapy, and oncolytic virus therapy. The inherent challenges of clinical application are also summarized. This review provides a comprehensive landscape for analyzing TM, especially the TM-related mechanisms and TM-based treatment in cancer.

肿瘤内微生物组(TM)是指肿瘤组织中的微生物,包括细菌、真菌、病毒等,有别于肠道微生物组和循环微生物组。TM与肿瘤的发生、发展、转移和治疗反应密切相关。本文重点介绍了 TM 的现状。TM 的主要来源包括溃疡源、邻近正常组织、循环系统和伴随肿瘤的共同转移。本文全面总结了 TM 分析的先进技术。此外,TM 通过多种机制参与肿瘤进展,包括 DNA 损伤、激活致癌信号通路(磷酸肌酸 3-激酶[PI3K]、转录信号转导和激活剂[STAT]、WNT/β-catenin 和细胞外调节蛋白激酶[ERK])、细胞因子的影响和诱导炎症反应以及与肿瘤微环境的相互作用(抗肿瘤免疫、促肿瘤免疫和微生物衍生代谢产物)。此外,TM 在肿瘤治疗中的应用方向包括免疫疗法、化疗、放疗、益生菌/益生元/合成益生菌的应用、粪便微生物组移植、工程微生物群、噬菌体疗法和溶瘤病毒疗法。此外,还总结了临床应用所面临的固有挑战。这篇综述为分析 TM,尤其是 TM 相关机制和基于 TM 的癌症治疗提供了一个全面的视角。
{"title":"Beyond the Gut: The intratumoral microbiome's influence on tumorigenesis and treatment response.","authors":"Hao Zhang, Li Fu, Xinwen Leiliang, Chunrun Qu, Wantao Wu, Rong Wen, Ning Huang, Qiuguang He, Quan Cheng, Guodong Liu, Yuan Cheng","doi":"10.1002/cac2.12597","DOIUrl":"https://doi.org/10.1002/cac2.12597","url":null,"abstract":"<p><p>The intratumoral microbiome (TM) refers to the microorganisms in the tumor tissues, including bacteria, fungi, viruses, and so on, and is distinct from the gut microbiome and circulating microbiota. TM is strongly associated with tumorigenesis, progression, metastasis, and response to therapy. This paper highlights the current status of TM. Tract sources, adjacent normal tissue, circulatory system, and concomitant tumor co-metastasis are the main origin of TM. The advanced techniques in TM analysis are comprehensively summarized. Besides, TM is involved in tumor progression through several mechanisms, including DNA damage, activation of oncogenic signaling pathways (phosphoinositide 3-kinase [PI3K], signal transducer and activator of transcription [STAT], WNT/β-catenin, and extracellular regulated protein kinases [ERK]), influence of cytokines and induce inflammatory responses, and interaction with the tumor microenvironment (anti-tumor immunity, pro-tumor immunity, and microbial-derived metabolites). Moreover, promising directions of TM in tumor therapy include immunotherapy, chemotherapy, radiotherapy, the application of probiotics/prebiotics/synbiotics, fecal microbiome transplantation, engineered microbiota, phage therapy, and oncolytic virus therapy. The inherent challenges of clinical application are also summarized. This review provides a comprehensive landscape for analyzing TM, especially the TM-related mechanisms and TM-based treatment in cancer.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141859128","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Advances and clinical applications of immune checkpoint inhibitors in hematological malignancies. 免疫检查点抑制剂在血液恶性肿瘤中的进展和临床应用。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-28 DOI: 10.1002/cac2.12587
Wenyue Sun, Shunfeng Hu, Xin Wang

Immune checkpoints are differentially expressed on various immune cells to regulate immune responses in tumor microenvironment. Tumor cells can activate the immune checkpoint pathway to establish an immunosuppressive tumor microenvironment and inhibit the anti-tumor immune response, which may lead to tumor progression by evading immune surveillance. Interrupting co-inhibitory signaling pathways with immune checkpoint inhibitors (ICIs) could reinvigorate the anti-tumor immune response and promote immune-mediated eradication of tumor cells. As a milestone in tumor treatment, ICIs have been firstly used in solid tumors and subsequently expanded to hematological malignancies, which are in their infancy. Currently, immune checkpoints have been investigated as promising biomarkers and therapeutic targets in hematological malignancies, and novel immune checkpoints, such as signal regulatory protein α (SIRPα) and tumor necrosis factor-alpha-inducible protein 8-like 2 (TIPE2), are constantly being discovered. Numerous ICIs have received clinical approval for clinical application in the treatment of hematological malignancies, especially when used in combination with other strategies, including oncolytic viruses (OVs), neoantigen vaccines, bispecific antibodies (bsAb), bio-nanomaterials, tumor vaccines, and cytokine-induced killer (CIK) cells. Moreover, the proportion of individuals with hematological malignancies benefiting from ICIs remains lower than expected due to multiple mechanisms of drug resistance and immune-related adverse events (irAEs). Close monitoring and appropriate intervention are needed to mitigate irAEs while using ICIs. This review provided a comprehensive overview of immune checkpoints on different immune cells, the latest advances of ICIs and highlighted the clinical applications of immune checkpoints in hematological malignancies, including biomarkers, targets, combination of ICIs with other therapies, mechanisms of resistance to ICIs, and irAEs, which can provide novel insight into the future exploration of ICIs in tumor treatment.

免疫检查点在各种免疫细胞上不同程度地表达,以调节肿瘤微环境中的免疫反应。肿瘤细胞可激活免疫检查点通路,建立免疫抑制性肿瘤微环境,抑制抗肿瘤免疫反应,从而逃避免疫监视,导致肿瘤进展。利用免疫检查点抑制剂(ICIs)中断协同抑制信号通路,可以重振抗肿瘤免疫反应,促进免疫介导的肿瘤细胞清除。作为肿瘤治疗的里程碑,ICIs 首先用于实体瘤,随后扩展到处于起步阶段的血液恶性肿瘤。目前,免疫检查点已作为血液恶性肿瘤中很有前景的生物标记物和治疗靶点得到研究,新型免疫检查点,如信号调节蛋白α(SIRPα)和肿瘤坏死因子-α-诱导蛋白8样2(TIPE2),也在不断被发现。许多 ICIs 已获得临床批准,可用于血液恶性肿瘤的临床治疗,尤其是与其他策略联合使用时,包括溶瘤病毒(OV)、新抗原疫苗、双特异性抗体(bsAb)、生物纳米材料、肿瘤疫苗和细胞因子诱导的杀伤细胞(CIK)。此外,由于多种耐药机制和免疫相关不良事件(irAEs),血液恶性肿瘤患者从 ICIs 中获益的比例仍然低于预期。在使用 ICIs 时,需要进行密切监测和适当干预,以减轻 irAEs。本综述全面概述了不同免疫细胞上的免疫检查点、ICIs的最新进展,并重点介绍了免疫检查点在血液恶性肿瘤中的临床应用,包括生物标志物、靶点、ICIs与其他疗法的联合应用、ICIs的耐药机制和irAEs等,为今后探索ICIs在肿瘤治疗中的应用提供了新的启示。
{"title":"Advances and clinical applications of immune checkpoint inhibitors in hematological malignancies.","authors":"Wenyue Sun, Shunfeng Hu, Xin Wang","doi":"10.1002/cac2.12587","DOIUrl":"https://doi.org/10.1002/cac2.12587","url":null,"abstract":"<p><p>Immune checkpoints are differentially expressed on various immune cells to regulate immune responses in tumor microenvironment. Tumor cells can activate the immune checkpoint pathway to establish an immunosuppressive tumor microenvironment and inhibit the anti-tumor immune response, which may lead to tumor progression by evading immune surveillance. Interrupting co-inhibitory signaling pathways with immune checkpoint inhibitors (ICIs) could reinvigorate the anti-tumor immune response and promote immune-mediated eradication of tumor cells. As a milestone in tumor treatment, ICIs have been firstly used in solid tumors and subsequently expanded to hematological malignancies, which are in their infancy. Currently, immune checkpoints have been investigated as promising biomarkers and therapeutic targets in hematological malignancies, and novel immune checkpoints, such as signal regulatory protein α (SIRPα) and tumor necrosis factor-alpha-inducible protein 8-like 2 (TIPE2), are constantly being discovered. Numerous ICIs have received clinical approval for clinical application in the treatment of hematological malignancies, especially when used in combination with other strategies, including oncolytic viruses (OVs), neoantigen vaccines, bispecific antibodies (bsAb), bio-nanomaterials, tumor vaccines, and cytokine-induced killer (CIK) cells. Moreover, the proportion of individuals with hematological malignancies benefiting from ICIs remains lower than expected due to multiple mechanisms of drug resistance and immune-related adverse events (irAEs). Close monitoring and appropriate intervention are needed to mitigate irAEs while using ICIs. This review provided a comprehensive overview of immune checkpoints on different immune cells, the latest advances of ICIs and highlighted the clinical applications of immune checkpoints in hematological malignancies, including biomarkers, targets, combination of ICIs with other therapies, mechanisms of resistance to ICIs, and irAEs, which can provide novel insight into the future exploration of ICIs in tumor treatment.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141787311","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Abrogation of nuclear entry of TERT by fructose 1,6-bisphosphatase 1-mediated dephosphorylation. 果糖-1,6-二磷酸酶 1 介导的去磷酸化作用可抑制 TERT 进入细胞核。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-28 DOI: 10.1002/cac2.12599
Pengbo Yao, Gaoxiang Zhao, Min Li, Wensheng Qiu, Zhimin Lu
{"title":"Abrogation of nuclear entry of TERT by fructose 1,6-bisphosphatase 1-mediated dephosphorylation.","authors":"Pengbo Yao, Gaoxiang Zhao, Min Li, Wensheng Qiu, Zhimin Lu","doi":"10.1002/cac2.12599","DOIUrl":"https://doi.org/10.1002/cac2.12599","url":null,"abstract":"","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141787310","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
期刊
Cancer Communications
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