首页 > 最新文献

Cancer Communications最新文献

英文 中文
The regulatory roles and clinical significance of glycolysis in tumor 糖酵解在肿瘤中的调节作用和临床意义。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-06-08 DOI: 10.1002/cac2.12549
Qiqi Qiao, Shunfeng Hu, Xin Wang

Metabolic reprogramming has been demonstrated to have a significant impact on the biological behaviors of tumor cells, among which glycolysis is an important form. Recent research has revealed that the heightened glycolysis levels, the abnormal expression of glycolytic enzymes, and the accumulation of glycolytic products could regulate the growth, proliferation, invasion, and metastasis of tumor cells and provide a favorable microenvironment for tumor development and progression. Based on the distinctive glycolytic characteristics of tumor cells, novel imaging tests have been developed to evaluate tumor proliferation and metastasis. In addition, glycolytic enzymes have been found to serve as promising biomarkers in tumor, which could provide assistance in the early diagnosis and prognostic assessment of tumor patients. Numerous glycolytic enzymes have been identified as potential therapeutic targets for tumor treatment, and various small molecule inhibitors targeting glycolytic enzymes have been developed to inhibit tumor development and some of them are already applied in the clinic. In this review, we systematically summarized recent advances of the regulatory roles of glycolysis in tumor progression and highlighted the potential clinical significance of glycolytic enzymes and products as novel biomarkers and therapeutic targets in tumor treatment.

代谢重编程已被证明对肿瘤细胞的生物学行为有重大影响,而糖酵解是其中的一种重要形式。最新研究发现,糖酵解水平的升高、糖酵解酶的异常表达以及糖酵解产物的积累可调控肿瘤细胞的生长、增殖、侵袭和转移,并为肿瘤的发生和发展提供有利的微环境。基于肿瘤细胞独特的糖酵解特性,人们开发了新型成像检测方法来评估肿瘤的增殖和转移。此外,人们还发现糖酵解酶可作为肿瘤的生物标记物,有助于肿瘤患者的早期诊断和预后评估。许多糖酵解酶已被确定为治疗肿瘤的潜在靶点,针对糖酵解酶的各种小分子抑制剂已被开发出来以抑制肿瘤的发展,其中一些已应用于临床。在这篇综述中,我们系统地总结了糖酵解在肿瘤进展中的调控作用的最新进展,并强调了糖酵解酶及其产物作为新型生物标记物和治疗靶点在肿瘤治疗中的潜在临床意义。
{"title":"The regulatory roles and clinical significance of glycolysis in tumor","authors":"Qiqi Qiao,&nbsp;Shunfeng Hu,&nbsp;Xin Wang","doi":"10.1002/cac2.12549","DOIUrl":"10.1002/cac2.12549","url":null,"abstract":"<p>Metabolic reprogramming has been demonstrated to have a significant impact on the biological behaviors of tumor cells, among which glycolysis is an important form. Recent research has revealed that the heightened glycolysis levels, the abnormal expression of glycolytic enzymes, and the accumulation of glycolytic products could regulate the growth, proliferation, invasion, and metastasis of tumor cells and provide a favorable microenvironment for tumor development and progression. Based on the distinctive glycolytic characteristics of tumor cells, novel imaging tests have been developed to evaluate tumor proliferation and metastasis. In addition, glycolytic enzymes have been found to serve as promising biomarkers in tumor, which could provide assistance in the early diagnosis and prognostic assessment of tumor patients. Numerous glycolytic enzymes have been identified as potential therapeutic targets for tumor treatment, and various small molecule inhibitors targeting glycolytic enzymes have been developed to inhibit tumor development and some of them are already applied in the clinic. In this review, we systematically summarized recent advances of the regulatory roles of glycolysis in tumor progression and highlighted the potential clinical significance of glycolytic enzymes and products as novel biomarkers and therapeutic targets in tumor treatment.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-06-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11260772/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141293127","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Exposure of benzo[a]pyrene induces HCC exosome-circular RNA to activate lung fibroblasts and trigger organotropic metastasis 接触苯并[a]芘会诱导 HCC 外泌体环状 RNA 激活肺成纤维细胞并引发器官转移。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-06-05 DOI: 10.1002/cac2.12574
Wei Mu, Pengfei Gu, Huating Li, Jinjin Zhou, Yulun Jian, Weiping Jia, Yang Ge

Background

Benzo[a]pyrene (B[a]P), a carcinogen pollutant produced by combustion processes, is present in the western diet with grilled meats. Chronic exposure of B[a]P in hepatocellular carcinoma (HCC) cells promotes metastasis rather than primary proliferation, implying an unknown mechanism of B[a]P-induced malignancy. Given that exosomes carry bioactive molecules to distant sites, we investigated whether and how exosomes mediate cancer-stroma communications for a toxicologically associated microenvironment.

Method

Exosomes were isolated from B[a]P stimulated BEL7404 HCC cells (7404-100Bap Exo) at an environmental relevant dose (100 nmol/L). Lung pre-education animal model was prepared via injection of exosomes and cytokines. The inflammatory genes of educated lungs were evaluated using quantitative reverse transcription PCR array. HCC LM3 cells transfected with firefly luciferase were next injected to monitor tumor burdens and organotropic metastasis. Profile of B[a]P-exposed exosomes were determined by ceRNA microarray. Interactions between circular RNA (circRNA) and microRNAs (miRNAs) were detected using RNA pull-down in target lung fibroblasts. Fluorescence in situ hybridization and RNA immunoprecipitation assay was used to evaluate the “on-off” interaction of circRNA-miRNA pairs. We further developed an adeno-associated virus inhalation model to examine mRNA expression specific in lung, thereby exploring the mRNA targets of B[a]P induced circRNA-miRNA cascade.

Results

Lung fibroblasts exert activation phenotypes, including focal adhesion and motility were altered by 7404-100Bap Exo. In the exosome-educated in vivo model, fibrosis factors and pro-inflammatory molecules of are up-regulated when injected with exosomes. Compared to non-exposed 7404 cells, circ_0011496 was up-regulated following B[a]P treatment and was mainly packaged into 7404-100Bap Exo. Exosomal circ_0011496 were delivered and competitively bound to miR-486-5p in recipient fibroblasts. The down-regulation of miR-486-5p converted fibroblast to cancer-associated fibroblast via regulating the downstream of Twinfilin-1 (TWF1) and matrix metalloproteinase-9 (MMP9) cascade. Additionally, increased TWF1, specifically in exosomal circ_0011496 educated lungs, could promote cancer-stroma crosstalk via activating vascular endothelial growth factor (VEGF). These modulated fibroblasts promoted endothelial cells angiogenesis and recruited primary HCC cells invasion, as a consequence of a pre-metastatic niche formation.

背景:苯并[a]芘(B[a]P)是燃烧过程中产生的一种致癌污染物,存在于西方饮食中的烤肉中。肝细胞癌(HCC)细胞长期暴露于 B[a]P 会促进转移而非原发增殖,这意味着 B[a]P 诱导恶性肿瘤的机制不明。鉴于外泌体可将生物活性分子携带至远处,我们研究了外泌体是否以及如何介导癌症与肿瘤之间的沟通,以形成与毒性相关的微环境:方法:从B[a]P刺激的BEL7404 HCC细胞(7404-100Bap Exo)中分离出外泌体,其剂量为环境相关剂量(100 nmol/L)。通过注射外泌体和细胞因子制备了肺预教育动物模型。使用定量反转录 PCR 阵列评估受教育肺部的炎症基因。然后注射转染了萤火虫荧光素酶的 HCC LM3 细胞,以监测肿瘤负荷和器官转移。通过ceRNA芯片确定了B[a]P暴露的外泌体的特征。在靶肺成纤维细胞中使用 RNA pull-down 方法检测环状 RNA(circRNA)和 microRNA(miRNA)之间的相互作用。荧光原位杂交和 RNA 免疫沉淀试验被用来评估 circRNA-miRNA 对的 "开关 "相互作用。我们进一步开发了腺相关病毒吸入模型来检测肺部特定的mRNA表达,从而探索B[a]P诱导的circRNA-miRNA级联的mRNA靶标:结果:7404-100Bap外泌体改变了肺成纤维细胞的活化表型,包括病灶粘附和运动。在外泌体教育的体内模型中,注射外泌体后,纤维化因子和促炎分子上调。与未接触外泌体的7404细胞相比,circ_0011496在B[a]P处理后上调,并主要被包装到7404-100Bap外泌体中。外泌体circ_0011496被递送并与受体成纤维细胞中的miR-486-5p竞争性结合。miR-486-5p的下调通过调节Twinfilin-1(TWF1)下游和基质金属蛋白酶-9(MMP9)级联,使成纤维细胞转化为癌相关成纤维细胞。此外,TWF1的增加,特别是在外泌体circ_0011496教育肺中的增加,可通过激活血管内皮生长因子(VEGF)促进癌症-基质串联。这些被调控的成纤维细胞促进了内皮细胞血管生成,并招募原发性 HCC 细胞入侵,这是转移前生态位形成的结果:我们证明,B[a]P诱导的肿瘤外泌体可传递circ_0011496,激活肺成纤维细胞中的miR-486-5p/TWF1/MMP9级联,产生促进HCC转移的反馈回路。
{"title":"Exposure of benzo[a]pyrene induces HCC exosome-circular RNA to activate lung fibroblasts and trigger organotropic metastasis","authors":"Wei Mu,&nbsp;Pengfei Gu,&nbsp;Huating Li,&nbsp;Jinjin Zhou,&nbsp;Yulun Jian,&nbsp;Weiping Jia,&nbsp;Yang Ge","doi":"10.1002/cac2.12574","DOIUrl":"10.1002/cac2.12574","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <h3> Background</h3>\u0000 \u0000 <p>Benzo[a]pyrene (B[a]P), a carcinogen pollutant produced by combustion processes, is present in the western diet with grilled meats. Chronic exposure of B[a]P in hepatocellular carcinoma (HCC) cells promotes metastasis rather than primary proliferation, implying an unknown mechanism of B[a]P-induced malignancy. Given that exosomes carry bioactive molecules to distant sites, we investigated whether and how exosomes mediate cancer-stroma communications for a toxicologically associated microenvironment.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Method</h3>\u0000 \u0000 <p>Exosomes were isolated from B[a]P stimulated BEL7404 HCC cells (7404-100Bap Exo) at an environmental relevant dose (100 nmol/L). Lung pre-education animal model was prepared via injection of exosomes and cytokines. The inflammatory genes of educated lungs were evaluated using quantitative reverse transcription PCR array. HCC LM3 cells transfected with firefly luciferase were next injected to monitor tumor burdens and organotropic metastasis. Profile of B[a]P-exposed exosomes were determined by ceRNA microarray. Interactions between circular RNA (circRNA) and microRNAs (miRNAs) were detected using RNA pull-down in target lung fibroblasts. Fluorescence in situ hybridization and RNA immunoprecipitation assay was used to evaluate the “on-off” interaction of circRNA-miRNA pairs. We further developed an adeno-associated virus inhalation model to examine mRNA expression specific in lung, thereby exploring the mRNA targets of B[a]P induced circRNA-miRNA cascade.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Results</h3>\u0000 \u0000 <p>Lung fibroblasts exert activation phenotypes, including focal adhesion and motility were altered by 7404-100Bap Exo. In the exosome-educated in vivo model, fibrosis factors and pro-inflammatory molecules of are up-regulated when injected with exosomes. Compared to non-exposed 7404 cells, circ_0011496 was up-regulated following B[a]P treatment and was mainly packaged into 7404-100Bap Exo. Exosomal circ_0011496 were delivered and competitively bound to miR-486-5p in recipient fibroblasts. The down-regulation of miR-486-5p converted fibroblast to cancer-associated fibroblast via regulating the downstream of Twinfilin-1 (TWF1) and matrix metalloproteinase-9 (MMP9) cascade. Additionally, increased TWF1, specifically in exosomal circ_0011496 educated lungs, could promote cancer-stroma crosstalk via activating vascular endothelial growth factor (VEGF). These modulated fibroblasts promoted endothelial cells angiogenesis and recruited primary HCC cells invasion, as a consequence of a pre-metastatic niche formation.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 ","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11260768/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141261131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Beyond success: unveiling the hidden potential of radiotherapy and immunotherapy in solid tumors 超越成功:发掘放射治疗和免疫治疗在实体瘤中的潜能
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-06-05 DOI: 10.1002/cac2.12576
Yuze Wu, Ming Yi, Mengke Niu, Binghan Zhou, Qi Mei, Kongming Wu

Immunotherapy, particularly with immune checkpoint inhibitors, has significantly transformed cancer treatment. Despite its success, many patients struggle to respond adequately or sustain long-lasting clinical improvement. A growing consensus has emerged that radiotherapy (RT) enhances the response rate and overall efficacy of immunotherapy. Although combining RT and immunotherapy has been extensively investigated in preclinical models and has shown promising results, establishing itself as a dynamic and thriving area of research, clinical evidence for this combination strategy over the past five years has shown both positive and disappointing results, suggesting the need for a more nuanced understanding. This review provides a balanced and updated analysis of the combination of immunotherapy and RT. We summarized the preclinical mechanisms through which RT boosts antitumor immune responses and mainly focused on the outcomes of recently updated clinical trials, including those that may not have met expectations. We investigated the optimization of the therapeutic potential of this combined strategy, including key challenges, such as fractionation and scheduling, lymph node irradiation, and toxicity. Finally, we offered insights into the prospects and challenges associated with the clinical translation of this combination therapy, providing a realistic perspective on the current state of research and potential future directions.

免疫疗法,尤其是免疫检查点抑制剂,极大地改变了癌症治疗。尽管免疫疗法取得了成功,但许多患者仍难以获得充分的反应或持久的临床改善。放疗(RT)可提高免疫疗法的反应率和总体疗效,这一点已逐渐成为共识。尽管在临床前模型中已对 RT 与免疫疗法的结合进行了广泛研究,并取得了可喜的成果,使其成为一个充满活力、蓬勃发展的研究领域,但在过去五年中,这种结合策略的临床证据既显示出积极的结果,也显示出令人失望的结果,这表明我们需要对其有更细致的了解。本综述对免疫疗法和 RT 的联合应用进行了均衡的最新分析。我们总结了 RT 促进抗肿瘤免疫反应的临床前机制,并主要关注最近更新的临床试验结果,包括那些可能未达到预期的结果。我们研究了如何优化这种联合策略的治疗潜力,包括分次和时间安排、淋巴结照射和毒性等关键挑战。最后,我们对这种联合疗法的临床转化前景和挑战提出了见解,为当前的研究状况和未来的潜在方向提供了现实的视角。
{"title":"Beyond success: unveiling the hidden potential of radiotherapy and immunotherapy in solid tumors","authors":"Yuze Wu,&nbsp;Ming Yi,&nbsp;Mengke Niu,&nbsp;Binghan Zhou,&nbsp;Qi Mei,&nbsp;Kongming Wu","doi":"10.1002/cac2.12576","DOIUrl":"10.1002/cac2.12576","url":null,"abstract":"<p>Immunotherapy, particularly with immune checkpoint inhibitors, has significantly transformed cancer treatment. Despite its success, many patients struggle to respond adequately or sustain long-lasting clinical improvement. A growing consensus has emerged that radiotherapy (RT) enhances the response rate and overall efficacy of immunotherapy. Although combining RT and immunotherapy has been extensively investigated in preclinical models and has shown promising results, establishing itself as a dynamic and thriving area of research, clinical evidence for this combination strategy over the past five years has shown both positive and disappointing results, suggesting the need for a more nuanced understanding. This review provides a balanced and updated analysis of the combination of immunotherapy and RT. We summarized the preclinical mechanisms through which RT boosts antitumor immune responses and mainly focused on the outcomes of recently updated clinical trials, including those that may not have met expectations. We investigated the optimization of the therapeutic potential of this combined strategy, including key challenges, such as fractionation and scheduling, lymph node irradiation, and toxicity. Finally, we offered insights into the prospects and challenges associated with the clinical translation of this combination therapy, providing a realistic perspective on the current state of research and potential future directions.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12576","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141258171","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Long-term survival outcomes and immune checkpoint inhibitor retreatment in patients with advanced cervical cancer treated with camrelizumab plus apatinib in the phase II CLAP study 在 CLAP II 期研究中,接受坎瑞珠单抗加阿帕替尼治疗的晚期宫颈癌患者的长期生存结果和免疫检查点抑制剂再治疗情况。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-05-13 DOI: 10.1002/cac2.12547
Chunyan Lan, Huaiwu Lu, Lin Zhou, Kunlun Liao, Junxiu Liu, Zhiwen Xie, Haixi Liang, Guorong Zou, Ting Yang, Qin Xu, Xin Huang

Background

Camrelizumab plus apatinib have demonstrated robust antitumor activity and safety in patients with advanced cervical cancer (CLAP study; NCT03816553). We herein present the updated long-term results of the CLAP study and explore potential biomarkers for survival. The outcomes of patients who underwent immune checkpoint inhibitor (ICI) retreatment were also reported.

Methods

In this phase II trial, eligible patients received camrelizumab 200 mg intravenously every two weeks and apatinib 250 mg orally once daily in 4-week cycles for up to two years. Treatment was continued until disease progression, unacceptable toxicity, or withdrawal of consent.

Results

Between January 21 and August 1, 2019, a total of 45 patients were enrolled. Data were analyzed as of July 31, 2023, representing > 48 months since treatment initiation for all patients. Nine (20.0%) patients completed the 2-year study. The median duration of response (DOR) was 16.6 months, and 45.0% of patients achieved a DOR of ≥ 24 months. The 12-month progression-free survival (PFS) rate was 40.7% (95% confidence interval [CI], 25.2-55.6), with an 18-month PFS rate of 37.8% (95% CI, 22.7-52.8). The median overall survival (OS) was 20.3 months (95% CI, 9.3-36.9), and the 24-month OS rate was 47.8% (95% CI, 31.7-62.3). Age > 50 years, programmed death-ligand 1 (PD-L1) combined positive score (CPS) ≥ 1 (versus [vs.] < 1), CPS ≥ 10 (vs. < 1), high tumor mutational burden, and PIK3CA mutations were associated with improved PFS (hazard ratio [HR] < 1) and longer OS (HR < 1). Eight patients who initially responded in the CLAP trial but later experienced disease progression were retreated with ICIs. Among them, 2 (25.0%) achieved a partial response, while 5 (62.5%) had stable disease. Notably, four patients who received retreatment with ICIs survived for more than 45 months. No new safety signals were identified in the present study.

Conclusion

Long-term survival follow-up data demonstrated that camrelizumab plus apatinib has robust, sustained, and durable efficacy in patients with advanced cervical cancer who progress after first-line platinum-based chemotherapy. No new safety signals were noted with long-term treatment.

研究背景康瑞珠单抗联合阿帕替尼已在晚期宫颈癌患者中显示出强大的抗肿瘤活性和安全性(CLAP 研究;NCT03816553)。我们在此介绍 CLAP 研究的最新长期结果,并探讨潜在的生存生物标志物。同时还报告了接受免疫检查点抑制剂(ICI)再治疗的患者的结果:在这项II期试验中,符合条件的患者每两周静脉注射一次康瑞珠单抗(camrelizumab)200毫克,同时口服阿帕替尼250毫克,每天一次,4周为一个周期,疗程最长为两年。治疗一直持续到疾病进展、出现不可接受的毒性或撤回同意为止:2019年1月21日至8月1日期间,共有45名患者入组。对截至 2023 年 7 月 31 日的数据进行了分析,所有患者的治疗时间均大于 48 个月。9名患者(20.0%)完成了为期2年的研究。中位应答持续时间(DOR)为16.6个月,45.0%的患者达到了≥24个月的DOR。12个月无进展生存期(PFS)率为40.7%(95%置信区间[CI],25.2-55.6),18个月PFS率为37.8%(95%置信区间[CI],22.7-52.8)。中位总生存期(OS)为20.3个月(95% CI,9.3-36.9),24个月的OS率为47.8%(95% CI,31.7-62.3)。年龄大于50岁、程序性死亡配体1(PD-L1)联合阳性评分(CPS)≥1(与[vs.]<1)、CPS≥10(与[vs.]<1)、高肿瘤突变负荷和PIK3CA突变与PFS改善(危险比[HR]<1)和OS延长(HR<1)相关。在CLAP试验中,有8名患者起初有反应,但后来出现了疾病进展,他们接受了ICIs治疗。其中,2 人(25.0%)获得部分应答,5 人(62.5%)病情稳定。值得注意的是,4 名接受 ICIs 治疗的患者存活时间超过 45 个月。本研究未发现新的安全信号:长期生存随访数据表明,康瑞珠单抗联合阿帕替尼对一线铂类化疗后病情进展的晚期宫颈癌患者具有稳健、持续和持久的疗效。长期治疗未发现新的安全性信号。
{"title":"Long-term survival outcomes and immune checkpoint inhibitor retreatment in patients with advanced cervical cancer treated with camrelizumab plus apatinib in the phase II CLAP study","authors":"Chunyan Lan,&nbsp;Huaiwu Lu,&nbsp;Lin Zhou,&nbsp;Kunlun Liao,&nbsp;Junxiu Liu,&nbsp;Zhiwen Xie,&nbsp;Haixi Liang,&nbsp;Guorong Zou,&nbsp;Ting Yang,&nbsp;Qin Xu,&nbsp;Xin Huang","doi":"10.1002/cac2.12547","DOIUrl":"10.1002/cac2.12547","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <h3> Background</h3>\u0000 \u0000 <p>Camrelizumab plus apatinib have demonstrated robust antitumor activity and safety in patients with advanced cervical cancer (CLAP study; NCT03816553). We herein present the updated long-term results of the CLAP study and explore potential biomarkers for survival. The outcomes of patients who underwent immune checkpoint inhibitor (ICI) retreatment were also reported.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Methods</h3>\u0000 \u0000 <p>In this phase II trial, eligible patients received camrelizumab 200 mg intravenously every two weeks and apatinib 250 mg orally once daily in 4-week cycles for up to two years. Treatment was continued until disease progression, unacceptable toxicity, or withdrawal of consent.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Results</h3>\u0000 \u0000 <p>Between January 21 and August 1, 2019, a total of 45 patients were enrolled. Data were analyzed as of July 31, 2023, representing &gt; 48 months since treatment initiation for all patients. Nine (20.0%) patients completed the 2-year study. The median duration of response (DOR) was 16.6 months, and 45.0% of patients achieved a DOR of ≥ 24 months. The 12-month progression-free survival (PFS) rate was 40.7% (95% confidence interval [CI], 25.2-55.6), with an 18-month PFS rate of 37.8% (95% CI, 22.7-52.8). The median overall survival (OS) was 20.3 months (95% CI, 9.3-36.9), and the 24-month OS rate was 47.8% (95% CI, 31.7-62.3). Age &gt; 50 years, programmed death-ligand 1 (PD-L1) combined positive score (CPS) ≥ 1 (versus [vs.] &lt; 1), CPS ≥ 10 (vs. &lt; 1), high tumor mutational burden, and <i>PIK3CA</i> mutations were associated with improved PFS (hazard ratio [HR] &lt; 1) and longer OS (HR &lt; 1). Eight patients who initially responded in the CLAP trial but later experienced disease progression were retreated with ICIs. Among them, 2 (25.0%) achieved a partial response, while 5 (62.5%) had stable disease. Notably, four patients who received retreatment with ICIs survived for more than 45 months. No new safety signals were identified in the present study.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Conclusion</h3>\u0000 \u0000 <p>Long-term survival follow-up data demonstrated that camrelizumab plus apatinib has robust, sustained, and durable efficacy in patients with advanced cervical cancer who progress after first-line platinum-based chemotherapy. No new safety signals were noted with long-term treatment.</p>\u0000 </section>\u0000 </div>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11194449/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140915872","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Protein tyrosine phosphatases: emerging role in cancer therapy resistance 蛋白酪氨酸磷酸酶:在抗癌治疗中的新角色。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-05-13 DOI: 10.1002/cac2.12548
Min Zhao, Wen Shuai, Zehao Su, Ping Xu, Aoxue Wang, Qiu Sun, Guan Wang

Background

Tyrosine phosphorylation of intracellular proteins is a post-translational modification that plays a regulatory role in signal transduction during cellular events. Dephosphorylation of signal transduction proteins caused by protein tyrosine phosphatases (PTPs) contributed their role as a convergent node to mediate cross-talk between signaling pathways. In the context of cancer, PTP-mediated pathways have been identified as signaling hubs that enabled cancer cells to mitigate stress induced by clinical therapy. This is achieved by the promotion of constitutive activation of growth-stimulatory signaling pathways or modulation of the immune-suppressive tumor microenvironment. Preclinical evidences suggested that anticancer drugs will release their greatest therapeutic potency when combined with PTP inhibitors, reversing drug resistance that was responsible for clinical failures during cancer therapy.

Areas covered

This review aimed to elaborate recent insights that supported the involvement of PTP-mediated pathways in the development of resistance to targeted therapy and immune-checkpoint therapy.

Expert opinion

This review proposed the notion of PTP inhibition in anticancer combination therapy as a potential strategy in clinic to achieve long-term tumor regression. Ongoing clinical trials are currently underway to assess the safety and efficacy of combination therapy in advanced-stage tumors.

背景:细胞内蛋白质的酪氨酸磷酸化是一种翻译后修饰,在细胞事件的信号转导过程中发挥着调节作用。蛋白酪氨酸磷酸酶(PTPs)引起的信号转导蛋白的去磷酸化有助于它们发挥聚合节点的作用,介导信号通路之间的交叉对话。在癌症方面,PTP 介导的通路已被确定为信号枢纽,可使癌细胞减轻临床治疗引起的压力。这是通过促进生长刺激信号通路的组成性激活或调节免疫抑制性肿瘤微环境来实现的。临床前证据表明,抗癌药物与 PTP 抑制剂联合使用时将释放出最大的治疗效力,从而逆转导致癌症治疗临床失败的耐药性:本综述旨在阐述支持 PTP 介导的通路参与靶向疗法和免疫检查点疗法耐药性产生的最新观点:这篇综述提出了在抗癌联合疗法中抑制PTP的概念,将其作为实现长期肿瘤消退的潜在临床策略。目前正在进行临床试验,以评估联合疗法在晚期肿瘤中的安全性和有效性。
{"title":"Protein tyrosine phosphatases: emerging role in cancer therapy resistance","authors":"Min Zhao,&nbsp;Wen Shuai,&nbsp;Zehao Su,&nbsp;Ping Xu,&nbsp;Aoxue Wang,&nbsp;Qiu Sun,&nbsp;Guan Wang","doi":"10.1002/cac2.12548","DOIUrl":"10.1002/cac2.12548","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <h3> Background</h3>\u0000 \u0000 <p>Tyrosine phosphorylation of intracellular proteins is a post-translational modification that plays a regulatory role in signal transduction during cellular events. Dephosphorylation of signal transduction proteins caused by protein tyrosine phosphatases (PTPs) contributed their role as a convergent node to mediate cross-talk between signaling pathways. In the context of cancer, PTP-mediated pathways have been identified as signaling hubs that enabled cancer cells to mitigate stress induced by clinical therapy. This is achieved by the promotion of constitutive activation of growth-stimulatory signaling pathways or modulation of the immune-suppressive tumor microenvironment. Preclinical evidences suggested that anticancer drugs will release their greatest therapeutic potency when combined with PTP inhibitors, reversing drug resistance that was responsible for clinical failures during cancer therapy.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Areas covered</h3>\u0000 \u0000 <p>This review aimed to elaborate recent insights that supported the involvement of PTP-mediated pathways in the development of resistance to targeted therapy and immune-checkpoint therapy.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Expert opinion</h3>\u0000 \u0000 <p>This review proposed the notion of PTP inhibition in anticancer combination therapy as a potential strategy in clinic to achieve long-term tumor regression. Ongoing clinical trials are currently underway to assess the safety and efficacy of combination therapy in advanced-stage tumors.</p>\u0000 </section>\u0000 </div>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11194456/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140915877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Metabolic landscape of head and neck squamous cell carcinoma informs a novel kynurenine/Siglec-15 axis in immune escape 头颈部鳞状细胞癌的代谢状况揭示了免疫逃逸中的新型犬尿氨酸/Siglec-15轴。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-05-12 DOI: 10.1002/cac2.12545
Xin-Yu Zhang, Jian-Bo Shi, Shu-Fang Jin, Rui-Jie Wang, Ming-Yu Li, Zhi-Yuan Zhang, Xi Yang, Hai-Long Ma

Background

Metabolic reprograming and immune escape are two hallmarks of cancer. However, how metabolic disorders drive immune escape in head and neck squamous cell carcinoma (HNSCC) remains unclear. Therefore, the aim of the present study was to investigate the metabolic landscape of HNSCC and its mechanism of driving immune escape.

Methods

Analysis of paired tumor tissues and adjacent normal tissues from 69 HNSCC patients was performed using liquid/gas chromatography-mass spectrometry and RNA-sequencing. The tumor-promoting function of kynurenine (Kyn) was explored in vitro and in vivo. The downstream target of Kyn was investigated in CD8+ T cells. The regulation of CD8+ T cells was investigated after Siglec-15 overexpression in vivo. An engineering nanoparticle was established to deliver Siglec-15 small interfering RNA (siS15), and its association with immunotherapy response were investigated. The association between Siglec-15 and CD8+ programmed cell death 1 (PD-1)+ T cells was analyzed in a HNSCC patient cohort.

Results

A total of 178 metabolites showed significant dysregulation in HNSCC, including carbohydrates, lipids and lipid-like molecules, and amino acids. Among these, amino acid metabolism was the most significantly altered, especially Kyn, which promoted tumor proliferation and metastasis. In addition, most immune checkpoint molecules were upregulated in Kyn-high patients based on RNA-sequencing. Furthermore, tumor-derived Kyn was transferred into CD8+ T cells and induced T cell functional exhaustion, and blocking Kyn transporters restored its killing activity. Accroding to the results, mechanistically, Kyn transcriptionally regulated the expression of Siglec-15 via aryl hydrocarbon receptor (AhR), and overexpression of Siglec-15 promoted immune escape by suppressing T cell infiltration and activation. Targeting AhR in vivo reduced Kyn-mediated Siglec-15 expression and promoted intratumoral CD8+ T cell infiltration and killing capacity. Finally, a NH2-modified mesoporous silica nanoparticle was designed to deliver siS15, which restored CD8+ T cell function status and enhanced anti-PD-1 efficacy in tumor-bearing immunocompetent mice. Clinically, Siglec-15 was positively correlated with AhR expression and CD8+PD-1+ T cell infiltration in HNSCC tissues.

Conclusions

The findings describe the

背景:代谢重编程和免疫逃逸是癌症的两大特征。然而,代谢紊乱如何驱动头颈部鳞状细胞癌(HNSCC)的免疫逃逸仍不清楚。因此,本研究旨在调查 HNSCC 的代谢情况及其驱动免疫逃逸的机制:方法:采用液/气相色谱-质谱法和 RNA 序列分析法对 69 例 HNSCC 患者的配对肿瘤组织和邻近正常组织进行分析。在体外和体内探讨了犬尿氨酸(Kyn)的促瘤功能。在 CD8+ T 细胞中研究了 Kyn 的下游靶标。研究了 Siglec-15 在体内过表达后对 CD8+ T 细胞的调控。建立了一种工程纳米粒子来递送 Siglec-15 小干扰 RNA(siS15),并研究了其与免疫治疗反应的关系。在HNSCC患者队列中分析了Siglec-15与CD8+程序性细胞死亡1(PD-1)+ T细胞之间的关联:结果:共有178种代谢物在HNSCC中出现明显失调,包括碳水化合物、脂类和类脂分子以及氨基酸。其中,氨基酸代谢的改变最为明显,尤其是Kyn,它能促进肿瘤的增殖和转移。此外,根据 RNA 序列分析,Kyn 高的患者体内大多数免疫检查点分子都出现了上调。此外,肿瘤衍生的Kyn被转移到CD8+ T细胞中,诱导T细胞功能衰竭,而阻断Kyn转运体可恢复其杀伤活性。研究结果表明,从机理上讲,Kyn通过芳基烃受体(AhR)转录调节Siglec-15的表达,而Siglec-15的过表达会抑制T细胞的浸润和活化,从而促进免疫逃逸。在体内以AhR为靶点可减少Kyn介导的Siglec-15表达,促进瘤内CD8+ T细胞浸润和杀伤能力。最后,我们设计了一种 NH2 修饰介孔二氧化硅纳米粒子来递送 siS15,从而恢复了 CD8+ T 细胞的功能状态,并增强了肿瘤免疫功能健全小鼠的抗 PD-1 疗效。在临床上,Siglec-15与HNSCC组织中的AhR表达和CD8+PD-1+ T细胞浸润呈正相关:结论:研究结果全面描述了 HNSCC 的代谢情况,揭示了 Kyn/Siglec-15 轴可能是一种新的潜在免疫代谢机制,为癌症的治疗提供了一种前景广阔的策略。
{"title":"Metabolic landscape of head and neck squamous cell carcinoma informs a novel kynurenine/Siglec-15 axis in immune escape","authors":"Xin-Yu Zhang,&nbsp;Jian-Bo Shi,&nbsp;Shu-Fang Jin,&nbsp;Rui-Jie Wang,&nbsp;Ming-Yu Li,&nbsp;Zhi-Yuan Zhang,&nbsp;Xi Yang,&nbsp;Hai-Long Ma","doi":"10.1002/cac2.12545","DOIUrl":"10.1002/cac2.12545","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <h3> Background</h3>\u0000 \u0000 <p>Metabolic reprograming and immune escape are two hallmarks of cancer. However, how metabolic disorders drive immune escape in head and neck squamous cell carcinoma (HNSCC) remains unclear. Therefore, the aim of the present study was to investigate the metabolic landscape of HNSCC and its mechanism of driving immune escape.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Methods</h3>\u0000 \u0000 <p>Analysis of paired tumor tissues and adjacent normal tissues from 69 HNSCC patients was performed using liquid/gas chromatography-mass spectrometry and RNA-sequencing. The tumor-promoting function of kynurenine (Kyn) was explored in vitro and in vivo. The downstream target of Kyn was investigated in CD8<sup>+</sup> T cells. The regulation of CD8<sup>+</sup> T cells was investigated after Siglec-15 overexpression in vivo. An engineering nanoparticle was established to deliver Siglec-15 small interfering RNA (siS15), and its association with immunotherapy response were investigated. The association between Siglec-15 and CD8<sup>+</sup> programmed cell death 1 (PD-1)<sup>+</sup> T cells was analyzed in a HNSCC patient cohort.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Results</h3>\u0000 \u0000 <p>A total of 178 metabolites showed significant dysregulation in HNSCC, including carbohydrates, lipids and lipid-like molecules, and amino acids. Among these, amino acid metabolism was the most significantly altered, especially Kyn, which promoted tumor proliferation and metastasis. In addition, most immune checkpoint molecules were upregulated in Kyn-high patients based on RNA-sequencing. Furthermore, tumor-derived Kyn was transferred into CD8<sup>+</sup> T cells and induced T cell functional exhaustion, and blocking Kyn transporters restored its killing activity. Accroding to the results, mechanistically, Kyn transcriptionally regulated the expression of Siglec-15 via aryl hydrocarbon receptor (AhR), and overexpression of Siglec-15 promoted immune escape by suppressing T cell infiltration and activation. Targeting AhR in vivo reduced Kyn-mediated Siglec-15 expression and promoted intratumoral CD8<sup>+</sup> T cell infiltration and killing capacity. Finally, a NH<sub>2</sub>-modified mesoporous silica nanoparticle was designed to deliver siS15, which restored CD8<sup>+</sup> T cell function status and enhanced anti-PD-1 efficacy in tumor-bearing immunocompetent mice. Clinically, Siglec-15 was positively correlated with AhR expression and CD8<sup>+</sup>PD-1<sup>+</sup> T cell infiltration in HNSCC tissues.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Conclusions</h3>\u0000 \u0000 <p>The findings describe the","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11194450/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140907967","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Converting “cold” to “hot”: epigenetics strategies to improve immune therapy effect by regulating tumor-associated immune suppressive cells 化 "冷 "为 "热":通过调节肿瘤相关免疫抑制细胞提高免疫治疗效果的表观遗传学策略。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-05-07 DOI: 10.1002/cac2.12546
Yijia Tang, Guangzu Cui, Haicong Liu, Ying Han, Changjing Cai, Ziyang Feng, Hong Shen, Shan Zeng

Significant developments in cancer treatment have been made since the advent of immune therapies. However, there are still some patients with malignant tumors who do not benefit from immunotherapy. Tumors without immunogenicity are called “cold” tumors which are unresponsive to immunotherapy, and the opposite are “hot” tumors. Immune suppressive cells (ISCs) refer to cells which can inhibit the immune response such as tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), regulatory T (Treg) cells and so on. The more ISCs infiltrated, the weaker the immunogenicity of the tumor, showing the characteristics of “cold” tumor. The dysfunction of ISCs in the tumor microenvironment (TME) may play essential roles in insensitive therapeutic reaction. Previous studies have found that epigenetic mechanisms play an important role in the regulation of ISCs. Regulating ISCs may be a new approach to transforming “cold” tumors into “hot” tumors. Here, we focused on the function of ISCs in the TME and discussed how epigenetics is involved in regulating ISCs. In addition, we summarized the mechanisms by which the epigenetic drugs convert immunotherapy-insensitive tumors into immunotherapy-sensitive tumors which would be an innovative tendency for future immunotherapy in “cold” tumor.

自免疫疗法出现以来,癌症治疗取得了重大进展。然而,仍有一些恶性肿瘤患者无法从免疫疗法中获益。没有免疫原性的肿瘤被称为对免疫疗法无反应的 "冷 "肿瘤,反之则为 "热 "肿瘤。免疫抑制细胞(ISCs)是指能够抑制免疫反应的细胞,如肿瘤相关巨噬细胞(TAMs)、髓源抑制细胞(MDSCs)、调节性T细胞(Treg)等。ISCs浸润越多,肿瘤的免疫原性越弱,呈现出 "冷 "肿瘤的特征。肿瘤微环境(TME)中 ISCs 的功能失调可能是导致治疗反应不敏感的重要原因。以往的研究发现,表观遗传机制在调控 ISCs 方面发挥着重要作用。调控ISCs可能是将 "冷 "肿瘤转化为 "热 "肿瘤的一种新方法。在此,我们重点探讨了ISCs在TME中的功能,并讨论了表观遗传学是如何参与调控ISCs的。此外,我们还总结了表观遗传学药物将免疫治疗不敏感肿瘤转化为免疫治疗敏感肿瘤的机制,这将是未来 "冷 "肿瘤免疫治疗的创新趋势。
{"title":"Converting “cold” to “hot”: epigenetics strategies to improve immune therapy effect by regulating tumor-associated immune suppressive cells","authors":"Yijia Tang,&nbsp;Guangzu Cui,&nbsp;Haicong Liu,&nbsp;Ying Han,&nbsp;Changjing Cai,&nbsp;Ziyang Feng,&nbsp;Hong Shen,&nbsp;Shan Zeng","doi":"10.1002/cac2.12546","DOIUrl":"10.1002/cac2.12546","url":null,"abstract":"<p>Significant developments in cancer treatment have been made since the advent of immune therapies. However, there are still some patients with malignant tumors who do not benefit from immunotherapy. Tumors without immunogenicity are called “cold” tumors which are unresponsive to immunotherapy, and the opposite are “hot” tumors. Immune suppressive cells (ISCs) refer to cells which can inhibit the immune response such as tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), regulatory T (Treg) cells and so on. The more ISCs infiltrated, the weaker the immunogenicity of the tumor, showing the characteristics of “cold” tumor. The dysfunction of ISCs in the tumor microenvironment (TME) may play essential roles in insensitive therapeutic reaction. Previous studies have found that epigenetic mechanisms play an important role in the regulation of ISCs. Regulating ISCs may be a new approach to transforming “cold” tumors into “hot” tumors. Here, we focused on the function of ISCs in the TME and discussed how epigenetics is involved in regulating ISCs. In addition, we summarized the mechanisms by which the epigenetic drugs convert immunotherapy-insensitive tumors into immunotherapy-sensitive tumors which would be an innovative tendency for future immunotherapy in “cold” tumor.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11194457/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140875955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Cover Image, Volume 44, Issue 4 封面图片,第 44 卷第 4 期
IF 16.2 1区 医学 Q1 Medicine Pub Date : 2024-04-18 DOI: 10.1002/cac2.12544
Xuechun Wang, Anna Juncker-Jensen, Gang Huang, Mate Levente Nagy, Xuemin Lu, Liang Cheng, Xin Lu

The cover image is based on the Correspondence Spatial relationship of tertiary lymphoid structures and tumor-associated neutrophils in bladder cancer and prognostic potential for anti-PD-L1 immunotherapy by Xuechun Wang et al., https://doi.org/10.1002/cac2.12491.

封面图像基于 Xuechun Wang 等人的 Correspondence Spatial relationship of tertiary lymphoid structures and tumor-associated neutrophils in bladder cancer and prognostic potential for anti-PD-L1 immunotherapy,https://doi.org/10.1002/cac2.12491。
{"title":"Cover Image, Volume 44, Issue 4","authors":"Xuechun Wang,&nbsp;Anna Juncker-Jensen,&nbsp;Gang Huang,&nbsp;Mate Levente Nagy,&nbsp;Xuemin Lu,&nbsp;Liang Cheng,&nbsp;Xin Lu","doi":"10.1002/cac2.12544","DOIUrl":"https://doi.org/10.1002/cac2.12544","url":null,"abstract":"<p>The cover image is based on the Correspondence <i>Spatial relationship of tertiary lymphoid structures and tumor-associated neutrophils in bladder cancer and prognostic potential for anti-PD-L1 immunotherapy</i> by Xuechun Wang et al., https://doi.org/10.1002/cac2.12491.\u0000\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":16.2,"publicationDate":"2024-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12544","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140606406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Dietary long-chain fatty acid metabolism boosts antitumor immune response 膳食长链脂肪酸代谢可增强抗肿瘤免疫反应。
IF 16.2 1区 医学 Q1 Medicine Pub Date : 2024-04-12 DOI: 10.1002/cac2.12543
Jiaming Wang, Xuetao Cao

Overcoming resistance to immune checkpoint blockade (ICB) therapy will pave the way for effective ICB cancer immunotherapy since a large proportion of cancer patients are not responsive to ICB immunotherapy [1]. The molecular mechanisms of ICB resistance are diverse, including tumor-intrinsic resistant factors (such as genetic and epigenetic disorders), immunosuppressive/disabled factors (such as T cell exhaustion), and environmental restrictive factors (such as neuroendocrine stress and metabolic reprogramming). Deciphering the mechanisms for ICB resistance will provide immense potential for designing new immunotherapeutic strategies in refractory cancers. The tumor microenvironment (TME) includes diverse types of cells, such as immune cells, cancer-associated fibroblasts, and endothelial cells, as well as intercellular cytokines/chemokines, growth factors, and metabolites, which have been recognized as crucial determinants in ICB responsiveness. The recent advent of high-throughput metabolomics and lipidomics analysis reveals that metabolic reprogramming in TME is closely associated with cancer cell invasion, regulated cell death, immune escape, and chemoresistance [2]. On the other hand, immunometabolism also modulates tumor-associated immune cell function and immunotherapy efficacy. For instance, higher expression of major facilitator superfamily domain containing 2A (MFSD2A) in gastric cancer cells inhibits transforming growth factor beta 1 (TGF-β1) production by suppressing cyclooxygenase 2 (COX2)-prostaglandin synthesis, thus promoting antitumor immunity via reprogramming TME [3].

As a major component of lipids in TME, long-chain fatty acids (LCFAs) are important energy supply and cellular membrane components for cancer cells. Indeed, various types of LCFAs have been found in TME, showing different and sometimes opposite influences on tumor progression and antitumor immunity [4]. For example, palmitic acid promotes metastasis in oral carcinomas and melanoma mouse models through stimulating intratumoral Schwann cells and innervation [5], whereas linoleic acid potentiates CD8+ T cell antitumor functions via enhancing endoplasmic reticulum-mitochondria contact formation and energetics fitness [6]. Therefore, better understanding of the molecular mechanism for each individual LCFA in tumor immunity is meaningful since it may improve cancer immunotherapy through targeting metabolic reprogramming of TME. In a recent study published in Cell Metabolism, Lai et al. [7] found dietary elaidic acid (EA) supplementation elevates tumoral major histocompatibility complex-1 (MHC-I) expression via acyl-coenzyme A synthetase long chain family member 5 (ACSL5), thus suppressing tumor growth and enhancing anti-programmed cell death protein 1 (anti-PD-1) efficacy (Figure 1).

Acyl-coenzyme A synthetase long-chain family members (ACS

虽然表达ACSL5的LLC肿瘤中CD8+ T细胞数量增加,但CD4+ T细胞数量和T细胞产生的细胞因子不受影响。因此,作者推测 ACSL5 可能会调节肿瘤对 T 细胞介导的杀伤的敏感性。事实上,他们发现,在体外共培养系统中,ACSL5 的缺乏会使肿瘤细胞对 CD8+ T 细胞介导的细胞毒性更具抵抗力。肿瘤免疫原性不足是导致 ICB 抗性的关键原因,因为 CTL 通过 T 细胞受体(TCR)识别 MHC-I 分子呈递的肿瘤抗原来介导肿瘤细胞杀伤 [9]。因此,作者发现 ACSL5 缺失的肿瘤细胞中 MHC-I 表达下调。通过对野生型和 ACSL5 基因缺陷型细胞进行转录组分析,作者注意到,ACSL5 基因敲除后,MHC-I 的关键转录激活因子之一 NLR 家族含 CARD 结构域 5(NLRC5)的表达减少。沉默 NLRC5 可完全消除 ACSL5 异位表达对 MHC-I 的上调,这表明 ACSL5-NLRC5-MHC-I 轴决定了肿瘤细胞的免疫原性,从而诱导抗肿瘤免疫。根据各种 LCFAs 的筛选,作者发现油酸反式异构体 EA(动物脂肪和植物油的常见成分)能有效增强基础和 IFNγ 刺激的 MHC-I 表达。ACSL5或NLRC5缺乏会影响EA对MHC-I的诱导,这进一步验证了EA-ACSL5-NLRC5-MHC-I轴在抗肿瘤免疫代谢调节中的作用。此外,腹腔注射和胃内注射EA都能以免疫依赖的方式抑制体内B16-F10肿瘤的生长。同样,EA与抗PD-1疗法联合使用可显著减少LLC肿瘤负荷,而LLC原本对ICB具有抗药性,这突显了膳食补充EA在促进对ICB的反应性方面具有良好的治疗潜力。最后,作者发现血浆EA水平越高,患者对ICB的反应越好,无进展生存期也越长,这意味着血浆EA可能是ICB疗效的理想预测指标。尽管大量研究表明反式脂肪酸(TFAs)有害,尤其是对心血管疾病有害,但这项研究揭示了反式脂肪酸在增强肿瘤免疫原性和提高抗肿瘤免疫反应方面的意想不到的作用。然而,仍有许多问题需要解决。代谢酶ACSL5如何调控核转录因子NLRC5的表达,以及NLRC5如何作为脂肪酸代谢改变的传感器,这些问题都不清楚。与这一发现相反,有报道称 EA 可通过激活 Wnt 和细胞外信号调节激酶(ERK)信号来促进小鼠结直肠癌细胞的生长和转移[10]。EA在不同肿瘤模型中的相反功能是否是由于TME的异质性造成的,还需要进一步研究。尽管作者关注的是 EA 对肿瘤细胞的抗肿瘤作用,但 EA 是否也直接影响 CD8+ T 细胞的功能尚不清楚。研究摄入 EA 期间免疫细胞的脂质代谢重构、脂肪酸介导的信号转导和抗肿瘤功能调控也很有意义。作者利用同型标记技术证明了 ACSL5 能将 EA 转化为 EA-CoA,然而,EA-CoA 是否直接通过调控靶蛋白酰化(如棕榈酰化)来刺激 MHC-I 的表达仍是未知数。转化为 EA-CoA 后,活化的脂肪酸是下游脂质代谢途径(如脂肪酸 β-氧化(FAO)、脂质生物合成和蛋白质酰化)所必需的(图 1)。我们未发表的数据表明,ACSL5 可调控磷脂合成和细胞膜特性,这也可能影响肿瘤细胞中 MHC-1 和免疫细胞中其他受体的膜转位。虽然作者在本文[7]中排除了FAO对ACSL5介导的MHC-I表达的影响,但ACSL5的代谢扰动仍然复杂,值得进一步探讨EA-ACSL5轴在肿瘤免疫学中的深层分子机制。曹雪涛指导并修改了手稿。作者声明他们没有利益冲突。
{"title":"Dietary long-chain fatty acid metabolism boosts antitumor immune response","authors":"Jiaming Wang,&nbsp;Xuetao Cao","doi":"10.1002/cac2.12543","DOIUrl":"10.1002/cac2.12543","url":null,"abstract":"<p>Overcoming resistance to immune checkpoint blockade (ICB) therapy will pave the way for effective ICB cancer immunotherapy since a large proportion of cancer patients are not responsive to ICB immunotherapy [<span>1</span>]. The molecular mechanisms of ICB resistance are diverse, including tumor-intrinsic resistant factors (such as genetic and epigenetic disorders), immunosuppressive/disabled factors (such as T cell exhaustion), and environmental restrictive factors (such as neuroendocrine stress and metabolic reprogramming). Deciphering the mechanisms for ICB resistance will provide immense potential for designing new immunotherapeutic strategies in refractory cancers. The tumor microenvironment (TME) includes diverse types of cells, such as immune cells, cancer-associated fibroblasts, and endothelial cells, as well as intercellular cytokines/chemokines, growth factors, and metabolites, which have been recognized as crucial determinants in ICB responsiveness. The recent advent of high-throughput metabolomics and lipidomics analysis reveals that metabolic reprogramming in TME is closely associated with cancer cell invasion, regulated cell death, immune escape, and chemoresistance [<span>2</span>]. On the other hand, immunometabolism also modulates tumor-associated immune cell function and immunotherapy efficacy. For instance, higher expression of major facilitator superfamily domain containing 2A (MFSD2A) in gastric cancer cells inhibits transforming growth factor beta 1 (TGF-β1) production by suppressing cyclooxygenase 2 (COX2)-prostaglandin synthesis, thus promoting antitumor immunity via reprogramming TME [<span>3</span>].</p><p>As a major component of lipids in TME, long-chain fatty acids (LCFAs) are important energy supply and cellular membrane components for cancer cells. Indeed, various types of LCFAs have been found in TME, showing different and sometimes opposite influences on tumor progression and antitumor immunity [<span>4</span>]. For example, palmitic acid promotes metastasis in oral carcinomas and melanoma mouse models through stimulating intratumoral Schwann cells and innervation [<span>5</span>], whereas linoleic acid potentiates CD8<sup>+</sup> T cell antitumor functions via enhancing endoplasmic reticulum-mitochondria contact formation and energetics fitness [<span>6</span>]. Therefore, better understanding of the molecular mechanism for each individual LCFA in tumor immunity is meaningful since it may improve cancer immunotherapy through targeting metabolic reprogramming of TME. In a recent study published in <i>Cell Metabolism</i>, Lai <i>et al.</i> [<span>7</span>] found dietary elaidic acid (EA) supplementation elevates tumoral major histocompatibility complex-1 (MHC-I) expression via acyl-coenzyme A synthetase long chain family member 5 (ACSL5), thus suppressing tumor growth and enhancing anti-programmed cell death protein 1 (anti-PD-1) efficacy (Figure 1).</p><p>Acyl-coenzyme A synthetase long-chain family members (ACS","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":16.2,"publicationDate":"2024-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12543","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140710827","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Proteomics of cell-free breast cancer scaffolds identify clinically relevant imprinted proteins and cancer-progressing properties 无细胞乳腺癌支架的蛋白质组学鉴定临床相关的印迹蛋白和癌症进展特性
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-04-04 DOI: 10.1002/cac2.12542
Anna Gustafsson, Emma Jonasson, Anders Ståhlberg, Göran Landberg

The composition of the extracellular tumor microenvironment (TME) has not been fully delineated, limiting the understanding of general cancer-progressing properties within the cancer niche. The interplay and dynamics between cancer cells and the surrounding structures and cells clearly differ between various subtypes of cancer, adding to the complexity of precision medicine [1].

To better understand the composition and to define the imprinted proteins of the TME in breast cancer and its potential associations with clinical properties of the disease, we performed global proteomic analysis on a cohort of 63 decellularized patient-derived scaffolds (PDSs). PDSs represents the cell-free TME and were prepared using primary lesions from breast cancer patients with available clinicopathological data (Figure 1A, Supplementary file of methods). The PDS method has earlier been shown to maintain tumor tissue heterogeniety in vitro, producing quantitative assessments of the activity of the TME when studying cancer cell lines adapted to various PDS-based cell cultures [2]. Specific gene changes in the cancer population induced by the heterogenous PDS culture conditions have also been linked to clinical observations, validating the selected strategy for this study [2-5].

The analysis of the cell-free PDSs identified 1,844 unique proteins (Supplementary Table S1), showing enrichment for proteins related to metabolism, translation, transport, immunity, and extracellular matrix (ECM). Surprisingly, most proteins were annotated as cytoplasmic, suggesting that intracellular proteins were also associated with the PDSs, as further deliberated below (Supplementary Figure S1). When comparing the protein contents from primary cancer samples with adjacent normal breast tissues, 1,280 of the 1,844 detected proteins from the cell-free compartments were differentially expressed. Normal breast tissues were, in comparison to the PDSs, enriched for proteins involved in oxidation-reducing processes, secretion, regulation of exocytosis, and ECM organization (Supplementary Figure S2).

Next, we used k-means clustering to identify potential subgroups of patients based on the protein composition of the cell-free PDSs. PDS Clusters 1-3 were recognized, distinguished by the expression of proteins in Clusters A-C. Interestingly, the PDS clusters were significantly associated with cancer subtypes (Figure 1B and Supplementary Table S2). Cluster 1 was mainly derived from lobular cancers (63%) and low-grade cancers, showing high levels of 458 proteins involved in ECM organization, biological adhesion, and leukocyte-mediated immunity forming protein Cluster A. Cluster 2 was primarily derived from ductal cancers (77%) and showed pronounced expression of protein Cluster B, comprising of 396 proteins related to exocytosis, secretion and neutrophil degranulation. Cluster 3 included a mixture of ductal and lobular cancers with the highe

接下来,我们根据无细胞 PDS 的蛋白质组成,使用 k-means 聚类方法来识别潜在的患者亚群。根据簇 A-C 中蛋白质的表达情况,我们识别出了 PDS 簇 1-3。有趣的是,PDS 群与癌症亚型显著相关(图 1B 和补充表 S2)。簇 1 主要来自小叶癌(63%)和低级别癌症,显示出高水平的 458 种蛋白质,这些蛋白质涉及 ECM 组织、生物粘附和白细胞介导的免疫,形成了蛋白质簇 A。第 3 组包括导管癌和小叶癌的混合体,其中 C 组蛋白质含量最高,含有 990 个主要参与代谢过程和建立细胞定位的蛋白质。在独立于已定义的簇鉴定两种癌症类型的蛋白质时(图 1C 和补充表 S3),导管癌富含被归类为修饰酶、代谢物相互转换酶、RNA 代谢蛋白和翻译蛋白的蛋白质。与此相反,小叶癌的 ECM 蛋白和防御/免疫蛋白含量较高(图 1D),这支持了早先的研究结果,即小叶癌亚群可能对免疫靶向癌症疗法更敏感[1]。与进展性疾病和复发相关的 52 个蛋白质主要分为转运体和代谢物相互转换酶,而与非复发性癌症相关的 31 个蛋白质主要是细胞骨架和支架/适配蛋白。有趣的是,在与侵袭性疾病相关的蛋白质中,角蛋白和溶质载体(SLC)家族的成员非常突出,在14种已鉴定的角蛋白中,有8种在后来疾病复发患者的PDS中含量更高(图1F)。在单变量分析(图 1G)和多变量分析(危险比 = 14.86,P = 0.01)中,PDSs 中较高水平的 KRT78 与较差的无病生存率显著相关。为了进一步确定乳腺癌中的支架内容,我们根据传统上具有 TME 代表性的 126 个 ECM 相关蛋白子集对 PDSs 进行了聚类(图 1H 和补充表 S4)。观察到三个新的基于 ECM 的 PDS 聚类,它们与之前报告的聚类只有部分重叠(补充图 S3),并与癌症分级和患者年龄有显著关联(补充表 S5-S6)。ECM-1群组显示低级别癌症比例高,患者年龄参差不齐,其特征是胶原、微纤相关蛋白(EMILIN1、MFAP2)、纤连蛋白(FBN1、FBN2)、潜伏转化生长因子β结合蛋白LTBP1和过氧化物酶PRDX4含量高,以及整合素亚基ITGB2含量低。群组 ECM-2 与高级别癌症、患者年龄较大有关,并与较低水平的 ECM 蛋白(尤其是胶原和层粘连蛋白亚基)以及结构蛋白 tenascin XB (TNXB) 紧密相关。相比之下,ECM-3 组主要包括各年龄段的低级别癌症,ECM 蛋白含量较高。当详细研究 PDS 中胶原蛋白的丰度与患者年龄的关系时,在 21 种检测到的胶原蛋白中,有 10 种随着患者年龄的增加而明显逐渐减少(图 1I)。重要的是,所提供的数据表明,在恶性程度较低的低级别 TME 中,与 ECM 相关的蛋白质通常更为丰富。这表明,模仿侵袭性乳腺癌的三维生长模型不能仅使用 ECM 蛋白进行再现,还应该辅以癌症龛位中存在的其他蛋白。癌症患者的年龄也与 PDS 的 ECM 组成有关,年轻患者表现出更高水平的 ECM 蛋白,特别是胶原蛋白。胶原蛋白的更替是组织稳态的正常组成部分,而在包括皮肤在内的老年组织中,胶原蛋白通常会变得更加疏松、杂乱和破碎[7]。老年患者的药代动力学特征也与年轻患者不同。
{"title":"Proteomics of cell-free breast cancer scaffolds identify clinically relevant imprinted proteins and cancer-progressing properties","authors":"Anna Gustafsson,&nbsp;Emma Jonasson,&nbsp;Anders Ståhlberg,&nbsp;Göran Landberg","doi":"10.1002/cac2.12542","DOIUrl":"10.1002/cac2.12542","url":null,"abstract":"<p>The composition of the extracellular tumor microenvironment (TME) has not been fully delineated, limiting the understanding of general cancer-progressing properties within the cancer niche. The interplay and dynamics between cancer cells and the surrounding structures and cells clearly differ between various subtypes of cancer, adding to the complexity of precision medicine [<span>1</span>].</p><p>To better understand the composition and to define the imprinted proteins of the TME in breast cancer and its potential associations with clinical properties of the disease, we performed global proteomic analysis on a cohort of 63 decellularized patient-derived scaffolds (PDSs). PDSs represents the cell-free TME and were prepared using primary lesions from breast cancer patients with available clinicopathological data (Figure 1A, Supplementary file of methods). The PDS method has earlier been shown to maintain tumor tissue heterogeniety in vitro, producing quantitative assessments of the activity of the TME when studying cancer cell lines adapted to various PDS-based cell cultures [<span>2</span>]. Specific gene changes in the cancer population induced by the heterogenous PDS culture conditions have also been linked to clinical observations, validating the selected strategy for this study [<span>2-5</span>].</p><p>The analysis of the cell-free PDSs identified 1,844 unique proteins (Supplementary Table S1), showing enrichment for proteins related to metabolism, translation, transport, immunity, and extracellular matrix (ECM). Surprisingly, most proteins were annotated as cytoplasmic, suggesting that intracellular proteins were also associated with the PDSs, as further deliberated below (Supplementary Figure S1). When comparing the protein contents from primary cancer samples with adjacent normal breast tissues, 1,280 of the 1,844 detected proteins from the cell-free compartments were differentially expressed. Normal breast tissues were, in comparison to the PDSs, enriched for proteins involved in oxidation-reducing processes, secretion, regulation of exocytosis, and ECM organization (Supplementary Figure S2).</p><p>Next, we used k-means clustering to identify potential subgroups of patients based on the protein composition of the cell-free PDSs. PDS Clusters 1-3 were recognized, distinguished by the expression of proteins in Clusters A-C. Interestingly, the PDS clusters were significantly associated with cancer subtypes (Figure 1B and Supplementary Table S2). Cluster 1 was mainly derived from lobular cancers (63%) and low-grade cancers, showing high levels of 458 proteins involved in ECM organization, biological adhesion, and leukocyte-mediated immunity forming protein Cluster A. Cluster 2 was primarily derived from ductal cancers (77%) and showed pronounced expression of protein Cluster B, comprising of 396 proteins related to exocytosis, secretion and neutrophil degranulation. Cluster 3 included a mixture of ductal and lobular cancers with the highe","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12542","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140598842","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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 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
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