首页 > 最新文献

Cancer Science最新文献

英文 中文
Pembrolizumab for Early-Stage Triple-Negative Breast Cancer: KEYNOTE-522 Japan Subgroup Analysis. 派姆单抗治疗早期三阴性乳腺癌:KEYNOTE-522日本亚组分析
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-14 DOI: 10.1111/cas.70307
Masato Takahashi, Hirofumi Mukai, Toshimi Takano, Koichiro Tsugawa, Kenichi Inoue, Mitsuya Itoh, Junichiro Watanabe, Yuko Tanabe, Naohito Yamamoto, Yasuo Miyoshi, Kenichi Watanabe, Toru Mukohara, Yibin Kong, Masashi Shimura, Francisco Beca, Peter Schmid, Hiroji Iwata

The phase 3 KEYNOTE-522 study in high-risk early-stage triple-negative breast cancer (TNBC) showed significantly improved efficacy outcomes with neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab versus neoadjuvant chemotherapy alone. We present findings from the KEYNOTE-522 Japan subgroup. Eligible participants (aged ≥ 18 years) with untreated locally advanced TNBC (stage T1c N1-2 or T2-4 N0-2) were randomized 2:1 to neoadjuvant pembrolizumab 200 mg or placebo plus chemotherapy every 3 weeks for 8 cycles followed by surgery and adjuvant pembrolizumab or placebo for ≤ 9 cycles. Primary endpoints were pathologic complete response (pCR; ypT0/Tis ypN0) at the time of surgery and event-free survival (EFS). Of 76 participants enrolled in Japan, 45 were randomized to the pembrolizumab arm and 31 to the placebo arm. Median time from randomization to data cutoff (March 22, 2024) was 76.3 months. Twenty-four participants (53%) in the pembrolizumab arm and 15 (48%) in the placebo arm achieved pCR (between-treatment arm difference, 4.9%; 95% CI, -17.6% to 27.1%); findings were similar regardless of PD-L1 expression. Rates of EFS at 60 months were 84% and 73%, respectively (HR, 0.54; 95% CI, 0.20-1.50). Grade 3 or 4 treatment-related AEs occurred in 37 of 45 participants (82%) treated with pembrolizumab and 23 of 30 participants (77%) treated with placebo; there were no grade 5 AEs. In conclusion, neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab showed improved efficacy outcomes and manageable safety versus neoadjuvant chemotherapy alone in Japanese participants, supporting the use of this regimen in Japanese patients with high-risk early-stage TNBC. Trial Registration: The study (ClinicalTrials.gov, NCT03036488) was conducted in compliance with local and/or national regulations and International Council for Harmonization Good Clinical Practice guidelines and in accordance with the ethical principles originating from the Declaration of Helsinki.

高风险早期三阴性乳腺癌(TNBC)的3期KEYNOTE-522研究显示,新辅助派姆单抗联合化疗后,辅助派姆单抗与单独新辅助化疗相比,疗效显著改善。我们介绍KEYNOTE-522日本亚组的研究结果。符合条件的参与者(年龄≥18岁)未经治疗的局部晚期TNBC (T1c N1-2或T2-4 N0-2)随机分为2:1,每3周接受新辅助派姆单抗200 mg或安慰剂加化疗,共8个周期,随后进行手术和辅助派姆单抗或安慰剂治疗≤9个周期。主要终点是手术时的病理完全缓解(pCR; ypT0/Tis ypN0)和无事件生存期(EFS)。在日本招募的76名参与者中,45名随机分配到派姆单抗组,31名随机分配到安慰剂组。从随机分配到数据截止(2024年3月22日)的中位时间为76.3个月。派姆单抗组的24名参与者(53%)和安慰剂组的15名参与者(48%)实现了pCR(治疗组间差异为4.9%;95% CI, -17.6%至27.1%);无论PD-L1表达如何,结果相似。60个月时EFS发生率分别为84%和73% (HR, 0.54; 95% CI, 0.20-1.50)。接受派姆单抗治疗的45名参与者中有37名(82%)发生3级或4级治疗相关不良事件,30名接受安慰剂治疗的参与者中有23名(77%)发生3级或4级不良事件;无5级ae。总之,在日本参与者中,新辅助派姆单抗加化疗后再辅助派姆单抗与单独新辅助化疗相比,显示出更好的疗效结果和可管理的安全性,支持在日本高风险早期TNBC患者中使用该方案。试验注册:该研究(ClinicalTrials.gov, NCT03036488)符合地方和/或国家法规和国际协调良好临床实践委员会指南,并符合源自赫尔辛基宣言的伦理原则。
{"title":"Pembrolizumab for Early-Stage Triple-Negative Breast Cancer: KEYNOTE-522 Japan Subgroup Analysis.","authors":"Masato Takahashi, Hirofumi Mukai, Toshimi Takano, Koichiro Tsugawa, Kenichi Inoue, Mitsuya Itoh, Junichiro Watanabe, Yuko Tanabe, Naohito Yamamoto, Yasuo Miyoshi, Kenichi Watanabe, Toru Mukohara, Yibin Kong, Masashi Shimura, Francisco Beca, Peter Schmid, Hiroji Iwata","doi":"10.1111/cas.70307","DOIUrl":"https://doi.org/10.1111/cas.70307","url":null,"abstract":"<p><p>The phase 3 KEYNOTE-522 study in high-risk early-stage triple-negative breast cancer (TNBC) showed significantly improved efficacy outcomes with neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab versus neoadjuvant chemotherapy alone. We present findings from the KEYNOTE-522 Japan subgroup. Eligible participants (aged ≥ 18 years) with untreated locally advanced TNBC (stage T1c N1-2 or T2-4 N0-2) were randomized 2:1 to neoadjuvant pembrolizumab 200 mg or placebo plus chemotherapy every 3 weeks for 8 cycles followed by surgery and adjuvant pembrolizumab or placebo for ≤ 9 cycles. Primary endpoints were pathologic complete response (pCR; ypT0/Tis ypN0) at the time of surgery and event-free survival (EFS). Of 76 participants enrolled in Japan, 45 were randomized to the pembrolizumab arm and 31 to the placebo arm. Median time from randomization to data cutoff (March 22, 2024) was 76.3 months. Twenty-four participants (53%) in the pembrolizumab arm and 15 (48%) in the placebo arm achieved pCR (between-treatment arm difference, 4.9%; 95% CI, -17.6% to 27.1%); findings were similar regardless of PD-L1 expression. Rates of EFS at 60 months were 84% and 73%, respectively (HR, 0.54; 95% CI, 0.20-1.50). Grade 3 or 4 treatment-related AEs occurred in 37 of 45 participants (82%) treated with pembrolizumab and 23 of 30 participants (77%) treated with placebo; there were no grade 5 AEs. In conclusion, neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab showed improved efficacy outcomes and manageable safety versus neoadjuvant chemotherapy alone in Japanese participants, supporting the use of this regimen in Japanese patients with high-risk early-stage TNBC. Trial Registration: The study (ClinicalTrials.gov, NCT03036488) was conducted in compliance with local and/or national regulations and International Council for Harmonization Good Clinical Practice guidelines and in accordance with the ethical principles originating from the Declaration of Helsinki.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971470","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In-Tumor CRISPR-Cas9 Knockout Screening and Novel Therapy Development for Malignant Transformation of Ovarian Teratoma. 卵巢畸胎瘤恶性转化的肿瘤内CRISPR-Cas9基因敲除筛选及新疗法开发
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-13 DOI: 10.1111/cas.70315
Satoshi Tamauchi, Kosuke Yoshida, Wang Xinyuan, Atsushi Nakagawa, Akira Yokoi, Nobuhisa Yoshikawa, Kaoru Niimi, Yusuke Yamamoto, Hiroaki Kajiyama

Malignant transformation of mature cystic teratoma (MTMCT) of the ovary is a rare but aggressive malignancy for which no standardized chemotherapy or effective targeted therapies currently exist. To identify therapeutic vulnerabilities in MTMCT, we performed a genome-wide CRISPR-Cas9 knockout screen using the MTMCT-derived NOSCC1 cell line. Two parallel selective pressures were applied: in vivo tumorigenicity in immunodeficient mice and cisplatin exposure in vitro. From this screen, 67 negatively selected genes were identified, among which SOD1 and NDUFB4 emerged as top candidates based on high basal expression levels and clinical relevance. Integration with spatial transcriptomic data from three independent MTMCT patient tumors further supported the prioritization of these targets. SOD1 was selected for further investigation due to the availability of known pharmacological inhibitors. Both siRNA-mediated knockdown and small-molecule inhibition of SOD1 using LCS-1 significantly suppressed MTMCT cell proliferation in vitro by inducing oxidative stress and impairing cell cycle progression. This antiproliferative effect was reversed by co-treatment with N-acetylcysteine, a reactive oxygen species scavenger. In vivo validation using patient-derived xenograft models demonstrated that oral administration of LCS-1 led to significant tumor growth suppression and increased expression of apoptotic and DNA damage markers, including cleaved caspase-3 and γH2AX. These findings establish SOD1 as a critical vulnerability in MTMCT and provide preclinical evidence supporting redox modulation as a therapeutic strategy for this highly chemoresistant and understudied ovarian cancer subtype. Our integrative approach combining functional genomics, spatial transcriptomics, and pharmacologic validation offers a framework for the discovery of novel targets in rare gynecologic malignancies.

卵巢成熟囊性畸胎瘤(MTMCT)是一种罕见但侵袭性的恶性肿瘤,目前尚无标准化的化疗或有效的靶向治疗方法。为了确定MTMCT的治疗脆弱性,我们使用MTMCT衍生的NOSCC1细胞系进行了全基因组CRISPR-Cas9敲除筛选。采用两种平行的选择压力:免疫缺陷小鼠的体内致瘤性和体外顺铂暴露。从这个筛选中,鉴定出67个负选择基因,其中SOD1和NDUFB4是基于高基础表达水平和临床相关性的最佳候选基因。与来自三个独立MTMCT患者肿瘤的空间转录组数据的整合进一步支持了这些靶点的优先级。我们选择SOD1作为进一步研究的对象,是因为有已知的药理抑制剂。sirna介导的SOD1敲除和LCS-1小分子抑制均通过诱导氧化应激和损害细胞周期进程显著抑制MTMCT细胞体外增殖。通过与n -乙酰半胱氨酸(一种活性氧清除剂)共同处理,这种抗增殖作用被逆转。使用患者来源的异种移植模型进行体内验证表明,口服LCS-1可显著抑制肿瘤生长,并增加凋亡和DNA损伤标志物的表达,包括cleaved caspase-3和γH2AX。这些发现证实SOD1是MTMCT的关键易感性,并提供临床前证据支持氧化还原调节作为这种高度耐药且研究不足的卵巢癌亚型的治疗策略。我们的综合方法结合了功能基因组学、空间转录组学和药理学验证,为发现罕见妇科恶性肿瘤的新靶点提供了一个框架。
{"title":"In-Tumor CRISPR-Cas9 Knockout Screening and Novel Therapy Development for Malignant Transformation of Ovarian Teratoma.","authors":"Satoshi Tamauchi, Kosuke Yoshida, Wang Xinyuan, Atsushi Nakagawa, Akira Yokoi, Nobuhisa Yoshikawa, Kaoru Niimi, Yusuke Yamamoto, Hiroaki Kajiyama","doi":"10.1111/cas.70315","DOIUrl":"https://doi.org/10.1111/cas.70315","url":null,"abstract":"<p><p>Malignant transformation of mature cystic teratoma (MTMCT) of the ovary is a rare but aggressive malignancy for which no standardized chemotherapy or effective targeted therapies currently exist. To identify therapeutic vulnerabilities in MTMCT, we performed a genome-wide CRISPR-Cas9 knockout screen using the MTMCT-derived NOSCC1 cell line. Two parallel selective pressures were applied: in vivo tumorigenicity in immunodeficient mice and cisplatin exposure in vitro. From this screen, 67 negatively selected genes were identified, among which SOD1 and NDUFB4 emerged as top candidates based on high basal expression levels and clinical relevance. Integration with spatial transcriptomic data from three independent MTMCT patient tumors further supported the prioritization of these targets. SOD1 was selected for further investigation due to the availability of known pharmacological inhibitors. Both siRNA-mediated knockdown and small-molecule inhibition of SOD1 using LCS-1 significantly suppressed MTMCT cell proliferation in vitro by inducing oxidative stress and impairing cell cycle progression. This antiproliferative effect was reversed by co-treatment with N-acetylcysteine, a reactive oxygen species scavenger. In vivo validation using patient-derived xenograft models demonstrated that oral administration of LCS-1 led to significant tumor growth suppression and increased expression of apoptotic and DNA damage markers, including cleaved caspase-3 and γH2AX. These findings establish SOD1 as a critical vulnerability in MTMCT and provide preclinical evidence supporting redox modulation as a therapeutic strategy for this highly chemoresistant and understudied ovarian cancer subtype. Our integrative approach combining functional genomics, spatial transcriptomics, and pharmacologic validation offers a framework for the discovery of novel targets in rare gynecologic malignancies.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145967763","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neoadjuvant Therapy-Induced Remodeling in Pancreatic Ductal Adenocarcinoma: Multimodal Spatial Analysis and Prognosis. 新辅助治疗诱导的胰腺导管腺癌重构:多模式空间分析和预后。
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-13 DOI: 10.1111/cas.70320
Xiaofei Zhang, Ruoxin Lan, Danting Li, Yongjun Liu, Sonu Kalyan, Momin Iqbal, Nancy Liu, Jerry Zhang, Iman Hanna, Mala Gupta, Chaohui L Zhao, Weiguo Liu, Jonathan Melamed, Michael Shusterman, Jessica Widmer, John Allendorf, Yao-Zhong Liu

Neoadjuvant therapy (NAT) is increasingly used for pancreatic ductal adenocarcinoma (PDAC); yet most patients only achieve partial response. Pathological treatment response grading focuses on assessing residual tumor burden, often overlooking changes in tumor microenvironment (TME). To address this gap, we compared tumor cells and TME of 13 NAT-naïve and 23 post-NAT PDACs using integrated spatial pathomics and transcriptomics, with validation in an independent single-cell spatial dataset. NAT significantly reduced tumor burden (14.7%-6.2%, p = 0.004), but systemic comparison of 13 cytomorphometric features of tumor cells alone did not reliably distinguish between naïve and NAT cases. In contrast, NAT profoundly remodeled TME by increasing cancer-associated fibroblast (CAF) and CD8+ T cell densities, promoting CD8+ T cell-tumor cell proximity and fibrosis, reducing tumor-associated neutrophils, and redistributing tertiary lymphoid structures (TLSs). Spatial transcriptomics shows NAT induced apoptosis, DNA-damage response, and AGC-kinase (S_TK_X) signaling in tumor cells, and upregulated complement pathway, p53 signaling, and cellular senescence program in TME. Cross-platform single-cell spatial analysis revealed decreased regulatory T cells (Treg) and a shift from myofibroblastic (mCAF) to inflammatory CAF (iCAF). Importantly, post-NAT patients with more fibrosis had longer overall survival (p = 0.02), and higher B-cell density showed a favorable trend (p = 0.06). Together, these results suggest that beyond tumor debulking, NAT induces a coordinated TME remodeling characterized by fibroblast reprogramming, matrix fibrosis, and immune spatial reorganization. Incorporating assessment of NAT-induced stromal and immune changes into TRG may improve prognostication and guide more precise therapy in post-NAT PDAC.

新辅助治疗(NAT)越来越多地用于胰腺导管腺癌(PDAC);然而,大多数患者只能达到部分反应。病理治疗反应分级侧重于评估残余肿瘤负荷,往往忽略肿瘤微环境(TME)的变化。为了解决这一差距,我们使用整合的空间病理学和转录组学方法比较了13个NAT-naïve和23个nat后pdac的肿瘤细胞和TME,并在独立的单细胞空间数据集中进行了验证。NAT显著降低了肿瘤负荷(14.7%-6.2%,p = 0.004),但仅系统比较肿瘤细胞的13个细胞形态特征并不能可靠地区分naïve和NAT病例。相反,NAT通过增加癌症相关成纤维细胞(CAF)和CD8+ T细胞密度,促进CD8+ T细胞-肿瘤细胞接近和纤维化,减少肿瘤相关中性粒细胞和重新分配三级淋巴样结构(TLSs),深刻地重塑了TME。空间转录组学显示,NAT诱导肿瘤细胞凋亡、dna损伤反应和agc -激酶(S_TK_X)信号,以及TME中补体通路、p53信号和细胞衰老程序的上调。跨平台单细胞空间分析显示调节性T细胞(Treg)减少,从肌成纤维细胞(mCAF)向炎性细胞(iCAF)转变。重要的是,nat后纤维化程度越高的患者总生存期越长(p = 0.02), b细胞密度越高的患者总体生存期越长(p = 0.06)。综上所述,这些结果表明,除了肿瘤缩小之外,NAT还诱导了以成纤维细胞重编程、基质纤维化和免疫空间重组为特征的协调TME重塑。将nat诱导的基质和免疫变化的评估纳入TRG可能改善预后并指导nat后PDAC的更精确治疗。
{"title":"Neoadjuvant Therapy-Induced Remodeling in Pancreatic Ductal Adenocarcinoma: Multimodal Spatial Analysis and Prognosis.","authors":"Xiaofei Zhang, Ruoxin Lan, Danting Li, Yongjun Liu, Sonu Kalyan, Momin Iqbal, Nancy Liu, Jerry Zhang, Iman Hanna, Mala Gupta, Chaohui L Zhao, Weiguo Liu, Jonathan Melamed, Michael Shusterman, Jessica Widmer, John Allendorf, Yao-Zhong Liu","doi":"10.1111/cas.70320","DOIUrl":"https://doi.org/10.1111/cas.70320","url":null,"abstract":"<p><p>Neoadjuvant therapy (NAT) is increasingly used for pancreatic ductal adenocarcinoma (PDAC); yet most patients only achieve partial response. Pathological treatment response grading focuses on assessing residual tumor burden, often overlooking changes in tumor microenvironment (TME). To address this gap, we compared tumor cells and TME of 13 NAT-naïve and 23 post-NAT PDACs using integrated spatial pathomics and transcriptomics, with validation in an independent single-cell spatial dataset. NAT significantly reduced tumor burden (14.7%-6.2%, p = 0.004), but systemic comparison of 13 cytomorphometric features of tumor cells alone did not reliably distinguish between naïve and NAT cases. In contrast, NAT profoundly remodeled TME by increasing cancer-associated fibroblast (CAF) and CD8<sup>+</sup> T cell densities, promoting CD8<sup>+</sup> T cell-tumor cell proximity and fibrosis, reducing tumor-associated neutrophils, and redistributing tertiary lymphoid structures (TLSs). Spatial transcriptomics shows NAT induced apoptosis, DNA-damage response, and AGC-kinase (S_TK_X) signaling in tumor cells, and upregulated complement pathway, p53 signaling, and cellular senescence program in TME. Cross-platform single-cell spatial analysis revealed decreased regulatory T cells (Treg) and a shift from myofibroblastic (mCAF) to inflammatory CAF (iCAF). Importantly, post-NAT patients with more fibrosis had longer overall survival (p = 0.02), and higher B-cell density showed a favorable trend (p = 0.06). Together, these results suggest that beyond tumor debulking, NAT induces a coordinated TME remodeling characterized by fibroblast reprogramming, matrix fibrosis, and immune spatial reorganization. Incorporating assessment of NAT-induced stromal and immune changes into TRG may improve prognostication and guide more precise therapy in post-NAT PDAC.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145967721","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Resveratrol as a Novel YAP Inhibitor Targeting Glioblastoma Progression and Sensitizing to Chemotherapy. 白藜芦醇作为一种针对胶质母细胞瘤进展和化疗增敏的新型YAP抑制剂。
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-10 DOI: 10.1111/cas.70317
Wannawat Khotchawan, Chanchao Lorthongpanich, Pakpoom Kheolamai, Sith Sathornsumetee, Surapol Issaragrisil

Dysregulation of YAP, the terminal effector of the Hippo pathway, contributes to cancer progression and drug resistance. Its role in glioblastoma (GBM), the most aggressive brain cancer, remains incompletely understood. Single-cell RNA sequencing data from a published GBM dataset were reanalyzed to assess YAP expression across cell populations. YAP was silenced via shRNA in GBM cell lines (U-251 MG, U-87 MG) and patient-derived GBM cells. Resveratrol (RV), a natural blood-brain barrier-permeable compound, was evaluated for growth inhibition and YAP-targeted effects. Functional assays measured proliferation, spheroid formation, migration, invasion, and drug sensitivity. YAP and its cofactor TEAD were highly upregulated in GBM cells compared with normal brain and stromal cells. YAP depletion by shRNA suppressed proliferation, spheroid formation, migration, and invasion. RV treatment similarly inhibited YAP expression, reducing proliferation and viability in monolayer and 3D spheroid cultures, and impairing migration and invasion via epithelial-mesenchymal transition (EMT) inhibition. RV-mediated YAP suppression also enhanced sensitivity to temozolomide (TMZ) and carmustine (BCNU), increasing their cytotoxicity in GBM cells. RV acts as a novel YAP inhibitor in GBM, impairing malignant phenotypes and potentiating the effects of standard chemotherapy. These findings support RV as a potential adjunct in YAP-targeted GBM therapy, warranting further in vivo validation for clinical translation.

Hippo通路的末端效应物YAP的失调有助于癌症的进展和耐药性。它在最具侵袭性的脑癌胶质母细胞瘤(GBM)中的作用仍不完全清楚。重新分析来自已发表的GBM数据集的单细胞RNA测序数据,以评估YAP在细胞群中的表达。YAP通过shRNA在GBM细胞系(U-251 MG、U-87 MG)和患者源性GBM细胞中沉默。白藜芦醇(Resveratrol, RV)是一种天然的血脑屏障渗透化合物,研究了其生长抑制和yap靶向作用。功能试验测量细胞增殖、球体形成、迁移、侵袭和药物敏感性。与正常脑细胞和间质细胞相比,YAP及其辅助因子TEAD在GBM细胞中高度上调。shRNA耗竭YAP抑制增殖、球状体形成、迁移和侵袭。RV处理同样抑制YAP表达,降低单层和3D球形培养物的增殖和活力,并通过上皮-间充质转化(EMT)抑制损害迁移和侵袭。rv介导的YAP抑制也增强了对替莫唑胺(TMZ)和卡莫司汀(BCNU)的敏感性,增加了它们在GBM细胞中的细胞毒性。RV在GBM中作为一种新的YAP抑制剂,损害恶性表型并增强标准化疗的效果。这些发现支持RV作为yap靶向GBM治疗的潜在辅助药物,需要进一步的临床转化体内验证。
{"title":"Resveratrol as a Novel YAP Inhibitor Targeting Glioblastoma Progression and Sensitizing to Chemotherapy.","authors":"Wannawat Khotchawan, Chanchao Lorthongpanich, Pakpoom Kheolamai, Sith Sathornsumetee, Surapol Issaragrisil","doi":"10.1111/cas.70317","DOIUrl":"https://doi.org/10.1111/cas.70317","url":null,"abstract":"<p><p>Dysregulation of YAP, the terminal effector of the Hippo pathway, contributes to cancer progression and drug resistance. Its role in glioblastoma (GBM), the most aggressive brain cancer, remains incompletely understood. Single-cell RNA sequencing data from a published GBM dataset were reanalyzed to assess YAP expression across cell populations. YAP was silenced via shRNA in GBM cell lines (U-251 MG, U-87 MG) and patient-derived GBM cells. Resveratrol (RV), a natural blood-brain barrier-permeable compound, was evaluated for growth inhibition and YAP-targeted effects. Functional assays measured proliferation, spheroid formation, migration, invasion, and drug sensitivity. YAP and its cofactor TEAD were highly upregulated in GBM cells compared with normal brain and stromal cells. YAP depletion by shRNA suppressed proliferation, spheroid formation, migration, and invasion. RV treatment similarly inhibited YAP expression, reducing proliferation and viability in monolayer and 3D spheroid cultures, and impairing migration and invasion via epithelial-mesenchymal transition (EMT) inhibition. RV-mediated YAP suppression also enhanced sensitivity to temozolomide (TMZ) and carmustine (BCNU), increasing their cytotoxicity in GBM cells. RV acts as a novel YAP inhibitor in GBM, impairing malignant phenotypes and potentiating the effects of standard chemotherapy. These findings support RV as a potential adjunct in YAP-targeted GBM therapy, warranting further in vivo validation for clinical translation.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145946669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SUSD2 Promotes Metastasis and Primary Tumor Growth in Pancreatic Cancer Cells via Integrin-FAK Signaling Activation. SUSD2通过整合素- fak信号激活促进胰腺癌细胞转移和原发肿瘤生长。
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-09 DOI: 10.1111/cas.70318
Junjiro Yoshida, Tomokazu Ohishi, Isao Momose, Shun-Ichi Ohba, Kyohei Kurosawa, Akiko Harakawa, Hiroyuki Inoue, Minori Senoo, Daisuke Tatsuda, Hikaru Abe, Atsushi Masamune, Manabu Kawada

Tumor tissues in pancreatic cancer develop with abundant cancer-associated fibroblasts (CAFs), promoting tumor progression. CAF-conditioned medium induces the expression of sushi domain-containing 2 (SUSD2) and enhances the invasive potential of pancreatic cancer cells. We showed that SUSD2 binds to integrin β1 and promotes pancreatic cancer cell motility by inducing phosphorylation of focal adhesion kinase (FAK), facilitating the formation of focal adhesion complexes in cells adhered to collagen 1 or fibronectin. Orthotopic transplantation of SUSD2-overexpressing human pancreatic cancer cell lines into the mouse pancreas enhanced liver metastasis in Panc-1 cells, whereas in KP2 cells, it increased primary tumor growth without promoting metastasis. In spheroid cultures of KP2 cells, forced SUSD2 expression elevated FAK phosphorylation independently of cell adhesion, suggesting that SUSD2 promotes cell proliferation even in non-metastatic cells. High SUSD2 expression in cancer cells contributes to tumor growth and metastasis, identifying SUSD2 as a potential therapeutic target in pancreatic cancer.

胰腺癌肿瘤组织中存在大量的癌相关成纤维细胞,促进肿瘤进展。ca条件培养基诱导含sushi结构域2 (SUSD2)的表达,增强胰腺癌细胞的侵袭潜能。我们发现,SUSD2与整合素β1结合,通过诱导局灶黏附激酶(FAK)的磷酸化促进胰腺癌细胞的运动,促进黏附于胶原1或纤维连接蛋白的细胞形成局灶黏附复合物。将过表达susd2的人胰腺癌细胞系原位移植到小鼠胰腺中,可促进Panc-1细胞的肝转移,而在KP2细胞中,可促进原发肿瘤生长,但不促进转移。在球形培养的KP2细胞中,强制SUSD2表达提高了FAK磷酸化,而不依赖于细胞粘附,这表明即使在非转移细胞中,SUSD2也能促进细胞增殖。SUSD2在癌细胞中的高表达有助于肿瘤的生长和转移,这表明SUSD2是胰腺癌的潜在治疗靶点。
{"title":"SUSD2 Promotes Metastasis and Primary Tumor Growth in Pancreatic Cancer Cells via Integrin-FAK Signaling Activation.","authors":"Junjiro Yoshida, Tomokazu Ohishi, Isao Momose, Shun-Ichi Ohba, Kyohei Kurosawa, Akiko Harakawa, Hiroyuki Inoue, Minori Senoo, Daisuke Tatsuda, Hikaru Abe, Atsushi Masamune, Manabu Kawada","doi":"10.1111/cas.70318","DOIUrl":"https://doi.org/10.1111/cas.70318","url":null,"abstract":"<p><p>Tumor tissues in pancreatic cancer develop with abundant cancer-associated fibroblasts (CAFs), promoting tumor progression. CAF-conditioned medium induces the expression of sushi domain-containing 2 (SUSD2) and enhances the invasive potential of pancreatic cancer cells. We showed that SUSD2 binds to integrin β1 and promotes pancreatic cancer cell motility by inducing phosphorylation of focal adhesion kinase (FAK), facilitating the formation of focal adhesion complexes in cells adhered to collagen 1 or fibronectin. Orthotopic transplantation of SUSD2-overexpressing human pancreatic cancer cell lines into the mouse pancreas enhanced liver metastasis in Panc-1 cells, whereas in KP2 cells, it increased primary tumor growth without promoting metastasis. In spheroid cultures of KP2 cells, forced SUSD2 expression elevated FAK phosphorylation independently of cell adhesion, suggesting that SUSD2 promotes cell proliferation even in non-metastatic cells. High SUSD2 expression in cancer cells contributes to tumor growth and metastasis, identifying SUSD2 as a potential therapeutic target in pancreatic cancer.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145946653","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PSMD12 Overexpression Promotes Lung Adenocarcinoma Progression via Ubiquitin-Proteasome Pathway Dysregulation. PSMD12过表达通过泛素-蛋白酶体通路失调促进肺腺癌进展。
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-06 DOI: 10.1111/cas.70290
Yuya Ono, Hajime Otsu, Takaaki Masuda, Keisuke Kosai, Shohei Shibuta, Kosuke Hirose, Takashi Ofuchi, Yuki Ando, Koto Kawata, Yasuo Tsuda, Yusuke Yonemura, Taro Tobo, Tomoyoshi Takenaka, Tomoharu Yoshizumi, Koshi Mimori

Lung adenocarcinoma (LUAD) is one of the most common cancers and a leading cause of cancer-related mortality worldwide, highlighting the need for novel therapeutic strategies. Proteasome 26S Subunit, Non-ATPase 12 (PSMD12), a component of the proteasomal 19S regulatory particle, is associated with tumorigenesis; however, its role in LUAD remains poorly understood. Integrative bioinformatic analysis of The Cancer Genome Atlas (TCGA) and other publicly available LUAD datasets identified PSMD12 as a candidate driver gene on chromosome 17q, a region frequently amplified in LUAD. Clinicopathological and prognostic analyses revealed that PSMD12 was significantly upregulated in tumor tissues because of DNA copy number gain. High PSMD12 expression was associated with poor prognosis and advanced pathological stages. Gene set enrichment analysis of TCGA LUAD dataset demonstrated that samples with high PSMD12 expression were enriched for cell cycle-related pathways. Using CRISPR-Cas9-mediated PSMD12 knockout and lentivirus-mediated overexpression models, we demonstrated that PSMD12 promoted tumor cell proliferation by accelerating the G2/M cell cycle transition in vitro, and xenograft experiments confirmed its tumor-promoting effect in vivo. Mechanistically, PSMD12 overexpression reduced the ubiquitination of CDK1, a key regulator of mitotic entry. Cycloheximide chase and MG132 assays confirmed that PSMD12 stabilized CDK1 by inhibiting proteasome-mediated degradation. In conclusion, we identified PSMD12 as a novel driver gene and prognostic biomarker of LUAD. PSMD12 promoted LUAD progression by modulating CDK1 ubiquitination and enhancing cell cycle progression. These findings suggest that PSMD12 is a promising molecular target for future LUAD therapies.

肺腺癌(LUAD)是最常见的癌症之一,也是全球癌症相关死亡的主要原因,强调了对新型治疗策略的需求。蛋白酶体26S亚基,非atp酶12 (PSMD12),蛋白酶体19S调节颗粒的一个组成部分,与肿瘤发生有关;然而,它在LUAD中的作用仍然知之甚少。对癌症基因组图谱(TCGA)和其他公开的LUAD数据集的综合生物信息学分析发现,PSMD12是LUAD中经常扩增的染色体17q上的候选驱动基因。临床病理和预后分析显示PSMD12在肿瘤组织中由于DNA拷贝数增加而显著上调。PSMD12高表达与预后差、病理分期晚期相关。TCGA LUAD数据集的基因集富集分析表明,PSMD12高表达的样本富集了细胞周期相关通路。通过crispr - cas9介导的PSMD12基因敲除和慢病毒介导的过表达模型,我们在体外证明了PSMD12通过加速G2/M细胞周期转变促进肿瘤细胞增殖,并且异种移植实验证实了其在体内的促瘤作用。从机制上讲,PSMD12过表达降低了CDK1的泛素化,CDK1是有丝分裂进入的关键调节因子。环己亚胺追踪和MG132实验证实PSMD12通过抑制蛋白酶体介导的降解来稳定CDK1。总之,我们确定PSMD12是LUAD的一个新的驱动基因和预后生物标志物。PSMD12通过调节CDK1泛素化和促进细胞周期进程促进LUAD进展。这些发现表明PSMD12是未来LUAD治疗的一个有希望的分子靶点。
{"title":"PSMD12 Overexpression Promotes Lung Adenocarcinoma Progression via Ubiquitin-Proteasome Pathway Dysregulation.","authors":"Yuya Ono, Hajime Otsu, Takaaki Masuda, Keisuke Kosai, Shohei Shibuta, Kosuke Hirose, Takashi Ofuchi, Yuki Ando, Koto Kawata, Yasuo Tsuda, Yusuke Yonemura, Taro Tobo, Tomoyoshi Takenaka, Tomoharu Yoshizumi, Koshi Mimori","doi":"10.1111/cas.70290","DOIUrl":"https://doi.org/10.1111/cas.70290","url":null,"abstract":"<p><p>Lung adenocarcinoma (LUAD) is one of the most common cancers and a leading cause of cancer-related mortality worldwide, highlighting the need for novel therapeutic strategies. Proteasome 26S Subunit, Non-ATPase 12 (PSMD12), a component of the proteasomal 19S regulatory particle, is associated with tumorigenesis; however, its role in LUAD remains poorly understood. Integrative bioinformatic analysis of The Cancer Genome Atlas (TCGA) and other publicly available LUAD datasets identified PSMD12 as a candidate driver gene on chromosome 17q, a region frequently amplified in LUAD. Clinicopathological and prognostic analyses revealed that PSMD12 was significantly upregulated in tumor tissues because of DNA copy number gain. High PSMD12 expression was associated with poor prognosis and advanced pathological stages. Gene set enrichment analysis of TCGA LUAD dataset demonstrated that samples with high PSMD12 expression were enriched for cell cycle-related pathways. Using CRISPR-Cas9-mediated PSMD12 knockout and lentivirus-mediated overexpression models, we demonstrated that PSMD12 promoted tumor cell proliferation by accelerating the G2/M cell cycle transition in vitro, and xenograft experiments confirmed its tumor-promoting effect in vivo. Mechanistically, PSMD12 overexpression reduced the ubiquitination of CDK1, a key regulator of mitotic entry. Cycloheximide chase and MG132 assays confirmed that PSMD12 stabilized CDK1 by inhibiting proteasome-mediated degradation. In conclusion, we identified PSMD12 as a novel driver gene and prognostic biomarker of LUAD. PSMD12 promoted LUAD progression by modulating CDK1 ubiquitination and enhancing cell cycle progression. These findings suggest that PSMD12 is a promising molecular target for future LUAD therapies.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145913518","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction to "Programmed Death Ligand 1 Regulates Epithelial-Mesenchymal Transition and Cancer Stem Cell Phenotypes in Hepatocellular Carcinoma Through the Serum and Glucocorticoid Kinase 2/β-Catenin Signaling Pathway". 对“程序性死亡配体1通过血清和糖皮质激素激酶2/β-Catenin信号通路调节肝癌上皮-间质转化和癌症干细胞表型”的更正。
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-05 DOI: 10.1111/cas.70316
{"title":"Correction to \"Programmed Death Ligand 1 Regulates Epithelial-Mesenchymal Transition and Cancer Stem Cell Phenotypes in Hepatocellular Carcinoma Through the Serum and Glucocorticoid Kinase 2/β-Catenin Signaling Pathway\".","authors":"","doi":"10.1111/cas.70316","DOIUrl":"https://doi.org/10.1111/cas.70316","url":null,"abstract":"","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145906865","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Longitudinal Impact of Urinary Diversion on Health-Related Quality of Life After Radical Cystectomy: A Multicenter Study in Japan. 泌尿分流对根治性膀胱切除术后健康相关生活质量的纵向影响:日本的一项多中心研究
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2026-01-04 DOI: 10.1111/cas.70289
Shuhei Yamada, Miho Sato, Takahiro Osawa, Toru Harabayashi, Jun Miki, Takashi Kobayashi, Katsuyoshi Hashine, Atsunari Kawashima, Takashi Matsumoto, Takanori Mochizuki, Rikiya Taoka, Fumihiko Urabe, Shuichi Tatarano, Atsuro Sawada, Takahiro Kojima, Atsushi Takahashi, Akira Yokomizo, Shigetaka Suekane, Kohei Hashimoto, Yasuhiro Hashimoto, Junji Yatsuda, Ken Morita, Keita Kobayashi, Yohei Satake, Ataru Sazawa, Yoshiyuki Matsui, Yoichi M Ito, Sayaka Shimizu, Shunichi Fukuhara, Hiroyuki Nishiyama, Hiroshi Kitamura, Nobuo Shinohara

This multicenter longitudinal study was conducted across 24 institutions in Japan to examine the impact of urinary diversion on health-related quality of life (HRQOL) among bladder cancer patients who underwent radical cystectomy (RC). We evaluated bladder cancer-specific HRQOL and general HRQOL via the bladder cancer index (BCI) and the QOL General (QGEN-8), respectively, before the operation and at 3, 6, and 12 months postoperatively. The scores were compared across urinary diversion groups as well as across different time points within each urinary diversion group with linear mixed-effects models. Data from 227 patients were analyzed (151 with ileal conduits, 45 with ureterostomy, and 31 with neobladders). Neobladder patients were more likely to experience longitudinal impacts of their urinary diversion on urinary function than ileal conduit or ureterostomy patients were. Compared with that at baseline, the bowel function of neobladder patients remained impaired 12 months after surgery. All urinary diversion groups had worse sexual function scores at 3 and 6 months than at baseline, and the ileal conduit and neobladder groups had significantly worse sexual function scores at 12 months than at baseline. On the other hand, there was no significant difference in bother scores in the urinary, bowel, or sexual domain. The generic HRQOL was maintained from the preoperative to the postoperative period in all urinary diversion groups. This study explored longitudinal changes in HRQOL after RC, and the findings may help inform patient counseling regarding possible QOL trajectories.

这项多中心纵向研究在日本的24个机构进行,旨在研究尿分流对接受根治性膀胱切除术(RC)的膀胱癌患者健康相关生活质量(HRQOL)的影响。术前、术后3个月、6个月和12个月分别通过膀胱癌指数(BCI)和一般生活质量(QGEN-8)评估膀胱癌特异性HRQOL和一般HRQOL。采用线性混合效应模型比较尿分流组之间的得分以及每个尿分流组内不同时间点的得分。分析了227例患者的数据(151例回肠导管,45例输尿管造口,31例新膀胱)。与回肠导管或输尿管造口患者相比,新膀胱患者更有可能经历尿改道对泌尿功能的纵向影响。与基线时相比,术后12个月新膀胱患者的肠功能仍然受损。所有尿改道组在3个月和6个月时的性功能评分均低于基线,回肠导管组和新膀胱组在12个月时的性功能评分明显低于基线。另一方面,在尿、肠或性方面的得分没有显著差异。各尿分流组术前至术后HRQOL均保持一般水平。本研究探讨了RC后HRQOL的纵向变化,研究结果可能有助于为患者提供有关可能的QOL轨迹的咨询。
{"title":"Longitudinal Impact of Urinary Diversion on Health-Related Quality of Life After Radical Cystectomy: A Multicenter Study in Japan.","authors":"Shuhei Yamada, Miho Sato, Takahiro Osawa, Toru Harabayashi, Jun Miki, Takashi Kobayashi, Katsuyoshi Hashine, Atsunari Kawashima, Takashi Matsumoto, Takanori Mochizuki, Rikiya Taoka, Fumihiko Urabe, Shuichi Tatarano, Atsuro Sawada, Takahiro Kojima, Atsushi Takahashi, Akira Yokomizo, Shigetaka Suekane, Kohei Hashimoto, Yasuhiro Hashimoto, Junji Yatsuda, Ken Morita, Keita Kobayashi, Yohei Satake, Ataru Sazawa, Yoshiyuki Matsui, Yoichi M Ito, Sayaka Shimizu, Shunichi Fukuhara, Hiroyuki Nishiyama, Hiroshi Kitamura, Nobuo Shinohara","doi":"10.1111/cas.70289","DOIUrl":"https://doi.org/10.1111/cas.70289","url":null,"abstract":"<p><p>This multicenter longitudinal study was conducted across 24 institutions in Japan to examine the impact of urinary diversion on health-related quality of life (HRQOL) among bladder cancer patients who underwent radical cystectomy (RC). We evaluated bladder cancer-specific HRQOL and general HRQOL via the bladder cancer index (BCI) and the QOL General (QGEN-8), respectively, before the operation and at 3, 6, and 12 months postoperatively. The scores were compared across urinary diversion groups as well as across different time points within each urinary diversion group with linear mixed-effects models. Data from 227 patients were analyzed (151 with ileal conduits, 45 with ureterostomy, and 31 with neobladders). Neobladder patients were more likely to experience longitudinal impacts of their urinary diversion on urinary function than ileal conduit or ureterostomy patients were. Compared with that at baseline, the bowel function of neobladder patients remained impaired 12 months after surgery. All urinary diversion groups had worse sexual function scores at 3 and 6 months than at baseline, and the ileal conduit and neobladder groups had significantly worse sexual function scores at 12 months than at baseline. On the other hand, there was no significant difference in bother scores in the urinary, bowel, or sexual domain. The generic HRQOL was maintained from the preoperative to the postoperative period in all urinary diversion groups. This study explored longitudinal changes in HRQOL after RC, and the findings may help inform patient counseling regarding possible QOL trajectories.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2026-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145901560","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Expression of the CXCR4 S338X Variant Improves Anti-Leukemia Efficacy of Anti-CD19 CAR-T Cells. 表达CXCR4 S338X变体提高抗cd19 CAR-T细胞抗白血病疗效
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2025-12-30 DOI: 10.1111/cas.70313
Yushu Mao, Xiaodan Wang, Chun-Hui Jin, Zhifeng Wei, Qinghao Ding, Ke-Ke Zhang, Bo-Wen Dong, Kai-Wen Zheng, Yufei Hou, Tao Zhang, Wen-Jie Zhao, Zheng Hu, Yong-Guang Yang

Chimeric antigen receptor T (CAR-T) cell therapy has demonstrated remarkable success in treating hematological malignancies; however, antigen-positive relapse remains a significant obstacle to achieving sustained remission in B-cell acute lymphoblastic leukemia (B-ALL). To enhance the therapeutic efficacy of anti-CD19 CAR (CAR19)-T cells, we overexpressed CXCR4 in CAR19-T cells to improve their trafficking to bone marrow (BM), a key sanctuary for minimal residual disease. We engineered CAR19-T cells with overexpression of wild-type CXCR4 (CAR19/CXCR4WT-T) or gain-of-function CXCR4 S338X mutant (CAR19/CXCR4S338X-T). Both CAR19/CXCR4WT-T and CAR19/CXCR4S338X-T cells exhibited enhanced CXCR4 surface expression in vitro and in vivo compared to control CAR19-T cells, with the latter showing significantly superior improvements under all tested conditions, including engagement with CAR-specific antigens or CXCR4 ligand CXCL12. Upon engagement with CXCL12, CAR19/CXCR4S338X-T cells, but not CAR19/CXCR4WT-T cells, displayed significantly increased activation of ERK1/2 and AKT signaling pathways, as well as elevated transcription of TNF-α, IFN-γ, granzyme B, CDK6, and BCL2A1, along with strengthened effector functions, chemotaxis, and activation of anti-apoptotic pathways. Furthermore, CAR19/CXCR4S338X-T cells demonstrated significantly improved migration to and retention in the BM accompanied by increased CD45RA+CCR7+ memory T cell populations, which correlated with enhanced anti-leukemic effects following injection into B-ALL-bearing mice. This study offers a potentially effective strategy to improve the functionality and durability of CAR-T cell responses in hematological malignancies.

嵌合抗原受体T (CAR-T)细胞疗法在治疗血液系统恶性肿瘤方面取得了显著成功;然而,抗原阳性复发仍然是实现b细胞急性淋巴细胞白血病(B-ALL)持续缓解的一个重大障碍。为了增强抗cd19 CAR (CAR19)-T细胞的治疗效果,我们在CAR19-T细胞中过表达CXCR4,以改善它们向骨髓(BM)的运输,骨髓是最小残留疾病的关键庇护所。我们设计了过表达野生型CXCR4 (CAR19/CXCR4WT-T)或功能获得型CXCR4S338X突变体(CAR19/CXCR4S338X-T)的CAR19- t细胞。与对照CAR19- t细胞相比,CAR19/CXCR4WT-T细胞和CAR19/CXCR4S338X-T细胞在体外和体内均表现出增强的CXCR4表面表达,后者在所有测试条件下均表现出显著的改善,包括与car特异性抗原或CXCR4配体CXCL12结合。与CXCL12结合后,CAR19/CXCR4S338X-T细胞,而不是CAR19/CXCR4WT-T细胞,表现出显著增加的ERK1/2和AKT信号通路的激活,以及TNF-α、IFN-γ、颗粒酶B、CDK6和BCL2A1的转录升高,同时增强了效应功能、趋化性和抗凋亡通路的激活。此外,CAR19/CXCR4S338X-T细胞在BM中的迁移和滞留显著改善,同时CD45RA+CCR7+记忆T细胞群增加,这与注射到携带b - all的小鼠后抗白血病作用增强相关。这项研究提供了一种潜在的有效策略来改善血液恶性肿瘤中CAR-T细胞反应的功能和持久性。
{"title":"Expression of the CXCR4 S338X Variant Improves Anti-Leukemia Efficacy of Anti-CD19 CAR-T Cells.","authors":"Yushu Mao, Xiaodan Wang, Chun-Hui Jin, Zhifeng Wei, Qinghao Ding, Ke-Ke Zhang, Bo-Wen Dong, Kai-Wen Zheng, Yufei Hou, Tao Zhang, Wen-Jie Zhao, Zheng Hu, Yong-Guang Yang","doi":"10.1111/cas.70313","DOIUrl":"https://doi.org/10.1111/cas.70313","url":null,"abstract":"<p><p>Chimeric antigen receptor T (CAR-T) cell therapy has demonstrated remarkable success in treating hematological malignancies; however, antigen-positive relapse remains a significant obstacle to achieving sustained remission in B-cell acute lymphoblastic leukemia (B-ALL). To enhance the therapeutic efficacy of anti-CD19 CAR (CAR19)-T cells, we overexpressed CXCR4 in CAR19-T cells to improve their trafficking to bone marrow (BM), a key sanctuary for minimal residual disease. We engineered CAR19-T cells with overexpression of wild-type CXCR4 (CAR19/CXCR4<sup>WT</sup>-T) or gain-of-function CXCR4 S338X mutant (CAR19/CXCR4<sup>S338X</sup>-T). Both CAR19/CXCR4<sup>WT</sup>-T and CAR19/CXCR4<sup>S338X</sup>-T cells exhibited enhanced CXCR4 surface expression in vitro and in vivo compared to control CAR19-T cells, with the latter showing significantly superior improvements under all tested conditions, including engagement with CAR-specific antigens or CXCR4 ligand CXCL12. Upon engagement with CXCL12, CAR19/CXCR4<sup>S338X</sup>-T cells, but not CAR19/CXCR4<sup>WT</sup>-T cells, displayed significantly increased activation of ERK1/2 and AKT signaling pathways, as well as elevated transcription of TNF-α, IFN-γ, granzyme B, CDK6, and BCL2A1, along with strengthened effector functions, chemotaxis, and activation of anti-apoptotic pathways. Furthermore, CAR19/CXCR4<sup>S338X</sup>-T cells demonstrated significantly improved migration to and retention in the BM accompanied by increased CD45RA<sup>+</sup>CCR7<sup>+</sup> memory T cell populations, which correlated with enhanced anti-leukemic effects following injection into B-ALL-bearing mice. This study offers a potentially effective strategy to improve the functionality and durability of CAR-T cell responses in hematological malignancies.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145858776","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Genotype-Phenotype Correlations of Li-Fraumeni Syndrome in Japan Children's Cancer Group LFS20 Study Cohort. 日本儿童癌症组LFS20研究队列中Li-Fraumeni综合征的基因型-表型相关性
IF 4.3 2区 医学 Q1 Medicine Pub Date : 2025-12-29 DOI: 10.1111/cas.70302
Fumito Yamazaki, Yoshiko Nakano, Masashi Sanada, Hiroki Kurahashi, Shunsuke Miyai, Arisa Ueki, Yuko Watanabe, Daisuke Hasegawa, Shuhei Karakawa, Toshifumi Ozaki, Akira Hirasawa, Akiko M Saito, Eisuke Inoue, Motohiro Kato, Hiroyoshi Hattori

Li-Fraumeni syndrome (LFS) is a cancer predisposition syndrome caused by germline pathogenic variants in the TP53 gene. With the increasing use of multi-gene panel testing, TP53 variants have been identified in individuals who do not meet established TP53 testing criteria, such as the Chompret criteria. The term "attenuated LFS" has been proposed for some of these cases, particularly those with adult-onset cancer. We analyzed participants of the Japanese nationwide prospective clinical trial of the cancer surveillance program (Japan Children's Cancer Group LFS-20), along with clinical information including their family histories, to better understand their genotypic and phenotypic characteristics. We identified 32 distinct TP53 variants from 41 families (45 participants), including four missense variants with conflicting classifications of pathogenicity in ClinVar. Among these families, 36 (88%) met the LFS criteria (hereafter referred to as "LFS" in contrast to attenuated LFS), while 5 (12%) were classified as attenuated LFS. Including 30 additional family members carrying the same variant, we analyzed 75 individuals with TP53 variants. Of these, 40 with LFS and 6 with attenuated LFS had cancer. Multiple primary cancers occurred in 22 individuals (21 LFS, 1 attenuated LFS). LFS-core tumors accounted for 66% (58/88) of cancers in the LFS group and 63% (5/8) in the attenuated LFS group; of note, all core tumors in the attenuated group were limited to breast cancer. Hotspot missense variants were detected in 11 of 36 LFS families and in none of 5 attenuated LFS families, and non-hotspot null variants were found in 14 and 1, respectively. Our study revealed genotype-phenotype correlations in several respects. UMIN-CTR: UMIN000045855.

Li-Fraumeni综合征(LFS)是一种由TP53基因的种系致病性变异引起的癌症易感综合征。随着多基因面板检测的使用越来越多,在不符合既定TP53检测标准(如Chompret标准)的个体中发现了TP53变异。“减毒LFS”一词已被提出用于其中一些病例,特别是那些成人发病的癌症。我们分析了日本全国癌症监测项目前瞻性临床试验(日本儿童癌症组LFS-20)的参与者,以及包括家族史在内的临床信息,以更好地了解他们的基因型和表型特征。我们从41个家族(45名参与者)中鉴定出32种不同的TP53变体,包括4种在ClinVar中具有相互冲突的致病性分类的错义变体。在这些家庭中,36个(88%)符合LFS标准(以下简称“LFS”,与减弱的LFS相对),5个(12%)属于减弱的LFS。包括另外30名携带相同变体的家庭成员,我们分析了75名携带TP53变体的个体。其中,40名LFS患者和6名LFS减弱患者发生了癌症。22例发生多发性原发癌(21例LFS, 1例减弱LFS)。LFS组中LFS核心肿瘤占66% (58/88),LFS减毒组中占63% (5/8);值得注意的是,减毒组的所有核心肿瘤仅限于乳腺癌。36个LFS家族中有11个存在热点错义变异,5个减弱LFS家族中没有发现热点错义变异,14个和1个LFS家族中分别存在非热点零变异。我们的研究揭示了基因型-表型在几个方面的相关性。UMIN-CTR: UMIN000045855。
{"title":"Genotype-Phenotype Correlations of Li-Fraumeni Syndrome in Japan Children's Cancer Group LFS20 Study Cohort.","authors":"Fumito Yamazaki, Yoshiko Nakano, Masashi Sanada, Hiroki Kurahashi, Shunsuke Miyai, Arisa Ueki, Yuko Watanabe, Daisuke Hasegawa, Shuhei Karakawa, Toshifumi Ozaki, Akira Hirasawa, Akiko M Saito, Eisuke Inoue, Motohiro Kato, Hiroyoshi Hattori","doi":"10.1111/cas.70302","DOIUrl":"https://doi.org/10.1111/cas.70302","url":null,"abstract":"<p><p>Li-Fraumeni syndrome (LFS) is a cancer predisposition syndrome caused by germline pathogenic variants in the TP53 gene. With the increasing use of multi-gene panel testing, TP53 variants have been identified in individuals who do not meet established TP53 testing criteria, such as the Chompret criteria. The term \"attenuated LFS\" has been proposed for some of these cases, particularly those with adult-onset cancer. We analyzed participants of the Japanese nationwide prospective clinical trial of the cancer surveillance program (Japan Children's Cancer Group LFS-20), along with clinical information including their family histories, to better understand their genotypic and phenotypic characteristics. We identified 32 distinct TP53 variants from 41 families (45 participants), including four missense variants with conflicting classifications of pathogenicity in ClinVar. Among these families, 36 (88%) met the LFS criteria (hereafter referred to as \"LFS\" in contrast to attenuated LFS), while 5 (12%) were classified as attenuated LFS. Including 30 additional family members carrying the same variant, we analyzed 75 individuals with TP53 variants. Of these, 40 with LFS and 6 with attenuated LFS had cancer. Multiple primary cancers occurred in 22 individuals (21 LFS, 1 attenuated LFS). LFS-core tumors accounted for 66% (58/88) of cancers in the LFS group and 63% (5/8) in the attenuated LFS group; of note, all core tumors in the attenuated group were limited to breast cancer. Hotspot missense variants were detected in 11 of 36 LFS families and in none of 5 attenuated LFS families, and non-hotspot null variants were found in 14 and 1, respectively. Our study revealed genotype-phenotype correlations in several respects. UMIN-CTR: UMIN000045855.</p>","PeriodicalId":48943,"journal":{"name":"Cancer Science","volume":" ","pages":""},"PeriodicalIF":4.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145850636","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer Science
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1