Objective:Patient-derived xenograft(PDX)models have shown great promise in preclinical and translational applications,but their consistency with primary tumors in phenotypic,genetic,and pharmacodynamic heterogeneity h...Objective:Patient-derived xenograft(PDX)models have shown great promise in preclinical and translational applications,but their consistency with primary tumors in phenotypic,genetic,and pharmacodynamic heterogeneity has not been well-studied.This study aimed to establish a PDX repository for non-small cell lung cancer(NSCLC)and to further elucidate whether it could preserve the heterogeneity within and between tumors in patients.Methods:A total of 75 surgically resected NSCLC specimens were implanted into immunodeficient NOD/SCID mice.Based on the successful establishment of the NSCLC PDX model,we compared the expressions of vimentin,Ki67,EGFR,and PD-L1 proteins between cancer tissues and PDX models using hematoxylin and eosin staining and immunohistochemical staining.In addition,we detected whole gene expression profiling between primary tumors and PDX generations.We also performed whole exome sequencing(WES)analysis in 17 first generation xenografts to further assess whether PDXs retained the patient heterogeneities.Finally,paclitaxel,cisplatin,doxorubicin,atezolizumab,afatininb,and AZD4547 were used to evaluate the responses of PDX models to the standard-of-care agents.Results:A large collection of serially transplantable PDX models for NSCLC were successfully developed.The histology and pathological immunohistochemistry of PDX xenografts were consistent with the patients’tumor samples.WES and RNA-seq further confirmed that PDX accurately replicated the molecular heterogeneities of primary tumors.Similar to clinical patients,PDX models responded differentially to the standard-of-care treatment,including chemo-,targeted-and immuno-therapeutics.Conclusions:Our established PDX models of NSCLC faithfully reproduced the molecular,histopathological,and therapeutic characteristics,as well as the corresponding tumor heterogeneities,which provides a clinically relevant platform for drug screening,biomarker discovery,and translational research.展开更多
目的:检测巴戟天对人源结肠癌细胞HCT-116移植瘤的抑制作用及初步探讨其作用机制。方法:于雄性裸鼠腋下注射人源结肠癌HCT-116细胞,建立移植瘤模型,随后采用巴戟天乙醇提取物(Ethanol extract of Morinda officinalis,EEMO)(12 mg/kg/d...目的:检测巴戟天对人源结肠癌细胞HCT-116移植瘤的抑制作用及初步探讨其作用机制。方法:于雄性裸鼠腋下注射人源结肠癌HCT-116细胞,建立移植瘤模型,随后采用巴戟天乙醇提取物(Ethanol extract of Morinda officinalis,EEMO)(12 mg/kg/d)和巴戟天生药(Crude drug of Morinda officinalis,CDMO)(200 mg/kg/d)为实验组进行灌胃给药,以5-氟尿嘧啶(5-Fluorouracil,5-FU)为阳性药(30 mg/kg,每3 d给药一次,腹腔注射),以水(一次/d)为对照组,共四组(EEMO组、CDMO组、5-FU组和对照组),持续给药23 d。通过检测各干预组动物的肿瘤重量、肿瘤体积、肿瘤组织病理变化判断巴戟天的抑癌作用;通过检测肿瘤组织血管生成因子(VEGF)、缺氧诱导因子-1α(HIF-1α)和环氧化酶-2(COX-2)的表达水平以及巨噬细胞的极化趋势初步探讨其抑癌途径。结果:EEMO和CDMO能不同程度抑制小鼠移植瘤体积的增长(P<0.05)、降低移植瘤的重量(P<0.01)和降低血清前列腺素E2(PGE2)的水平(P<0.05)。蛋白免疫印迹和免疫组化的检测结果显示EEMO和CDMO可显著下调肿瘤组织VEGF、HIF-1α和COX-2的表达水平(P<0.01和P<0.05)。免疫荧光检测结果显示,EEMO和CDMO极显著增加iNOS的表达水平(P<0.01),并且极显著提高i NOS/CD206的比例(P<0.01)。结论:EEMO和CDMO对小鼠体内的移植瘤发生有抑制作用,可能与其下调VEGF、HIF-1α和COX-2/PGE2信号通路的表达、增强巨噬细胞向M1型极化有关。展开更多
BACKGROUND Pancreatic cancer is a highly malignant tumor of the gastrointestinal system whose emerging resistance to chemotherapy has necessitated the development of novel antitumor treatments.Scoparone,a traditional ...BACKGROUND Pancreatic cancer is a highly malignant tumor of the gastrointestinal system whose emerging resistance to chemotherapy has necessitated the development of novel antitumor treatments.Scoparone,a traditional Chinese medicine monomer with a wide range of pharmacological properties,has attracted considerable attention for its antitumor activity.AIM To explore the potential antitumor effect of scoparone on pancreatic cancer and the possible molecular mechanism of action.METHODS The target genes of scoparone were determined using both the bioinformatics and multiplatform analyses.The effect of scoparone on pancreatic cancer cell proliferation,migration,invasion,cell cycle,and apoptosis was detected in vitro.The expression of hub genes was tested using quantitative reverse transcription polymerase chain reaction(qRT-PCR),and the molecular mechanism was analyzed using Western blot.The in vivo effect of scoparone on pancreatic cancer cell proliferation was detected using a xenograft tumor model in nude mice as well as immunohistochemistry.RESULTS The hub genes involved in the suppression of pancreatic cancer by scoparone were obtained by network bioinformatics analyses using publicly available databases and platforms,including SwissTargetPrediction,STITCH,GeneCards,CTD,STRING,WebGestalt,Cytoscape,and Gepia;AKT1 was confirmed using qRT-PCR to be the hub gene.Cell Counting Kit-8 assay revealed that the viability of Capan-2 and SW1990 cells was significantly reduced by scoparone treatment exhibiting IC50 values of 225.2μmol/L and 209.1μmol/L,respectively.Wound healing and transwell assays showed that scoparone inhibited the migration and invasion of pancreatic cancer cells.Additionally,flow cytometry confirmed that scoparone caused cell cycle arrest and induced apoptosis.Scoparone also increased the expression levels of Bax and cleaved caspase-3,decreased the levels of MMP9 and Bcl-2,and suppressed the phosphorylation of Akt without affecting total PI3K and Akt.Moreover,compared with the control group,xenograft 展开更多
基金supported by the National Natural Science Foundation of China(Grant Nos.81101143,81572617,and 81630101)the Sichuan Province Science and Technology Support Program(Grant Nos.2019JDRC0019 and 2018SZ0009)+2 种基金1.3.5 project for disciplines of excellence,West China Hospital,Sichuan University(Grant No.ZYJC18026)The Science and Technology Project of the Health Planning Committee of Sichuan(Grant No.19PJ242)Chengdu science and technology Support Program(Grant No.2019-YFYF-00090-SN)。
文摘Objective:Patient-derived xenograft(PDX)models have shown great promise in preclinical and translational applications,but their consistency with primary tumors in phenotypic,genetic,and pharmacodynamic heterogeneity has not been well-studied.This study aimed to establish a PDX repository for non-small cell lung cancer(NSCLC)and to further elucidate whether it could preserve the heterogeneity within and between tumors in patients.Methods:A total of 75 surgically resected NSCLC specimens were implanted into immunodeficient NOD/SCID mice.Based on the successful establishment of the NSCLC PDX model,we compared the expressions of vimentin,Ki67,EGFR,and PD-L1 proteins between cancer tissues and PDX models using hematoxylin and eosin staining and immunohistochemical staining.In addition,we detected whole gene expression profiling between primary tumors and PDX generations.We also performed whole exome sequencing(WES)analysis in 17 first generation xenografts to further assess whether PDXs retained the patient heterogeneities.Finally,paclitaxel,cisplatin,doxorubicin,atezolizumab,afatininb,and AZD4547 were used to evaluate the responses of PDX models to the standard-of-care agents.Results:A large collection of serially transplantable PDX models for NSCLC were successfully developed.The histology and pathological immunohistochemistry of PDX xenografts were consistent with the patients’tumor samples.WES and RNA-seq further confirmed that PDX accurately replicated the molecular heterogeneities of primary tumors.Similar to clinical patients,PDX models responded differentially to the standard-of-care treatment,including chemo-,targeted-and immuno-therapeutics.Conclusions:Our established PDX models of NSCLC faithfully reproduced the molecular,histopathological,and therapeutic characteristics,as well as the corresponding tumor heterogeneities,which provides a clinically relevant platform for drug screening,biomarker discovery,and translational research.
文摘目的:检测巴戟天对人源结肠癌细胞HCT-116移植瘤的抑制作用及初步探讨其作用机制。方法:于雄性裸鼠腋下注射人源结肠癌HCT-116细胞,建立移植瘤模型,随后采用巴戟天乙醇提取物(Ethanol extract of Morinda officinalis,EEMO)(12 mg/kg/d)和巴戟天生药(Crude drug of Morinda officinalis,CDMO)(200 mg/kg/d)为实验组进行灌胃给药,以5-氟尿嘧啶(5-Fluorouracil,5-FU)为阳性药(30 mg/kg,每3 d给药一次,腹腔注射),以水(一次/d)为对照组,共四组(EEMO组、CDMO组、5-FU组和对照组),持续给药23 d。通过检测各干预组动物的肿瘤重量、肿瘤体积、肿瘤组织病理变化判断巴戟天的抑癌作用;通过检测肿瘤组织血管生成因子(VEGF)、缺氧诱导因子-1α(HIF-1α)和环氧化酶-2(COX-2)的表达水平以及巨噬细胞的极化趋势初步探讨其抑癌途径。结果:EEMO和CDMO能不同程度抑制小鼠移植瘤体积的增长(P<0.05)、降低移植瘤的重量(P<0.01)和降低血清前列腺素E2(PGE2)的水平(P<0.05)。蛋白免疫印迹和免疫组化的检测结果显示EEMO和CDMO可显著下调肿瘤组织VEGF、HIF-1α和COX-2的表达水平(P<0.01和P<0.05)。免疫荧光检测结果显示,EEMO和CDMO极显著增加iNOS的表达水平(P<0.01),并且极显著提高i NOS/CD206的比例(P<0.01)。结论:EEMO和CDMO对小鼠体内的移植瘤发生有抑制作用,可能与其下调VEGF、HIF-1α和COX-2/PGE2信号通路的表达、增强巨噬细胞向M1型极化有关。
基金Supported by National Natural Science Foundation of China,No.817706555Special Project from the Central Government of Liaoning Province,No.2018107003+6 种基金Liaoning Province Medical Science and Technology Achievements Transformation Foundation,No.2018225120China Postdoctoral Science Foundation,No.2020M670101ZXDoctoral Scientific Research Foundation of Liaoning Province,No.2019-BS-276Science and Technology Program of Shenyang,No.19-112-4-103Youth Support Foundation of China Medical University,No.QGZ2018058Scientific Fund of Shengjing Hospital,No.201801345 Talent Project of Shengjing Hospital,No.52-30C.
文摘BACKGROUND Pancreatic cancer is a highly malignant tumor of the gastrointestinal system whose emerging resistance to chemotherapy has necessitated the development of novel antitumor treatments.Scoparone,a traditional Chinese medicine monomer with a wide range of pharmacological properties,has attracted considerable attention for its antitumor activity.AIM To explore the potential antitumor effect of scoparone on pancreatic cancer and the possible molecular mechanism of action.METHODS The target genes of scoparone were determined using both the bioinformatics and multiplatform analyses.The effect of scoparone on pancreatic cancer cell proliferation,migration,invasion,cell cycle,and apoptosis was detected in vitro.The expression of hub genes was tested using quantitative reverse transcription polymerase chain reaction(qRT-PCR),and the molecular mechanism was analyzed using Western blot.The in vivo effect of scoparone on pancreatic cancer cell proliferation was detected using a xenograft tumor model in nude mice as well as immunohistochemistry.RESULTS The hub genes involved in the suppression of pancreatic cancer by scoparone were obtained by network bioinformatics analyses using publicly available databases and platforms,including SwissTargetPrediction,STITCH,GeneCards,CTD,STRING,WebGestalt,Cytoscape,and Gepia;AKT1 was confirmed using qRT-PCR to be the hub gene.Cell Counting Kit-8 assay revealed that the viability of Capan-2 and SW1990 cells was significantly reduced by scoparone treatment exhibiting IC50 values of 225.2μmol/L and 209.1μmol/L,respectively.Wound healing and transwell assays showed that scoparone inhibited the migration and invasion of pancreatic cancer cells.Additionally,flow cytometry confirmed that scoparone caused cell cycle arrest and induced apoptosis.Scoparone also increased the expression levels of Bax and cleaved caspase-3,decreased the levels of MMP9 and Bcl-2,and suppressed the phosphorylation of Akt without affecting total PI3K and Akt.Moreover,compared with the control group,xenograft