Pub Date : 2024-10-01Epub Date: 2022-12-21DOI: 10.1089/cbr.2022.0039
Caijun Dong, Liangwei Yang, Guofang Zhao
Background: Circular RNAs (circ-RNAs) have been demonstrated to influence initiation, drug resistance, and metastasis of tumors. However, the effects of circular-phosphoglycerate mutase 1 (circ-PGAM1) on matrine resistance in nonsmall cell lung cancer (NSCLC) remain unknown. Materials and Methods: The reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to determine gene expression. The terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and cell colony formation assays were used to evaluate NSCLC apoptosis and cell proliferation after indicated treatments, respectively. Results: circ-PGAM1 was upregulated in human NSCLC cell lines (H1299 and A549) compared with the human normal lung epithelial (BEAS-2B) cells. circ-PGAM1 overexpression reversed the matrine treatment-induced inhibition on proliferation of NSCLC cells (A549 and H1299) and rescued the matrine treatment-stimulated apoptosis of these cells. miR-326 was demonstrated to interact with circ-PGAM1. circ-PGAM1 knockdown enhanced the antitumor effect of matrine on NSCLC cell proliferation and apoptosis, which was reversed by miR-326 inhibition. The authors also identified CXCR5 as a key downstream target of miR-326 in A549 cells. Conclusions: circ-PGAM1 enhances matrine resistance of NSCLC cells through the miR-326/CXCR5 axis. The authors' findings provide new insights into NSCLC-targeted therapy.
{"title":"Circ-PGAM1 Enhances Matrine Resistance of Non-Small Cell Lung Cancer via the miR-326/CXCR5 Axis.","authors":"Caijun Dong, Liangwei Yang, Guofang Zhao","doi":"10.1089/cbr.2022.0039","DOIUrl":"10.1089/cbr.2022.0039","url":null,"abstract":"<p><p><b><i>Background:</i></b> Circular RNAs (circ-RNAs) have been demonstrated to influence initiation, drug resistance, and metastasis of tumors. However, the effects of circular-phosphoglycerate mutase 1 (circ-PGAM1) on matrine resistance in nonsmall cell lung cancer (NSCLC) remain unknown. <b><i>Materials and Methods:</i></b> The reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to determine gene expression. The terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and cell colony formation assays were used to evaluate NSCLC apoptosis and cell proliferation after indicated treatments, respectively. <b><i>Results:</i></b> circ-PGAM1 was upregulated in human NSCLC cell lines (H1299 and A549) compared with the human normal lung epithelial (BEAS-2B) cells. circ-PGAM1 overexpression reversed the matrine treatment-induced inhibition on proliferation of NSCLC cells (A549 and H1299) and rescued the matrine treatment-stimulated apoptosis of these cells. miR-326 was demonstrated to interact with circ-PGAM1. circ-PGAM1 knockdown enhanced the antitumor effect of matrine on NSCLC cell proliferation and apoptosis, which was reversed by miR-326 inhibition. The authors also identified CXCR5 as a key downstream target of miR-326 in A549 cells. <b><i>Conclusions:</i></b> circ-PGAM1 enhances matrine resistance of NSCLC cells through the miR-326/CXCR5 axis. The authors' findings provide new insights into NSCLC-targeted therapy.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"593-599"},"PeriodicalIF":2.4,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10433558","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-08-02DOI: 10.1089/cbr.2024.0078
Yi-Tong Liu, He-Liang Wu, Yan-Dong Su, Yi Wang, Yan Li
Malignant peritoneal mesothelioma (MPeM) is a rare primary malignant tumor originating from peritoneal mesothelial cells. Insufficient specificity of the symptoms and their frequent reappearance following surgery make it challenging to diagnose, creating a need for more efficient treatment options. Natural killer cells (NK cells) are part of the innate immune system and are classified as lymphoid cells. Under the regulation of activating and inhibiting receptors, NK cells secrete various cytokines to exert cytotoxic effects and participate in antiforeign body, antiviral, and antitumor activities. This review provides a comprehensive summary of the specific alterations observed in NK cells following MPeM treatment, including changes in cell number, subpopulation distribution, active receptors, and cytotoxicity. In addition, we summarize the impact of various therapeutic interventions, such as chemotherapy, immunotherapy, and targeted therapy, on NK cell function post-MPeM treatment.
恶性腹膜间皮瘤(MPeM)是一种源自腹膜间皮细胞的罕见原发性恶性肿瘤。由于症状的特异性不足以及术后经常复发,使其诊断具有挑战性,因此需要更有效的治疗方案。自然杀伤细胞(NK 细胞)是先天性免疫系统的一部分,属于淋巴细胞。在激活受体和抑制受体的调节下,NK 细胞分泌各种细胞因子,发挥细胞毒作用,参与抗异物、抗病毒和抗肿瘤活动。本综述全面总结了经 MPeM 处理后在 NK 细胞中观察到的特定变化,包括细胞数量、亚群分布、活性受体和细胞毒性的变化。此外,我们还总结了化疗、免疫疗法和靶向疗法等各种治疗干预措施对 MPeM 治疗后 NK 细胞功能的影响。
{"title":"Development in the Study of Natural Killer Cells for Malignant Peritoneal Mesothelioma Treatment.","authors":"Yi-Tong Liu, He-Liang Wu, Yan-Dong Su, Yi Wang, Yan Li","doi":"10.1089/cbr.2024.0078","DOIUrl":"10.1089/cbr.2024.0078","url":null,"abstract":"<p><p>Malignant peritoneal mesothelioma (MPeM) is a rare primary malignant tumor originating from peritoneal mesothelial cells. Insufficient specificity of the symptoms and their frequent reappearance following surgery make it challenging to diagnose, creating a need for more efficient treatment options. Natural killer cells (NK cells) are part of the innate immune system and are classified as lymphoid cells. Under the regulation of activating and inhibiting receptors, NK cells secrete various cytokines to exert cytotoxic effects and participate in antiforeign body, antiviral, and antitumor activities. This review provides a comprehensive summary of the specific alterations observed in NK cells following MPeM treatment, including changes in cell number, subpopulation distribution, active receptors, and cytotoxicity. In addition, we summarize the impact of various therapeutic interventions, such as chemotherapy, immunotherapy, and targeted therapy, on NK cell function post-MPeM treatment.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"551-561"},"PeriodicalIF":2.4,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141879866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2021-09-02DOI: 10.1089/cbr.2021.0154
Shaoqiang Zhou, Kebao Qian, Shuhui Yu, Yutao Zhao, Qin Shen, Ya Li
Lung adenocarcinoma (ADC) is a common subtype of non-small cell lung cancer. MicroRNAs have been reported to be effective biomarkers for diagnosis and an important target for therapy. MiR-4429 is a newly identified miRNA, which can take part in tumor progression as a tumor inhibitor. Moreover, it is an exosomal miRNA that can be taken by lung ADC cell line A549. Nevertheless, its role in lung ADC has been poorly studied. This research discovered that miR-4429 was low expressed in lung ADC cells. MiR-4429 mimics could alleviate the capacities of cell proliferation and metastasis. The mimics are able to reverse epithelial-mesenchymal transition at the same time. Furthermore, it was verified that miR-4429 could bind to β-catenin and negatively regulate β-catenin expression. Interestingly, SKL2001 can reverse the role of miR-4429 on tumor. Consequently, miR-4429 can inactivate Wnt/β-catenin signaling pathway by targeting β-catenin and prevent oncogene expression in lung ADC cells.
{"title":"MiR-4429 Alleviates Malignant Behaviors of Lung Adenocarcioma Through <i>Wnt/β-Catenin</i> Pathway.","authors":"Shaoqiang Zhou, Kebao Qian, Shuhui Yu, Yutao Zhao, Qin Shen, Ya Li","doi":"10.1089/cbr.2021.0154","DOIUrl":"10.1089/cbr.2021.0154","url":null,"abstract":"<p><p>Lung adenocarcinoma (ADC) is a common subtype of non-small cell lung cancer. MicroRNAs have been reported to be effective biomarkers for diagnosis and an important target for therapy. MiR-4429 is a newly identified miRNA, which can take part in tumor progression as a tumor inhibitor. Moreover, it is an exosomal miRNA that can be taken by lung ADC cell line A549. Nevertheless, its role in lung ADC has been poorly studied. This research discovered that miR-4429 was low expressed in lung ADC cells. MiR-4429 mimics could alleviate the capacities of cell proliferation and metastasis. The mimics are able to reverse epithelial-mesenchymal transition at the same time. Furthermore, it was verified that miR-4429 could bind to <i>β-catenin</i> and negatively regulate <i>β-catenin</i> expression. Interestingly, SKL2001 can reverse the role of miR-4429 on tumor. Consequently, miR-4429 can inactivate <i>Wnt/β-catenin</i> signaling pathway by targeting <i>β-catenin</i> and prevent oncogene expression in lung ADC cells.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"562-572"},"PeriodicalIF":2.4,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39391598","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2021-11-12DOI: 10.1089/cbr.2021.0168
Yang Xue, Mingqiang Diao, Jing Lyu, Kang Li, Long He, Junfeng Chen, Xiangguo Li
Background: Numerous studies have recorded the function of long noncoding RNAs (lncRNAs) in cancer development, including lung adenocarcinoma (LUAD). Previous studies have reported the crucial role of lncRNA PTPRG antisense RNA 1 (PTPRG-AS1) in various cancers. However, the role of PTPRG-AS1 in LUAD remains unknown. Materials and Methods: Real-time quantitative polymerase chain reaction (RT-qPCR) was applied for detecting PTPRG-AS1 expression in LUAD cell lines. Functional assays and in vivo experiments explored cell proliferation, whereas flow cytometry analysis was used to detect cell cycle. In addition, fluorescent in situ hybridization (FISH) and subcellular fractionation assay measured the localization of PTPRG-AS1 in LUAD cells. RNA pulldown, luciferase reporter, and RNA immunoprecipitation (RIP) assays were used to investigate the interaction of PTPRG-AS1/miR-124-3p/cyclin D1 (CCND1) axis. Results: PTPRG-AS1 expression was notably high in LUAD cell lines. PTPRG-AS1 knockdown suppressed cell proliferation and cycle as well as the level of CCND1. Moreover, miR-124-3p was the mutual target of PTPRG-AS1 and CCND1. In addition, PTPRG-AS1 sponged miR-124-3p to upregulate CCND1 in LUAD cells. Moreover, miR-124-3p depletion reversed the suppression of PTPRG-AS1 silence on LUAD cell behaviors, but then CCND1 knockdown countervailed the promoting influence of downregulated miR-124-3p. Conclusions: PTPRG-AS1 propels cell proliferation and cell cycle of LUAD by targeting miR-124-3p/CCND1 axis.
{"title":"Long Noncoding RNAs PTPRG Antisense RNA 1 Targets Cyclin D1 to Facilitate Cell Proliferation in Lung Adenocarcinoma.","authors":"Yang Xue, Mingqiang Diao, Jing Lyu, Kang Li, Long He, Junfeng Chen, Xiangguo Li","doi":"10.1089/cbr.2021.0168","DOIUrl":"10.1089/cbr.2021.0168","url":null,"abstract":"<p><p><b><i>Background:</i></b> Numerous studies have recorded the function of long noncoding RNAs (lncRNAs) in cancer development, including lung adenocarcinoma (LUAD). Previous studies have reported the crucial role of lncRNA PTPRG antisense RNA 1 (PTPRG-AS1) in various cancers. However, the role of PTPRG-AS1 in LUAD remains unknown. <b><i>Materials and Methods:</i></b> Real-time quantitative polymerase chain reaction (RT-qPCR) was applied for detecting PTPRG-AS1 expression in LUAD cell lines. Functional assays and <i>in vivo</i> experiments explored cell proliferation, whereas flow cytometry analysis was used to detect cell cycle. In addition, fluorescent <i>in situ</i> hybridization (FISH) and subcellular fractionation assay measured the localization of PTPRG-AS1 in LUAD cells. RNA pulldown, luciferase reporter, and RNA immunoprecipitation (RIP) assays were used to investigate the interaction of PTPRG-AS1/miR-124-3p/cyclin D1 (<i>CCND1</i>) axis. <b><i>Results:</i></b> PTPRG-AS1 expression was notably high in LUAD cell lines. PTPRG-AS1 knockdown suppressed cell proliferation and cycle as well as the level of <i>CCND1</i>. Moreover, miR-124-3p was the mutual target of PTPRG-AS1 and <i>CCND1</i>. In addition, PTPRG-AS1 sponged miR-124-3p to upregulate <i>CCND1</i> in LUAD cells. Moreover, miR-124-3p depletion reversed the suppression of PTPRG-AS1 silence on LUAD cell behaviors, but then <i>CCND1</i> knockdown countervailed the promoting influence of downregulated miR-124-3p. <b><i>Conclusions:</i></b> PTPRG-AS1 propels cell proliferation and cell cycle of LUAD by targeting miR-124-3p/<i>CCND1</i> axis.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"573-583"},"PeriodicalIF":2.4,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39882219","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Qin Li, Bin Chen, Luz Angela Torres-de la Roche, Zimo Gong, Guilin Wang, Rui Zhuo, Rudy Leon De Wilde, Xiaopeng Chen, Wanwan Wang
Background: Breast cancer (BC) in women is the most common malignancy worldwide, but there is still a lack of validated tools to accurately assess patient prognosis and response to available chemotherapy treatment regimens. Method: We collected ultrasound images and transcriptome data of BC from our breast center and public database. Key ultrasound features were then identified by using the support vector machine (SVM) algorithm and correlated with prognostic genes. Long-term survival-related genes were identified through differential expression analysis, and a prognostic evaluation model was established by using Cox regression. In addition, VPS28 from the model was identified as a promising biomarker for BC. Results: Using univariate logistic regression and SVM algorithms, we identified 12 ultrasound features significantly associated with chemotherapy response. Subsequent correlation and differential expression analyses linked 401 genes to these features, from which five key signature genes were derived using Lasso and multivariate Cox regression models. This signature not only facilitates the stratification of patients into risk-specific treatment pathways but also predicts their chemotherapy response, thus supporting personalized medicine in clinical settings. Notably, VPS28, in the signature, emerged as a significant biomarker, strongly associated with poor prognosis, greater tumor invasiveness, and differing expression across demographic groups. Conclusion: In this study, we use ultrasound genomics to reveal a signature that can provide an effective tool for prognostic assessment and predicting chemotherapy response in patients with BC.
{"title":"Ultrasound Genomics Reveals a Signature for Predicting Breast Cancer Prognosis and Therapy Response.","authors":"Qin Li, Bin Chen, Luz Angela Torres-de la Roche, Zimo Gong, Guilin Wang, Rui Zhuo, Rudy Leon De Wilde, Xiaopeng Chen, Wanwan Wang","doi":"10.1089/cbr.2024.0127","DOIUrl":"10.1089/cbr.2024.0127","url":null,"abstract":"<p><p><b><i>Background:</i></b> Breast cancer (BC) in women is the most common malignancy worldwide, but there is still a lack of validated tools to accurately assess patient prognosis and response to available chemotherapy treatment regimens. <b><i>Method:</i></b> We collected ultrasound images and transcriptome data of BC from our breast center and public database. Key ultrasound features were then identified by using the support vector machine (SVM) algorithm and correlated with prognostic genes. Long-term survival-related genes were identified through differential expression analysis, and a prognostic evaluation model was established by using Cox regression. In addition, <i>VPS28</i> from the model was identified as a promising biomarker for BC. <b><i>Results:</i></b> Using univariate logistic regression and SVM algorithms, we identified 12 ultrasound features significantly associated with chemotherapy response. Subsequent correlation and differential expression analyses linked 401 genes to these features, from which five key signature genes were derived using Lasso and multivariate Cox regression models. This signature not only facilitates the stratification of patients into risk-specific treatment pathways but also predicts their chemotherapy response, thus supporting personalized medicine in clinical settings. Notably, <i>VPS28</i>, in the signature, emerged as a significant biomarker, strongly associated with poor prognosis, greater tumor invasiveness, and differing expression across demographic groups. <b><i>Conclusion:</i></b> In this study, we use ultrasound genomics to reveal a signature that can provide an effective tool for prognostic assessment and predicting chemotherapy response in patients with BC.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142309181","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ruicong Li,Xinyu Zhang,Yanan Ge,Zhen Zhao,Liangliang Feng,Xiaoming Li
Dihydroartemisinin (DHA), an artemisinin derivative, can influence certain malignancies' inflammatory response and growth. This study used Cell Counting Kit-8 and Transwell assays to show that DHA suppressed the growth, migration, and invasion of medullary thyroid cancer cells. Furthermore, the authors used enzyme-linked immunosorbent assay, Western blotting, and immunofluorescence to confirm the expression of the transcriptional coactivators Yes-associated protein (YAP)/transcriptional coactivator with a PDZ-binding domain (TAZ) downstream of the Hippo pathway and changes in the expression of the epithelial-mesenchymal transition (EMT) process markers E-cadherin and N-cadherin. These results demonstrate that DHA effectively reduced the expression of interleukin (IL)-6 in medullary thyroid carcinoma (MTC) cells and hindered the EMT process by regulating the Hippo pathway. This regulation was achieved by promoting YAP phosphorylation and inhibiting YAP/TAZ protein expression. Additional activation of the Hippo pathway by GA-017 alleviated the inhibitory effect of DHA on IL-6. Hippo pathway activation led to an increase in the expression of E-cadherin, a marker of EMT. In conclusion, DHA was demonstrated to regulate the Hippo pathway by inhibiting IL-6 secretion, leading to the inhibition of EMT in MTC. These findings provide a theoretical foundation for further exploration of the anticancer mechanisms of DHA and offer valuable insights into its potential clinical application as a combinatorial drug.
双氢青蒿素(DHA)是一种青蒿素衍生物,可影响某些恶性肿瘤的炎症反应和生长。这项研究利用细胞计数试剂盒-8 和 Transwell 试验表明,DHA 能抑制甲状腺髓样癌细胞的生长、迁移和侵袭。此外,作者还使用酶联免疫吸附测定法、Western印迹法和免疫荧光法证实了Hippo通路下游转录辅激活因子Yes-associated protein(YAP)/transcriptional coactivator with a PDZ-binding domain(TAZ)的表达,以及上皮-间质转化(EMT)过程标志物E-cadherin和N-cadherin表达的变化。这些结果表明,DHA能有效降低甲状腺髓样癌细胞中白细胞介素(IL)-6的表达,并通过调节Hippo通路阻碍EMT过程。这种调节是通过促进 YAP 磷酸化和抑制 YAP/TAZ 蛋白表达实现的。GA-017 对 Hippo 通路的进一步激活减轻了 DHA 对 IL-6 的抑制作用。Hippo通路的激活导致E-cadherin的表达增加,而E-cadherin是EMT的标志物。总之,DHA可通过抑制IL-6的分泌来调节Hippo通路,从而抑制MTC的EMT。这些发现为进一步探索 DHA 的抗癌机制提供了理论基础,并为其作为组合药物的潜在临床应用提供了宝贵的见解。
{"title":"Dihydroartemisinin Inhibits Epithelial-Mesenchymal Transition Progression in Medullary Thyroid Carcinoma Through the Hippo Signaling Pathway Regulated by Interleukin-6.","authors":"Ruicong Li,Xinyu Zhang,Yanan Ge,Zhen Zhao,Liangliang Feng,Xiaoming Li","doi":"10.1089/cbr.2023.0197","DOIUrl":"https://doi.org/10.1089/cbr.2023.0197","url":null,"abstract":"Dihydroartemisinin (DHA), an artemisinin derivative, can influence certain malignancies' inflammatory response and growth. This study used Cell Counting Kit-8 and Transwell assays to show that DHA suppressed the growth, migration, and invasion of medullary thyroid cancer cells. Furthermore, the authors used enzyme-linked immunosorbent assay, Western blotting, and immunofluorescence to confirm the expression of the transcriptional coactivators Yes-associated protein (YAP)/transcriptional coactivator with a PDZ-binding domain (TAZ) downstream of the Hippo pathway and changes in the expression of the epithelial-mesenchymal transition (EMT) process markers E-cadherin and N-cadherin. These results demonstrate that DHA effectively reduced the expression of interleukin (IL)-6 in medullary thyroid carcinoma (MTC) cells and hindered the EMT process by regulating the Hippo pathway. This regulation was achieved by promoting YAP phosphorylation and inhibiting YAP/TAZ protein expression. Additional activation of the Hippo pathway by GA-017 alleviated the inhibitory effect of DHA on IL-6. Hippo pathway activation led to an increase in the expression of E-cadherin, a marker of EMT. In conclusion, DHA was demonstrated to regulate the Hippo pathway by inhibiting IL-6 secretion, leading to the inhibition of EMT in MTC. These findings provide a theoretical foundation for further exploration of the anticancer mechanisms of DHA and offer valuable insights into its potential clinical application as a combinatorial drug.","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":"213 1","pages":""},"PeriodicalIF":3.4,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142257369","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Riptee Thakur,Manoj Kumar,Aishwarya Kumar,Raman Kumar Joshi,Divya Maheshwari,Afsal Mohammed Km,Manjunatha Venkataswamy,Bhabani Mohanty,Pradip Chaudhari,Hosahalli K Mohan,Pardeep Kumar
Introduction: Many studies have reported the role of P-glycoprotein (Pgp) in chemoresistance in various pathological conditions such as cancer and neurodegenerative diseases, such as Alzheimer's. In this study, we are reporting the high-performance liquid chromatography (HPLC)-based purification of fluorine-18 [18F]AVT-011 and its preclinical evaluation. Methods: AVT-011 was labeled with 18F using the nucleophilic substitution method by heating the reaction mixture at 110°C for 10 min, followed by purification using preparative HPLC and C18ec cartridge. The in vitro cell uptake study was carried out in U87 cells with and without an inhibitor. The preclinical toxicity was carried out in CD1 mice in three groups, including control, AVT-011 treated, and [18F]AVT-011 treated. The biodistribution study was done in CD1 mice (n = 12) after intravenous injection of 4-6 MBq [18F]AVT-011, and mice were sacrificed at various time intervals. A dose of 3.7 ± 0.7 MBq of [18F]AVT-011 was injected intravenously in the healthy Swiss albino mice, and the whole-body micro-positron emission tomography was acquired at 0-, 30-, 60-, and 120-min postinjection. Results: The radiochemical purity of [18F]AVT-011 was 97 ± 1.5% as evaluated by radio-HPLC with a yield of 14 ± 2% and was stable up to 95% under in vitro conditions in blood and in vivo conditions up to 4 h. The in vitro cell uptake study showed a significant difference in control (27.4 ± 2.1%) and blocked U987 cells (73.2 ± 3.2%) after incubation of 120 min. The tissue distribution in mice showed the highest uptake in the liver (17.3 ± 2.4%), kidneys (16.6 ± 3.1%), lungs (10.4 ± 2.9%), and spleen (5.6 ± 0.8%) at 15 min, and the activity was washed out with time. The radioactivity cleared through the hepatorenal pathway. The animal imaging study also demonstrates a similar biodistribution pattern. Conclusions: [18F]AVT-011 showed higher specific activity than the cartridge-based method but showed similar biological activity.
{"title":"Synthesis, Preclinical Toxicity, and Biodistribution of [18F]AVT-011 to Assess the P-Glycoprotein Function.","authors":"Riptee Thakur,Manoj Kumar,Aishwarya Kumar,Raman Kumar Joshi,Divya Maheshwari,Afsal Mohammed Km,Manjunatha Venkataswamy,Bhabani Mohanty,Pradip Chaudhari,Hosahalli K Mohan,Pardeep Kumar","doi":"10.1089/cbr.2024.0114","DOIUrl":"https://doi.org/10.1089/cbr.2024.0114","url":null,"abstract":"Introduction: Many studies have reported the role of P-glycoprotein (Pgp) in chemoresistance in various pathological conditions such as cancer and neurodegenerative diseases, such as Alzheimer's. In this study, we are reporting the high-performance liquid chromatography (HPLC)-based purification of fluorine-18 [18F]AVT-011 and its preclinical evaluation. Methods: AVT-011 was labeled with 18F using the nucleophilic substitution method by heating the reaction mixture at 110°C for 10 min, followed by purification using preparative HPLC and C18ec cartridge. The in vitro cell uptake study was carried out in U87 cells with and without an inhibitor. The preclinical toxicity was carried out in CD1 mice in three groups, including control, AVT-011 treated, and [18F]AVT-011 treated. The biodistribution study was done in CD1 mice (n = 12) after intravenous injection of 4-6 MBq [18F]AVT-011, and mice were sacrificed at various time intervals. A dose of 3.7 ± 0.7 MBq of [18F]AVT-011 was injected intravenously in the healthy Swiss albino mice, and the whole-body micro-positron emission tomography was acquired at 0-, 30-, 60-, and 120-min postinjection. Results: The radiochemical purity of [18F]AVT-011 was 97 ± 1.5% as evaluated by radio-HPLC with a yield of 14 ± 2% and was stable up to 95% under in vitro conditions in blood and in vivo conditions up to 4 h. The in vitro cell uptake study showed a significant difference in control (27.4 ± 2.1%) and blocked U987 cells (73.2 ± 3.2%) after incubation of 120 min. The tissue distribution in mice showed the highest uptake in the liver (17.3 ± 2.4%), kidneys (16.6 ± 3.1%), lungs (10.4 ± 2.9%), and spleen (5.6 ± 0.8%) at 15 min, and the activity was washed out with time. The radioactivity cleared through the hepatorenal pathway. The animal imaging study also demonstrates a similar biodistribution pattern. Conclusions: [18F]AVT-011 showed higher specific activity than the cartridge-based method but showed similar biological activity.","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":"164 1","pages":""},"PeriodicalIF":3.4,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142194030","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Exosomal programmed death ligand 1 (PD-L1), an exosomal membrane protein found in many tumor types, is reckoned to help regulate the immune microenvironment. However, the functions and the mechanisms underlying the exosome-mediated regulation of the immune microenvironment in colorectal cancer (CRC) remain unknown. Methods: Western blotting was used to investigate the levels of exosomal PD-L1 in the peripheral blood of patients with CRC and healthy controls. A CRC mouse model was constructed by administering 10 mg/kg azoxymethane (AOM) and dextrane sodium sulfate (DSS) intraperitoneally. The mice were then administered the control or CRC-derived exosomes to examine the regulatory effect of the exosomes on macrophage infiltration and CRC development. In vitro studies, using a coculture system, and flow cytometry analysis were conducted to examine the relationship between the regulatory effect of CRC-derived exosomes on CD4+ T cells and tumor-associated macrophages. RNA-seq and reverse transcription-quantitative polymerase chain reaction assays were used to investigate the mechanisms underlying the regulatory effect of the CRC-derived exosomes on macrophage proliferation and the regulation of the immune microenvironment during CRC development. Results: In patients with CRC, higher levels of exosomal PD-L1 were associated with a more severe form of disease. The treatment of mice with AOM/DSS-induced CRC with CRC-derived exosomes resulted in high levels of macrophage proliferation, increased PD-L1 levels in macrophages, and accelerated CRC progression. Importantly, analysis of an in vitro coculture system and flow cytometry analysis showed that the CRC-derived exosomes transported PD-L1 into macrophages and impaired CD4+ T cell function. Preliminary data suggest that the NF-κb signaling pathway regulates the function of CRC-derived exosomal PD-L1-dependent macrophages. Conclusion: CRC-derived exosomes induce the proliferation of macrophages and increase their PD-L1 levels. They also impair CD4+ T cell function and promote CRC progression. Our findings reveal a novel exosomal PD-L1-mediated crosstalk between the CRC cells and immune cells in the CRC microenvironment.
{"title":"Colorectal Cancer-Derived Exosomes Impair CD4+ T Cell Function and Accelerate Cancer Progression via Macrophage Activation.","authors":"Xiaolong Wang,Liang Chen,Wenwei Zhang,Wei Sun,Jianpeng Huang","doi":"10.1089/cbr.2024.0032","DOIUrl":"https://doi.org/10.1089/cbr.2024.0032","url":null,"abstract":"Background: Exosomal programmed death ligand 1 (PD-L1), an exosomal membrane protein found in many tumor types, is reckoned to help regulate the immune microenvironment. However, the functions and the mechanisms underlying the exosome-mediated regulation of the immune microenvironment in colorectal cancer (CRC) remain unknown. Methods: Western blotting was used to investigate the levels of exosomal PD-L1 in the peripheral blood of patients with CRC and healthy controls. A CRC mouse model was constructed by administering 10 mg/kg azoxymethane (AOM) and dextrane sodium sulfate (DSS) intraperitoneally. The mice were then administered the control or CRC-derived exosomes to examine the regulatory effect of the exosomes on macrophage infiltration and CRC development. In vitro studies, using a coculture system, and flow cytometry analysis were conducted to examine the relationship between the regulatory effect of CRC-derived exosomes on CD4+ T cells and tumor-associated macrophages. RNA-seq and reverse transcription-quantitative polymerase chain reaction assays were used to investigate the mechanisms underlying the regulatory effect of the CRC-derived exosomes on macrophage proliferation and the regulation of the immune microenvironment during CRC development. Results: In patients with CRC, higher levels of exosomal PD-L1 were associated with a more severe form of disease. The treatment of mice with AOM/DSS-induced CRC with CRC-derived exosomes resulted in high levels of macrophage proliferation, increased PD-L1 levels in macrophages, and accelerated CRC progression. Importantly, analysis of an in vitro coculture system and flow cytometry analysis showed that the CRC-derived exosomes transported PD-L1 into macrophages and impaired CD4+ T cell function. Preliminary data suggest that the NF-κb signaling pathway regulates the function of CRC-derived exosomal PD-L1-dependent macrophages. Conclusion: CRC-derived exosomes induce the proliferation of macrophages and increase their PD-L1 levels. They also impair CD4+ T cell function and promote CRC progression. Our findings reveal a novel exosomal PD-L1-mediated crosstalk between the CRC cells and immune cells in the CRC microenvironment.","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":"25 1","pages":""},"PeriodicalIF":3.4,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142194029","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Accumulating studies reveal that m6A RNA methylation plays a critical role in cancer pathogenesis and progression. METTL3 as a m6A methyltransferase acts as an oncogene in multiple malignancies including hepatocellular carcinoma (HCC). However, the role and underlying mechanism by which METTL3 contributes to HCC remain unclear. The association of METTL3 expression with clinicopathological characteristics and prognosis in patients with HCC was assessed by reverse transcription polymerase chain reaction, Western blot, and public TCGA dataset. MTT, colony formation, Transwell assays, and xenograft tumor models were executed to reveal the role of METTL3 in HCC. m6A dot blot, RNA immunoprecipitation (RIP), m6A methylated RIP, and Western blot assays were used to uncover the regulatory mechanism of METTL3 in HCC cells. We found that METTL3 was dramatically upregulated in HCC tissue samples and acted as an independent prognostic factor for poor survival and tumor recurrence in patients with HCC. Silencing of METTL3 repressed cell growth and invasion in vitro and in vivo, but restored expression of METTL3 boosted these effects. Mechanistical investigations revealed that METTL3 could directly interact with FMRP and harbor a positive correlation with FMRP expression. Knockdown of METTL3 reduced FMRP m6A levels as well as its mRNA and protein expression. FMRP overexpression drove cell colony formation and cell invasion and abolished METTL3 knockdown-induced antitumor effects and AKT/mTORC1 signaling inactivation. Elevated expression of FMRP could act as an independent prognostic factor for poor survival and tumor recurrence in patients with HCC. Our findings demonstrate that METTL3-mediated m6A modification of FMRP promotes growth and invasion of HCC cells and may provide a promising therapeutic target for HCC.
{"title":"METTL3-Mediated m6A Modification of FMRP Drives Hepatocellular Carcinoma Progression and Indicates Poor Prognosis.","authors":"Siyuan Fu,Dapeng Sun,Zongyan Wang,Peng Zhu,Wenbin Ding,Jian Huang,Xinggang Guo,Yun Yang,Fangming Gu","doi":"10.1089/cbr.2023.0186","DOIUrl":"https://doi.org/10.1089/cbr.2023.0186","url":null,"abstract":"Accumulating studies reveal that m6A RNA methylation plays a critical role in cancer pathogenesis and progression. METTL3 as a m6A methyltransferase acts as an oncogene in multiple malignancies including hepatocellular carcinoma (HCC). However, the role and underlying mechanism by which METTL3 contributes to HCC remain unclear. The association of METTL3 expression with clinicopathological characteristics and prognosis in patients with HCC was assessed by reverse transcription polymerase chain reaction, Western blot, and public TCGA dataset. MTT, colony formation, Transwell assays, and xenograft tumor models were executed to reveal the role of METTL3 in HCC. m6A dot blot, RNA immunoprecipitation (RIP), m6A methylated RIP, and Western blot assays were used to uncover the regulatory mechanism of METTL3 in HCC cells. We found that METTL3 was dramatically upregulated in HCC tissue samples and acted as an independent prognostic factor for poor survival and tumor recurrence in patients with HCC. Silencing of METTL3 repressed cell growth and invasion in vitro and in vivo, but restored expression of METTL3 boosted these effects. Mechanistical investigations revealed that METTL3 could directly interact with FMRP and harbor a positive correlation with FMRP expression. Knockdown of METTL3 reduced FMRP m6A levels as well as its mRNA and protein expression. FMRP overexpression drove cell colony formation and cell invasion and abolished METTL3 knockdown-induced antitumor effects and AKT/mTORC1 signaling inactivation. Elevated expression of FMRP could act as an independent prognostic factor for poor survival and tumor recurrence in patients with HCC. Our findings demonstrate that METTL3-mediated m6A modification of FMRP promotes growth and invasion of HCC cells and may provide a promising therapeutic target for HCC.","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":"37 1","pages":""},"PeriodicalIF":3.4,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142194028","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: This study aimed to predict therapeutic efficacy among diffuse large B-cell lymphoma (DLBCL) after R-CHOP (-like) therapy using baseline 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) radiomics. Methods: A total of 239 patients with DLBCL were enrolled in this study, with 82 patients having refractory/relapsed disease. The radiomics signatures were developed using a stacking ensemble approach. The efficacy of the radiomics signatures, the National Comprehensive Cancer Network-International Prognostic Index (NCCN-IPI), conventional PET parameters model, and their combinations in assessing refractory/relapse risk were evaluated using receiver operating characteristic (ROC) curves, sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), accuracy and decision curve analysis. Results: The stacking model, along with the integrated model that combines stacking with the NCCN-IPI and SDmax (the distance between the two lesions farthest apart, normalized to the patient's body surface area), showed remarkable predictive capabilities with a high area under the curve (AUC), sensitivity, specificity, PPV, NPV, accuracy, and significant net benefit of the AUC (NB-AUC). Although no significant differences were observed between the combined and stacking models in terms of the AUC in either the training cohort (AUC: 0.992 vs. 0.985, p = 0.139) or the testing cohort (AUC: 0.768 vs. 0.781, p = 0.668), the integrated model exhibited higher values for sensitivity, PPV, NPV, accuracy, and NB-AUC than the stacking model. Conclusion: Baseline PET radiomics could predict therapeutic efficacy in DLBCL after R-CHOP (-like) therapy, with improved predictive performance when incorporating clinical features and SDmax.
研究目的本研究旨在利用基线18F-氟脱氧葡萄糖正电子发射断层扫描(18F-FDG PET)放射组学预测弥漫大B细胞淋巴瘤(DLBCL)接受R-CHOP(类)治疗后的疗效。研究方法本研究共招募了239名DLBCL患者,其中82名患者患有难治性/复发性疾病。放射组学特征是采用堆叠集合方法开发的。使用接收器操作特征曲线(ROC)、灵敏度、特异性、阳性预测值(PPV)、阴性预测值(NPV)、准确性和决策曲线分析评估了放射组学特征、美国国家综合癌症网络-国际预后指数(NCCN-IPI)、传统 PET 参数模型及其组合在评估难治/复发风险方面的功效。结果:堆叠模型以及将堆叠与 NCCN-IPI 和 SDmax(相距最远的两个病灶之间的距离,以患者的体表面积归一化)结合起来的综合模型显示出卓越的预测能力,具有较高的曲线下面积(AUC)、灵敏度、特异性、PPV、NPV、准确性和显著的 AUC 净效益(NB-AUC)。虽然在训练队列(AUC:0.992 vs. 0.985,p = 0.139)或测试队列(AUC:0.768 vs. 0.781,p = 0.668)中,综合模型和堆叠模型的 AUC 均无明显差异,但综合模型的灵敏度、PPV、NPV、准确度和 NB-AUC 值均高于堆叠模型。结论基线PET放射组学可预测R-CHOP(类)治疗后DLBCL的疗效,在结合临床特征和SDmax后,其预测性能有所提高。
{"title":"Baseline <sup>18</sup>F-FDG PET Radiomics Predicting Therapeutic Efficacy of Diffuse Large B-Cell Lymphoma after R-CHOP (-Like) Therapy.","authors":"Fenglian Jing, Xinchao Zhang, Yunuan Liu, Xiaolin Chen, Xinming Zhao, Xiaoshan Chen, Huiqing Yuan, Meng Dai, Na Wang, Jingya Han, Jingmian Zhang","doi":"10.1089/cbr.2024.0115","DOIUrl":"https://doi.org/10.1089/cbr.2024.0115","url":null,"abstract":"<p><p><b><i>Objective:</i></b> This study aimed to predict therapeutic efficacy among diffuse large B-cell lymphoma (DLBCL) after R-CHOP (-like) therapy using baseline <sup>18</sup>F-fluorodeoxyglucose positron emission tomography (<sup>18</sup>F-FDG PET) radiomics. <b><i>Methods:</i></b> A total of 239 patients with DLBCL were enrolled in this study, with 82 patients having refractory/relapsed disease. The radiomics signatures were developed using a stacking ensemble approach. The efficacy of the radiomics signatures, the National Comprehensive Cancer Network-International Prognostic Index (NCCN-IPI), conventional PET parameters model, and their combinations in assessing refractory/relapse risk were evaluated using receiver operating characteristic (ROC) curves, sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), accuracy and decision curve analysis. <b><i>Results:</i></b> The stacking model, along with the integrated model that combines stacking with the NCCN-IPI and SDmax (the distance between the two lesions farthest apart, normalized to the patient's body surface area), showed remarkable predictive capabilities with a high area under the curve (AUC), sensitivity, specificity, PPV, NPV, accuracy, and significant net benefit of the AUC (NB-AUC). Although no significant differences were observed between the combined and stacking models in terms of the AUC in either the training cohort (AUC: 0.992 vs. 0.985, <i>p</i> = 0.139) or the testing cohort (AUC: 0.768 vs. 0.781, <i>p</i> = 0.668), the integrated model exhibited higher values for sensitivity, PPV, NPV, accuracy, and NB-AUC than the stacking model. <b><i>Conclusion:</i></b> Baseline PET radiomics could predict therapeutic efficacy in DLBCL after R-CHOP (-like) therapy, with improved predictive performance when incorporating clinical features and SDmax.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.4,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142127425","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}