首页 > 最新文献

American journal of cancer research最新文献

英文 中文
A novel reduced toxicity conditioning regimen for older myelodysplastic neoplasms patients undergoing haploidentical stem cell transplantation: a prospective cohort study.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/OFXJ3130
Wen-Jing Yu, Yu-Qian Sun, Xiao-Hui Zhang, Lan-Ping Xu, Xiao-Dong Mo, Meng Lv, Xiao-Jun Huang, Yu Wang

A novel reduced-toxicity conditioning (RTC) regimen of busulfan, fludarabine, cyclophosphamide, and antithymocyte globulin (Bu/Flu/Cy/ATG) followed by haploidentical hematopoietic stem cell transplantation (haplo-HSCT) in older patients with hematologic malignancies has been reported and the results was encouraging. However, the safety and efficacy of this regimen was unknown in older myelodysplastic neoplasms (MDS) patients. From January 2018 to December 2021, 68 consecutive older patients (aged over 50) using the RTC regimen for T-cell replete haplo-HSCT (RTC group) at our center were eligible, 68 patients aged under 50 using modified busulfan, cyclophosphamide plus antithymocyte globulin regimen (Bu/Cy/ATG) (Bu/Cy/ATG group) were randomly selected from 223 MDS patients during the same period in a 1:1 ratio matched-pair analysis for patient sex, World Health Organization (WHO) category, international prognostic scoring system (IPSS) risk group, time from diagnosis to HSCT, chemotherapy in advanced, response after chemotherapy, donor sex, infused mononuclear cells and the CD34-positive cell count. The transplant outcomes were also compared between the RTC group and the matched sibling donor (MSD) haploidentical stem cell transplantation (HSCT) with the busulfan and cyclophosphamide (Bu/Cy) conditioning regimen. The cumulative incidences of grade II-IV acute graft versus host disease (aGVHD) in the RTC group were significantly lower than that in the Bu/Cy/ATG group. The 3-year cumulative incidences of treatment related mortality (TRM) in the two groups were 12.3% versus 14.7% (P=0.613). The cumulative incidences of relapse, disease-free survival (DFS) and overall survival (OS) were comparable between the two groups. The outcomes were better in RTC group than those patients received MSD transplant, with lower incidence of TRM, and higher OS and DFS. The advantages were still significant when comparing patients receiving children donors HSCT in RTC group with MSD transplant in survival and TRM. Children donor with the RTC regimen could be a better choice than the MSD HSCT with Bu/Cy regimen for the elderly MDS patients. The encouraging results suggest that the RTC regimen followed by haplo-HSCT is a potentially promising method for older MDS patients. The trail number of the prospective study is "NCT03412409" and the trial URL is "https://clinicaltrials.gov/study/NCT03412409?term=NCT03412409&rank=1".

{"title":"A novel reduced toxicity conditioning regimen for older myelodysplastic neoplasms patients undergoing haploidentical stem cell transplantation: a prospective cohort study.","authors":"Wen-Jing Yu, Yu-Qian Sun, Xiao-Hui Zhang, Lan-Ping Xu, Xiao-Dong Mo, Meng Lv, Xiao-Jun Huang, Yu Wang","doi":"10.62347/OFXJ3130","DOIUrl":"10.62347/OFXJ3130","url":null,"abstract":"<p><p>A novel reduced-toxicity conditioning (RTC) regimen of busulfan, fludarabine, cyclophosphamide, and antithymocyte globulin (Bu/Flu/Cy/ATG) followed by haploidentical hematopoietic stem cell transplantation (haplo-HSCT) in older patients with hematologic malignancies has been reported and the results was encouraging. However, the safety and efficacy of this regimen was unknown in older myelodysplastic neoplasms (MDS) patients. From January 2018 to December 2021, 68 consecutive older patients (aged over 50) using the RTC regimen for T-cell replete haplo-HSCT (RTC group) at our center were eligible, 68 patients aged under 50 using modified busulfan, cyclophosphamide plus antithymocyte globulin regimen (Bu/Cy/ATG) (Bu/Cy/ATG group) were randomly selected from 223 MDS patients during the same period in a 1:1 ratio matched-pair analysis for patient sex, World Health Organization (WHO) category, international prognostic scoring system (IPSS) risk group, time from diagnosis to HSCT, chemotherapy in advanced, response after chemotherapy, donor sex, infused mononuclear cells and the CD34-positive cell count. The transplant outcomes were also compared between the RTC group and the matched sibling donor (MSD) haploidentical stem cell transplantation (HSCT) with the busulfan and cyclophosphamide (Bu/Cy) conditioning regimen. The cumulative incidences of grade II-IV acute graft versus host disease (aGVHD) in the RTC group were significantly lower than that in the Bu/Cy/ATG group. The 3-year cumulative incidences of treatment related mortality (TRM) in the two groups were 12.3% versus 14.7% (P=0.613). The cumulative incidences of relapse, disease-free survival (DFS) and overall survival (OS) were comparable between the two groups. The outcomes were better in RTC group than those patients received MSD transplant, with lower incidence of TRM, and higher OS and DFS. The advantages were still significant when comparing patients receiving children donors HSCT in RTC group with MSD transplant in survival and TRM. Children donor with the RTC regimen could be a better choice than the MSD HSCT with Bu/Cy regimen for the elderly MDS patients. The encouraging results suggest that the RTC regimen followed by haplo-HSCT is a potentially promising method for older MDS patients. The trail number of the prospective study is \"NCT03412409\" and the trial URL is \"https://clinicaltrials.gov/study/NCT03412409?term=NCT03412409&rank=1\".</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"182-194"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815378/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143413003","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Clinicopathologic features and exploration of new molecular mechanisms of radiation-induced bone injury: report of two cases and review of the literature.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/OABR4439
Lide Tao, Chaowen Bai, Xiang Gao, Mingchao Zhang, Xueli Qiu, Yuqian Yao, Shuai Wei, Xushen Zhao, Lijun Wu, Xiaozhong Zhou, Bingchen Shan, Jinyu Bai, Huajian Shan

Radiation-induced bone injury (RBI) has a multifaceted mechanism of occurrence, is influenced by various factors, and is difficult to prevent. Clinicopathologic features and manifestations of disease progression were observed in two patients with distinct fractures: one with a left-sided fourth rib fracture after postoperative radiotherapy for lung cancer, and the other with a right-sided intertrochanteric femur fracture following accidental exposure to a 192Ir radiation source 5 years prior. These two clinical case reports discussed the clinicopathologic features of RBI and disease progression and utilized transcriptome sequencing to explore potential new targets for treating this type of RBI. Both patients exhibited similar incurable osteolytic bone destruction and fatty infiltration in their pathology. It is proposed that XIST, the most significantly upregulated gene identified by transcriptome sequencing analysis, maybe a potential target for understanding these molecular mechanisms.

{"title":"Clinicopathologic features and exploration of new molecular mechanisms of radiation-induced bone injury: report of two cases and review of the literature.","authors":"Lide Tao, Chaowen Bai, Xiang Gao, Mingchao Zhang, Xueli Qiu, Yuqian Yao, Shuai Wei, Xushen Zhao, Lijun Wu, Xiaozhong Zhou, Bingchen Shan, Jinyu Bai, Huajian Shan","doi":"10.62347/OABR4439","DOIUrl":"10.62347/OABR4439","url":null,"abstract":"<p><p>Radiation-induced bone injury (RBI) has a multifaceted mechanism of occurrence, is influenced by various factors, and is difficult to prevent. Clinicopathologic features and manifestations of disease progression were observed in two patients with distinct fractures: one with a left-sided fourth rib fracture after postoperative radiotherapy for lung cancer, and the other with a right-sided intertrochanteric femur fracture following accidental exposure to a <sup>192</sup>Ir radiation source 5 years prior. These two clinical case reports discussed the clinicopathologic features of RBI and disease progression and utilized transcriptome sequencing to explore potential new targets for treating this type of RBI. Both patients exhibited similar incurable osteolytic bone destruction and fatty infiltration in their pathology. It is proposed that XIST, the most significantly upregulated gene identified by transcriptome sequencing analysis, maybe a potential target for understanding these molecular mechanisms.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"209-216"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815379/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143413009","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Establishment and clinical value of a circulating tumor cell system based on a multi-site immune lipid magnetic sphere technique in laryngopharyngeal head and neck tumors.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/MVRG3697
Wei Chen, Qin Lin, Desheng Wang, Wenting Xie, Chunyan Huang, Wenjing Fan, Shipu Wu, Xiaomei Fan, Chen Li

This study aimed to construct multi-site magnetic nanospheres to capture circulating tumor cells (CTCs) from peripheral blood specimens of laryngopharyngeal head and neck tumors. Separated CTCs were used to measure downstream molecular markers and to detect and analyze the disease status. A stable CTC multisite nano-enrichment system was used to determine changes in CTCs levels in Programmed Death-Ligand-1 (PD-L1)-positive patients and to assess the extent of real-time changes in CTCs over the course of the disease in correlation with clinicopathological indicators. The results demonstrated that the constructed immunomagnetic spheres could effectively capture CTCs and that the constructed lipid nanoparticles exhibited high capture efficiency and low cytotoxicity. The results of the concordance or complementarity analyses of PD-L1 expression at the CTC and tissue levels indicated good concordance between the two at up to 70%. The analysis of PD-L1 expression and the changes in CTCs in PD-L1-positive cells plays an auxiliary role in clinical diagnosis and can be used as a dynamic detection index for the course of head, neck, and throat tumor treatment and as a predictor of recurrence risk.

{"title":"Establishment and clinical value of a circulating tumor cell system based on a multi-site immune lipid magnetic sphere technique in laryngopharyngeal head and neck tumors.","authors":"Wei Chen, Qin Lin, Desheng Wang, Wenting Xie, Chunyan Huang, Wenjing Fan, Shipu Wu, Xiaomei Fan, Chen Li","doi":"10.62347/MVRG3697","DOIUrl":"10.62347/MVRG3697","url":null,"abstract":"<p><p>This study aimed to construct multi-site magnetic nanospheres to capture circulating tumor cells (CTCs) from peripheral blood specimens of laryngopharyngeal head and neck tumors. Separated CTCs were used to measure downstream molecular markers and to detect and analyze the disease status. A stable CTC multisite nano-enrichment system was used to determine changes in CTCs levels in Programmed Death-Ligand-1 (PD-L1)-positive patients and to assess the extent of real-time changes in CTCs over the course of the disease in correlation with clinicopathological indicators. The results demonstrated that the constructed immunomagnetic spheres could effectively capture CTCs and that the constructed lipid nanoparticles exhibited high capture efficiency and low cytotoxicity. The results of the concordance or complementarity analyses of PD-L1 expression at the CTC and tissue levels indicated good concordance between the two at up to 70%. The analysis of PD-L1 expression and the changes in CTCs in PD-L1-positive cells plays an auxiliary role in clinical diagnosis and can be used as a dynamic detection index for the course of head, neck, and throat tumor treatment and as a predictor of recurrence risk.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"19-31"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815362/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143413069","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ALKBH1-mediated N6-methyladenosine methylation of TRAF1 promotes osteosarcoma proliferation and metastasis.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/ALXR1853
Zhichao Wang, Yuli Fang, Yang Yu, Haile Pan

Osteosarcoma (OS) is a highly malignant bone tumor with poor prognosis and limited therapeutic options. Recent studies have highlighted the critical role of RNA modifications, particularly N6-methyladenosine (m6A) methylation, in cancer progression. This study aimed to investigate the role of ALKBH1, a m6A demethylase, in the proliferation and metastasis of OS through the regulation of TRAF1. Our findings showed that lower ALKBH1 expression correlates with poorer overall survival in OS patients. Knockdown of ALKBH1 significantly enhanced the proliferation, migration, and clonogenicity of OS cell lines (MG63 and HOS cells), while overexpression had the opposite effects. Transcriptomic analysis revealed that ALKBH1 regulates the expression of key oncogenes, including TRAF1, through m6A methylation. m6A-RIP and qPCR assays further confirmed that overexpression of ALKBH1 significantly decreased the m6A methylation and expression of TRAF1 in both MG63 and HOS cells, and ALKBH1 knockdown had the opposite roles. Combined knockdown of ALKBH1 and TRAF1 further reduced the oncogenic properties of osteosarcoma cells compared to individual knockdown for ALKBH1. In conclusion, ALKBH1 silence promotes osteosarcoma proliferation and metastasis by regulating TRAF1 expression through m6A methylation. Targeting the ALKBH1-TRAF1 axis may provide a novel therapeutic strategy for osteosarcoma.

骨肉瘤(Osteosarcoma,OS)是一种高度恶性的骨肿瘤,预后不良,治疗方案有限。最近的研究强调了 RNA 修饰(尤其是 N6-甲基腺苷(m6A)甲基化)在癌症进展中的关键作用。本研究旨在探讨m6A去甲基化酶ALKBH1通过调控TRAF1在OS增殖和转移中的作用。我们的研究结果表明,ALKBH1的低表达与OS患者较差的总生存率相关。敲除ALKBH1能显著增强OS细胞系(MG63和HOS细胞)的增殖、迁移和克隆性,而过表达则有相反的效果。m6A-RIP和qPCR检测进一步证实,在MG63和HOS细胞中,过表达ALKBH1能明显降低m6A甲基化和TRAF1的表达,而敲除ALKBH1则起相反的作用。与单独敲除ALKBH1相比,联合敲除ALKBH1和TRAF1可进一步降低骨肉瘤细胞的致癌特性。总之,ALKBH1沉默通过m6A甲基化调节TRAF1的表达,从而促进骨肉瘤的增殖和转移。靶向 ALKBH1-TRAF1 轴可能为骨肉瘤提供一种新的治疗策略。
{"title":"ALKBH1-mediated N6-methyladenosine methylation of TRAF1 promotes osteosarcoma proliferation and metastasis.","authors":"Zhichao Wang, Yuli Fang, Yang Yu, Haile Pan","doi":"10.62347/ALXR1853","DOIUrl":"10.62347/ALXR1853","url":null,"abstract":"<p><p>Osteosarcoma (OS) is a highly malignant bone tumor with poor prognosis and limited therapeutic options. Recent studies have highlighted the critical role of RNA modifications, particularly N6-methyladenosine (m6A) methylation, in cancer progression. This study aimed to investigate the role of ALKBH1, a m6A demethylase, in the proliferation and metastasis of OS through the regulation of TRAF1. Our findings showed that lower ALKBH1 expression correlates with poorer overall survival in OS patients. Knockdown of ALKBH1 significantly enhanced the proliferation, migration, and clonogenicity of OS cell lines (MG63 and HOS cells), while overexpression had the opposite effects. Transcriptomic analysis revealed that ALKBH1 regulates the expression of key oncogenes, including TRAF1, through m6A methylation. m6A-RIP and qPCR assays further confirmed that overexpression of ALKBH1 significantly decreased the m6A methylation and expression of TRAF1 in both MG63 and HOS cells, and ALKBH1 knockdown had the opposite roles. Combined knockdown of ALKBH1 and TRAF1 further reduced the oncogenic properties of osteosarcoma cells compared to individual knockdown for ALKBH1. In conclusion, ALKBH1 silence promotes osteosarcoma proliferation and metastasis by regulating TRAF1 expression through m6A methylation. Targeting the ALKBH1-TRAF1 axis may provide a novel therapeutic strategy for osteosarcoma.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"375-389"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815365/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143413018","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Retinoic acid receptor-β deletion in a model of early pancreatic ductal adenocarcinoma (PDAC) tumorigenesis.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/XFOT8509
Eduardo Mere Del Aguila, Xiao-Han Tang, Lorraine J Gudas

Vitamin A (VA, retinol) and its metabolites, including retinoic acid (RA), play a major role in the maintenance of cell populations in the adult pancreas. Pancreatic ductal adenocarcinomas (PDACs) contain lower amounts of VA and express lower levels of retinoic acid receptors (RARs) compared to normal human pancreatic tissues. Our goal was to determine if VA signaling directly impacts molecular events underlying pancreatic carcinogenesis using cell-type specific genetic approaches in mice. We knocked out retinoic acid receptor beta (RAR-β) selectively in pancreatic cells by tamoxifen treatment after crossing these adult RAR-βfl/fl mice with Pdx1/CreER (PCer) and lox-stop-lox KRasG12D transgenic mice. Our data show that the rounds of tamoxifen we used were able to induce the knockout of the RAR-β gene in pancreatic cells in this PCer;KRas;RAR-βfl/fl transgenic model. We detected increases in proteins involved in RA metabolism (CYP26A1, RBP1, and ALDH1A2) in the PCer;RAR-βD/wt pancreata, but the levels of RBP1 and ALDH1A2 were decreased in PCer;RAR-βD (both RAR-β alleles deleted) compared to PCer;KRas;RAR-βD and wild-type pancreata. Ki67 and vimentin proteins exhibited lower levels in the PCer;KRas;RAR-βD and PCer;RAR-βD pancreata compared to wild-type, indicating that deletion of RAR-β reduced cell proliferation in acinar cells. Expression of SOX9, a key protein required for formation and maintenance of PDAC, was higher in PCer;RAR-βD/wt and PCer;RAR-βD pancreata compared to wild-type, indicating that deletion of RAR-β increases SOX9 levels even without the KRas activating mutation. In summary, lack of RAR-β in pancreatic acinar cells reduced cell proliferation and increased SOX9 protein levels in this transgenic model.

{"title":"Retinoic acid receptor-β deletion in a model of early pancreatic ductal adenocarcinoma (PDAC) tumorigenesis.","authors":"Eduardo Mere Del Aguila, Xiao-Han Tang, Lorraine J Gudas","doi":"10.62347/XFOT8509","DOIUrl":"10.62347/XFOT8509","url":null,"abstract":"<p><p>Vitamin A (VA, retinol) and its metabolites, including retinoic acid (RA), play a major role in the maintenance of cell populations in the adult pancreas. Pancreatic ductal adenocarcinomas (PDACs) contain lower amounts of VA and express lower levels of retinoic acid receptors (RARs) compared to normal human pancreatic tissues. Our goal was to determine if VA signaling directly impacts molecular events underlying pancreatic carcinogenesis using cell-type specific genetic approaches in mice. We knocked out retinoic acid receptor beta (RAR-β) selectively in pancreatic cells by tamoxifen treatment after crossing these adult RAR-β<sup>fl/fl</sup> mice with Pdx1/CreER (PC<sup>er</sup>) and lox-stop-lox KRas<sup>G12D</sup> transgenic mice. Our data show that the rounds of tamoxifen we used were able to induce the knockout of the RAR-β gene in pancreatic cells in this PC<sup>er</sup>;KRas;RAR-β<sup>fl/fl</sup> transgenic model. We detected increases in proteins involved in RA metabolism (CYP26A1, RBP1, and ALDH1A2) in the PC<sup>er</sup>;RAR-β<sup>D/wt</sup> pancreata, but the levels of RBP1 and ALDH1A2 were decreased in PC<sup>er</sup>;RAR-β<sup>D</sup> (both RAR-β alleles deleted) compared to PC<sup>er</sup>;KRas;RAR-β<sup>D</sup> and wild-type pancreata. Ki67 and vimentin proteins exhibited lower levels in the PC<sup>er</sup>;KRas;RAR-β<sup>D</sup> and PC<sup>er</sup>;RAR-β<sup>D</sup> pancreata compared to wild-type, indicating that deletion of RAR-β reduced cell proliferation in acinar cells. Expression of SOX9, a key protein required for formation and maintenance of PDAC, was higher in PC<sup>er</sup>;RAR-β<sup>D/wt</sup> and PC<sup>er</sup>;RAR-β<sup>D</sup> pancreata compared to wild-type, indicating that deletion of RAR-β increases SOX9 levels even without the KRas activating mutation. In summary, lack of RAR-β in pancreatic acinar cells reduced cell proliferation and increased SOX9 protein levels in this transgenic model.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"127-140"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815370/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143412772","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of amino acid metabolism in tumor immune microenvironment of colorectal cancer.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/ZSOO2247
Minjing Zhu, Yanyan Hu, Yangjia Gu, Xuedan Lin, Xiang Jiang, Chaoju Gong, Zejun Fang

This review investigates the role of amino acid metabolism in the tumor microenvironment of colorectal cancer (CRC) and explores potential targeted therapeutic strategies. The paper synthesized current research on amino acid metabolism in the colorectal cancer tumor microenvironment, focusing on amino acids such as tryptophan, methionine, glutamine, and arginine. It examined their impact on tumor growth, immune evasion, and patient prognosis, as well as the metabolic reprogramming of tumor cells and complex tumor microenvironment interactions. Aberrant amino acid metabolism was a hallmark of colorectal cancer, influencing tumor proliferation, survival, and invasiveness. Key findings included: Tryptophan metabolism via the kynurenine and serotonin pathways significantly affected immune response and tumor progression in CRC. Methionine influenced T cell function and DNA methylation, playing a critical role in tumor development. Glutamine was extensively used by tumor cells for energy metabolism and supported immune cell function. Arginine metabolism impacted CD8+ T cell functionality and tumor growth. The review also discussed the dual roles of immune cells in the tumor microenvironment and the potential of targeting amino acid metabolic pathways for CRC treatment. In conclusion, amino acid metabolism significantly impacts the colorectal cancer tumor microenvironment and immunity. Understanding these metabolic pathways provides valuable insights into CRC pathogenesis and identifies potential therapeutic targets. Future research should focus on developing treatments that disrupt these metabolic processes to improve patient outcomes in CRC.

{"title":"Role of amino acid metabolism in tumor immune microenvironment of colorectal cancer.","authors":"Minjing Zhu, Yanyan Hu, Yangjia Gu, Xuedan Lin, Xiang Jiang, Chaoju Gong, Zejun Fang","doi":"10.62347/ZSOO2247","DOIUrl":"10.62347/ZSOO2247","url":null,"abstract":"<p><p>This review investigates the role of amino acid metabolism in the tumor microenvironment of colorectal cancer (CRC) and explores potential targeted therapeutic strategies. The paper synthesized current research on amino acid metabolism in the colorectal cancer tumor microenvironment, focusing on amino acids such as tryptophan, methionine, glutamine, and arginine. It examined their impact on tumor growth, immune evasion, and patient prognosis, as well as the metabolic reprogramming of tumor cells and complex tumor microenvironment interactions. Aberrant amino acid metabolism was a hallmark of colorectal cancer, influencing tumor proliferation, survival, and invasiveness. Key findings included: Tryptophan metabolism via the kynurenine and serotonin pathways significantly affected immune response and tumor progression in CRC. Methionine influenced T cell function and DNA methylation, playing a critical role in tumor development. Glutamine was extensively used by tumor cells for energy metabolism and supported immune cell function. Arginine metabolism impacted CD8+ T cell functionality and tumor growth. The review also discussed the dual roles of immune cells in the tumor microenvironment and the potential of targeting amino acid metabolic pathways for CRC treatment. In conclusion, amino acid metabolism significantly impacts the colorectal cancer tumor microenvironment and immunity. Understanding these metabolic pathways provides valuable insights into CRC pathogenesis and identifies potential therapeutic targets. Future research should focus on developing treatments that disrupt these metabolic processes to improve patient outcomes in CRC.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"233-247"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815375/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143412779","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pregnancy is associated with the prognosis of ovarian cancer patients with abdominal metastasis.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/JUJQ9225
Hai-Tao Cui, Qian-Yong Zhu, Hong-Wei Zhao, Hui-Li Liu, Na Wang

This study aims to explore a new approach to reduce the recurrence risk and improve the prognosis of ovarian cancer (OC) patients with abdominal metastasis by analyzing the clinical characteristics and prognostic factors. A total of 292 OC patients with abdominal metastasis, treated at Henan Provincial People's Hospital between 2021 and 2023 were included in this retrospective study. Follow-up was conducted for one year to observe the recurrence, with 285 patients completing the observation. The patients were then categorized into relapsing and non-relapsing groups based on whether they experienced a relapse within one-year follow-up. Independent sample t-tests and χ 2 tests were used for inter-group comparison. Both univariate and multivariate logistic regression analyses were utilized to screen factors affecting recurrence. The variance inflation factor (VIF) was used to analyze whether the variables in the model had multicollinearity. Receiver Operating Characteristic (ROC) curves and nomographs were used to construct models for predicting one-year recurrence in OC patients with abdominal metastasis. Area under curve (AUC) of ROC and Hosmer-Lemeshow goodness of fit test were used to evaluate the accuracy of the model. The prediction model was verified by internal verification and external verification. The number of pregnancies, the number of births, diabetes mellitus, tumor diameter, tumor reduction combined with intraperitoneal chemotherapy, CA-125, HE-4, NLR, PLR, MLR showed association with patient recurrence. Logistic regression analysis revealed that lower pregnancy frequency and elevated levels of CA-125, HE-4, PLR and MLR were independent risk factors for increased risk of recurrence. In addition, the nomogram-based model demonstrated strong predictive accuracy for one-year recurrence. OC patients with abdominal metastasis present diverse clinical manifestations, among which fewer pregnancies and elevated levels of CA-125, HE-4, PLR, and MLR may be independent risk factors for increased risk of recurrence. Individualized interventions based on these prognostic factors are essential to reduce risk and enhance patient quality of life.

{"title":"Pregnancy is associated with the prognosis of ovarian cancer patients with abdominal metastasis.","authors":"Hai-Tao Cui, Qian-Yong Zhu, Hong-Wei Zhao, Hui-Li Liu, Na Wang","doi":"10.62347/JUJQ9225","DOIUrl":"10.62347/JUJQ9225","url":null,"abstract":"<p><p>This study aims to explore a new approach to reduce the recurrence risk and improve the prognosis of ovarian cancer (OC) patients with abdominal metastasis by analyzing the clinical characteristics and prognostic factors. A total of 292 OC patients with abdominal metastasis, treated at Henan Provincial People's Hospital between 2021 and 2023 were included in this retrospective study. Follow-up was conducted for one year to observe the recurrence, with 285 patients completing the observation. The patients were then categorized into relapsing and non-relapsing groups based on whether they experienced a relapse within one-year follow-up. Independent sample t-tests and <i>χ</i> <sup>2</sup> tests were used for inter-group comparison. Both univariate and multivariate logistic regression analyses were utilized to screen factors affecting recurrence. The variance inflation factor (VIF) was used to analyze whether the variables in the model had multicollinearity. Receiver Operating Characteristic (ROC) curves and nomographs were used to construct models for predicting one-year recurrence in OC patients with abdominal metastasis. Area under curve (AUC) of ROC and Hosmer-Lemeshow goodness of fit test were used to evaluate the accuracy of the model. The prediction model was verified by internal verification and external verification. The number of pregnancies, the number of births, diabetes mellitus, tumor diameter, tumor reduction combined with intraperitoneal chemotherapy, CA-125, HE-4, NLR, PLR, MLR showed association with patient recurrence. Logistic regression analysis revealed that lower pregnancy frequency and elevated levels of CA-125, HE-4, PLR and MLR were independent risk factors for increased risk of recurrence. In addition, the nomogram-based model demonstrated strong predictive accuracy for one-year recurrence. OC patients with abdominal metastasis present diverse clinical manifestations, among which fewer pregnancies and elevated levels of CA-125, HE-4, PLR, and MLR may be independent risk factors for increased risk of recurrence. Individualized interventions based on these prognostic factors are essential to reduce risk and enhance patient quality of life.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"168-181"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815367/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143412792","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TRAIL receptor agonist TLY012 in combination with PD-1 inhibition promotes tumor regression in an immune-competent mouse model of pancreatic ductal adenocarcinoma.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/ROAT5658
Anna D Louie, Kelsey E Huntington, Young Lee, Jared Mompoint, Laura Jinxuan Wu, Seulki Lee, Thomas J Miner, Wafik S El-Deiry

Pancreatic ductal adenocarcinoma (PDAC) has an immunosuppressed, apoptosis-resistant phenotype. TLY012 is pegylated recombinant Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL), an orphan drug for chronic pancreatitis and systemic sclerosis. Innate immune TRAIL signaling suppresses cancer. We hypothesized that the combination of immune checkpoint-blocking anti-PD-1 antibody and TLY012 would have synergistic anti-tumor efficacy in immune-competent PDAC-bearing mice. PDAC tumor-bearing C57Bl/6 mice treated with 10 mg/kg anti-mouse PD-1 antibody twice weekly and 10 mg/kg TLY012 three times weekly had reduced tumor growth and tumor volume at 70 days compared to either drug alone (all P < 0.005). B-cell activating factor (BAFF), which promotes PDAC tumors, decreased to 44% of control mice with dual treatment at 7 days and remained decreased at 3 months. Long-term dual treatment showed the highest plasma levels of proinflammatory cytokines interferon-gamma (average 5.6 times control level, P=0.046), CCL5 (average 14.1 times control level, P=0.048), and interleukin-3 (IL-3, average 71.1 times control level, P=0.0053). Flow cytometry showed trends toward decreased circulating regulatory T cells, increased NK cells, and a higher proportion of CD8+ T cells within tumors in the dual treatment group. In summary, the combination of anti-PD-1 and TLY012 prevented the growth of PDAC in an immunocompetent mouse model while increasing tumor-infiltrating CD8+ T cells, decreasing circulating T-regulatory cells and altering plasma cytokine expression of CCL5, interferon-gamma, and IL-3 to promote proinflammatory, antitumor effects. Combining TLY012 and anti-mouse PD-1 modifies immune cell and cytokine levels to induce a more proinflammatory immune environment that contributes to decreased PDAC tumor growth.

{"title":"TRAIL receptor agonist TLY012 in combination with PD-1 inhibition promotes tumor regression in an immune-competent mouse model of pancreatic ductal adenocarcinoma.","authors":"Anna D Louie, Kelsey E Huntington, Young Lee, Jared Mompoint, Laura Jinxuan Wu, Seulki Lee, Thomas J Miner, Wafik S El-Deiry","doi":"10.62347/ROAT5658","DOIUrl":"10.62347/ROAT5658","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) has an immunosuppressed, apoptosis-resistant phenotype. TLY012 is pegylated recombinant Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL), an orphan drug for chronic pancreatitis and systemic sclerosis. Innate immune TRAIL signaling suppresses cancer. We hypothesized that the combination of immune checkpoint-blocking anti-PD-1 antibody and TLY012 would have synergistic anti-tumor efficacy in immune-competent PDAC-bearing mice. PDAC tumor-bearing C57Bl/6 mice treated with 10 mg/kg anti-mouse PD-1 antibody twice weekly and 10 mg/kg TLY012 three times weekly had reduced tumor growth and tumor volume at 70 days compared to either drug alone (all P < 0.005). B-cell activating factor (BAFF), which promotes PDAC tumors, decreased to 44% of control mice with dual treatment at 7 days and remained decreased at 3 months. Long-term dual treatment showed the highest plasma levels of proinflammatory cytokines interferon-gamma (average 5.6 times control level, P=0.046), CCL5 (average 14.1 times control level, P=0.048), and interleukin-3 (IL-3, average 71.1 times control level, P=0.0053). Flow cytometry showed trends toward decreased circulating regulatory T cells, increased NK cells, and a higher proportion of CD8+ T cells within tumors in the dual treatment group. In summary, the combination of anti-PD-1 and TLY012 prevented the growth of PDAC in an immunocompetent mouse model while increasing tumor-infiltrating CD8+ T cells, decreasing circulating T-regulatory cells and altering plasma cytokine expression of CCL5, interferon-gamma, and IL-3 to promote proinflammatory, antitumor effects. Combining TLY012 and anti-mouse PD-1 modifies immune cell and cytokine levels to induce a more proinflammatory immune environment that contributes to decreased PDAC tumor growth.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"286-298"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815385/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143412824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fedratinib and gandotinib induce apoptosis and enhance the efficacy of tyrosine kinase inhibitors in human mast cells.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/TYTU4465
Alina Makeeva, Simona Stivala, Elena Ratti, Laetitia Clauss, Etnik Sheremeti, Michel Arock, Martina Konantz, Karin Hartmann

Mastocytosis is characterized by an abnormal accumulation of mast cells (MC) in various organs. In most patients, the disease is driven by the KIT D816V mutation, leading to activation of the KIT receptor and subsequent downstream signaling, including the JAK/STAT pathway. In recent years, KIT-targeting tyrosine kinase inhibitors (TKI) have emerged for the treatment of systemic mastocytosis; however, the overall response rate is often not sufficient. In this study, we investigated whether targeting the JAK/STAT pathway might be a novel treatment approach in mastocytosis. Using human MC lines carrying the KIT D816V mutation and human primary cord blood-derived MC, we examined the effects of different JAK inhibitors. Our findings revealed that the JAK inhibitors fedratinib and gandotinib decreased viability, reduced proliferation, and induced apoptosis in KIT D816V-positive MC lines (HMC-1.2 and ROSA KIT D816V). In contrast, ruxolitinib, baricitinib, upadacitinib and abrocitinib failed to affect MC functions. Combinatorial treatment with fedratinib, gandotinib and the two TKI avapritinib and midostaurin was more effective than treatment with TKI alone. Fedratinib also induced apoptosis and enhanced the efficacy of TKI in primary cord blood-derived MC. These results indicate that fedratinib and gandotinib, but not the other JAK inhibitors used in this study, can suppress viability and induce apoptosis in KIT D816V-mutant and KIT WT MC and increase effects of TKI. These findings suggest to explore fedratinib and gandotinib as novel treatment option in mastocytosis.

{"title":"Fedratinib and gandotinib induce apoptosis and enhance the efficacy of tyrosine kinase inhibitors in human mast cells.","authors":"Alina Makeeva, Simona Stivala, Elena Ratti, Laetitia Clauss, Etnik Sheremeti, Michel Arock, Martina Konantz, Karin Hartmann","doi":"10.62347/TYTU4465","DOIUrl":"10.62347/TYTU4465","url":null,"abstract":"<p><p>Mastocytosis is characterized by an abnormal accumulation of mast cells (MC) in various organs. In most patients, the disease is driven by the <i>KIT</i> D816V mutation, leading to activation of the KIT receptor and subsequent downstream signaling, including the JAK/STAT pathway. In recent years, KIT-targeting tyrosine kinase inhibitors (TKI) have emerged for the treatment of systemic mastocytosis; however, the overall response rate is often not sufficient. In this study, we investigated whether targeting the JAK/STAT pathway might be a novel treatment approach in mastocytosis. Using human MC lines carrying the <i>KIT</i> D816V mutation and human primary cord blood-derived MC, we examined the effects of different JAK inhibitors. Our findings revealed that the JAK inhibitors fedratinib and gandotinib decreased viability, reduced proliferation, and induced apoptosis in <i>KIT</i> D816V-positive MC lines (HMC-1.2 and ROSA <sup><i>KIT</i> D816V</sup>). In contrast, ruxolitinib, baricitinib, upadacitinib and abrocitinib failed to affect MC functions. Combinatorial treatment with fedratinib, gandotinib and the two TKI avapritinib and midostaurin was more effective than treatment with TKI alone. Fedratinib also induced apoptosis and enhanced the efficacy of TKI in primary cord blood-derived MC. These results indicate that fedratinib and gandotinib, but not the other JAK inhibitors used in this study, can suppress viability and induce apoptosis in <i>KIT</i> D816V-mutant and <i>KIT</i> WT MC and increase effects of TKI. These findings suggest to explore fedratinib and gandotinib as novel treatment option in mastocytosis.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"84-98"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815366/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143413081","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hinokitiol reduces tumor metastasis by regulating epithelial cell adhesion molecule via protein kinase-B/mammalian target of rapamycin signaling pathway.
IF 3.6 3区 医学 Q2 ONCOLOGY Pub Date : 2025-01-15 eCollection Date: 2025-01-01 DOI: 10.62347/UZFZ9554
Pei-Shan Wu, Jing-Ru Weng, Shih-Han Chiu, Li-Hsien Wu, Pei-Hsuan Chen, Yun-Xuan Wang, Po-Yen Chiu, Che-Hsin Lee

Tumor metastasis is the leading cause of death in cancer patients. Epithelial cell adhesion molecule (EpCAM) is abundantly expressed in various malignant tumors and plays a crucial role in cell adhesion, metastasis, proliferation, and differentiation. This study investigated the effects of hinokitiol, a natural tropolone compound known for its antiviral, anti-inflammatory, and antibacterial properties, on tumor growth and metastasis. Specifically, the study focused on the expression of EpCAM in mouse tumor cells treated with hinokitiol. Hinokitiol was administered to mouse melanoma cells (B16F10) and mouse colorectal carcinoma cells (CT26), resulting in a significant decrease in EpCAM expression. Additionally, the protein levels involved in the protein kinase-B/mammalian target of rapamycin (AKT/mTOR) signaling pathway were reduced following hinokitiol treatment. Using wound healing and Transwell assays, the study demonstrated that hinokitiol effectively inhibits cancer cell migration. In vivo experiments were conducted using mice, which were injected intravenously with B16F10 or CT26 cells to induce tumor metastasis. The tumor cells were either treated with hinokitiol or left untreated. The results showed that tumor cells treated with hinokitiol exhibited significantly reduced tumor size and weight in the lungs, as well as prolonged survival, compared to untreated tumor cells. This study concludes that hinokitiol inhibits tumor migration by downregulating EpCAM via the AKT/mTOR signaling pathway and exhibits positive effects in vivo.

肿瘤转移是癌症患者死亡的主要原因。上皮细胞粘附分子(EpCAM)在各种恶性肿瘤中大量表达,在细胞粘附、转移、增殖和分化过程中起着至关重要的作用。本研究调查了以抗病毒、抗炎和抗菌特性而闻名的天然三苯酮化合物 hinokitiol 对肿瘤生长和转移的影响。具体来说,该研究重点关注用桧醇处理的小鼠肿瘤细胞中 EpCAM 的表达。对小鼠黑色素瘤细胞(B16F10)和小鼠结肠直肠癌细胞(CT26)施用桧醇后,EpCAM的表达明显减少。此外,参与蛋白激酶-B/哺乳动物雷帕霉素靶标(AKT/mTOR)信号通路的蛋白质水平在桧醇处理后也有所降低。研究利用伤口愈合和 Transwell 试验证明,桧醇能有效抑制癌细胞迁移。研究人员使用小鼠进行体内实验,向小鼠静脉注射 B16F10 或 CT26 细胞以诱导肿瘤转移。肿瘤细胞要么用 hinokitiol 处理,要么不处理。结果显示,与未经处理的肿瘤细胞相比,经 hinokitiol 处理的肿瘤细胞在肺部的肿瘤大小和重量明显减少,存活时间也有所延长。本研究的结论是,桧醇通过 AKT/mTOR 信号通路下调 EpCAM,从而抑制肿瘤迁移,并在体内发挥积极作用。
{"title":"Hinokitiol reduces tumor metastasis by regulating epithelial cell adhesion molecule via protein kinase-B/mammalian target of rapamycin signaling pathway.","authors":"Pei-Shan Wu, Jing-Ru Weng, Shih-Han Chiu, Li-Hsien Wu, Pei-Hsuan Chen, Yun-Xuan Wang, Po-Yen Chiu, Che-Hsin Lee","doi":"10.62347/UZFZ9554","DOIUrl":"10.62347/UZFZ9554","url":null,"abstract":"<p><p>Tumor metastasis is the leading cause of death in cancer patients. Epithelial cell adhesion molecule (EpCAM) is abundantly expressed in various malignant tumors and plays a crucial role in cell adhesion, metastasis, proliferation, and differentiation. This study investigated the effects of hinokitiol, a natural tropolone compound known for its antiviral, anti-inflammatory, and antibacterial properties, on tumor growth and metastasis. Specifically, the study focused on the expression of EpCAM in mouse tumor cells treated with hinokitiol. Hinokitiol was administered to mouse melanoma cells (B16F10) and mouse colorectal carcinoma cells (CT26), resulting in a significant decrease in EpCAM expression. Additionally, the protein levels involved in the protein kinase-B/mammalian target of rapamycin (AKT/mTOR) signaling pathway were reduced following hinokitiol treatment. Using wound healing and Transwell assays, the study demonstrated that hinokitiol effectively inhibits cancer cell migration. <i>In vivo</i> experiments were conducted using mice, which were injected intravenously with B16F10 or CT26 cells to induce tumor metastasis. The tumor cells were either treated with hinokitiol or left untreated. The results showed that tumor cells treated with hinokitiol exhibited significantly reduced tumor size and weight in the lungs, as well as prolonged survival, compared to untreated tumor cells. This study concludes that hinokitiol inhibits tumor migration by downregulating EpCAM via the AKT/mTOR signaling pathway and exhibits positive effects <i>in vivo</i>.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 1","pages":"59-68"},"PeriodicalIF":3.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11815376/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143413084","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
American journal of cancer research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1