首页 > 最新文献

Cancer Biology & Medicine最新文献

英文 中文
Expert consensus on the diagnosis and treatment of non-small cell lung cancer with MET alteration.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-04-08 DOI: 10.20892/j.issn.2095-3941.2024.0503
Huijing Feng, Yang Xia, Wenxian Wang, Chunwei Xu, Qian Wang, Zhengbo Song, Ziming Li, Jinpu Yu, Wenzhao Zhong, Zhijie Wang, Yongchang Zhang, Jingjing Liu, Shirong Zhang, Xiuyu Cai, Anwen Liu, Wen Li, Ping Zhan, Hongbing Liu, Tangfeng Lyu, Liyun Miao, Lingfeng Min, Gen Lin, Long Huang, Jingping Yuan, Zhansheng Jiang, Xingxiang Pu, Chuangzhou Rao, Dongqing Lyu, Zongyang Yu, Xiaoyan Li, Chuanhao Tang, Chengzhi Zhou, Qi Mei, Hui Guo, Qian Chu, Rui Meng, Xuewen Liu, Jingxun Wu, Jin Zhou, Zhengfei Zhu, Weiwei Pan, Fei Pang, Meizhen Hu, Kai Wang, Fan Wu, Bingwei Xu, Ling Xu, Liping Wang, Youcai Zhu, Jisheng Li, Yanru Xie, Xinqing Lin, Jing Cai, Lin Wang, Yingying Du, Wang Yao, Xuefei Shi, Xiaomin Niu, Dongmei Yuan, Yanwen Yao, Jing Kang, Jiatao Zhang, Chao Zhang, Wenbin Gao, Jianhui Huang, Yinbin Zhang, Pingli Sun, Hong Wang, Mingxiang Ye, Dong Wang, Zhaofeng Wang, Yue Hao, Zheng Wang, Bing Wan, Donglai Lyu, Xiaodong Jiao, Lin Shi, Gang Lan, Shengjie Yang, Yanhong Shang, Yina Wang, Bihui Li, Gang Jin, Kang Zheng, Jun Ma, Wenfeng Li, Zhang Zhang, Zhongwu Li, Yuan Li, Zhefeng Liu, Xuelei Ma, Nong Yang, Lin Wu, Qiming Wang, Guansong Wang, Zhuan Hong, Jiandong Wang, Meiyu Fang, Yong Fang, Xixu Zhu, Yi Shen, Ke Wang, Xiubao Ren, Yiping Zhang, Shenglin Ma, Junping Zhang, Yong Song, Wenfeng Fang, Yuanzhi Lu

Alterations in the mesenchymal-epithelial transition factor (MET) gene are critical drivers of non-small cell lung cancer (NSCLC). In recent years advances in precision therapies targeting MET alterations have significantly expanded treatment options for NSCLC patients. These alterations include MET exon 14 skipping mutations (MET exon 14 skipping), MET gene amplifications, MET point mutations (primarily kinase domain mutations), and MET protein overexpression. Accurate identification of these alterations and appropriate selection of patient populations and targeted therapies are essential for improving clinical outcomes. The East China Lung Cancer Group, Youth Committee (ECLUNG YOUNG, Yangtze River Delta Lung Cancer Cooperation Group) has synthesized insights from China's innovative drug development landscape and clinical practice to formulate an expert consensus on the diagnosis and treatment of NSCLC patients with MET alterations. This consensus addresses key areas, such as optimal testing timing, testing methods, testing strategies, quality control measures, and treatment approaches. By offering standardized recommendations, this guidance aims to streamline diagnostic and therapeutic processes and enhance clinical decision-making for NSCLC with MET alterations.

{"title":"Expert consensus on the diagnosis and treatment of non-small cell lung cancer with MET alteration.","authors":"Huijing Feng, Yang Xia, Wenxian Wang, Chunwei Xu, Qian Wang, Zhengbo Song, Ziming Li, Jinpu Yu, Wenzhao Zhong, Zhijie Wang, Yongchang Zhang, Jingjing Liu, Shirong Zhang, Xiuyu Cai, Anwen Liu, Wen Li, Ping Zhan, Hongbing Liu, Tangfeng Lyu, Liyun Miao, Lingfeng Min, Gen Lin, Long Huang, Jingping Yuan, Zhansheng Jiang, Xingxiang Pu, Chuangzhou Rao, Dongqing Lyu, Zongyang Yu, Xiaoyan Li, Chuanhao Tang, Chengzhi Zhou, Qi Mei, Hui Guo, Qian Chu, Rui Meng, Xuewen Liu, Jingxun Wu, Jin Zhou, Zhengfei Zhu, Weiwei Pan, Fei Pang, Meizhen Hu, Kai Wang, Fan Wu, Bingwei Xu, Ling Xu, Liping Wang, Youcai Zhu, Jisheng Li, Yanru Xie, Xinqing Lin, Jing Cai, Lin Wang, Yingying Du, Wang Yao, Xuefei Shi, Xiaomin Niu, Dongmei Yuan, Yanwen Yao, Jing Kang, Jiatao Zhang, Chao Zhang, Wenbin Gao, Jianhui Huang, Yinbin Zhang, Pingli Sun, Hong Wang, Mingxiang Ye, Dong Wang, Zhaofeng Wang, Yue Hao, Zheng Wang, Bing Wan, Donglai Lyu, Xiaodong Jiao, Lin Shi, Gang Lan, Shengjie Yang, Yanhong Shang, Yina Wang, Bihui Li, Gang Jin, Kang Zheng, Jun Ma, Wenfeng Li, Zhang Zhang, Zhongwu Li, Yuan Li, Zhefeng Liu, Xuelei Ma, Nong Yang, Lin Wu, Qiming Wang, Guansong Wang, Zhuan Hong, Jiandong Wang, Meiyu Fang, Yong Fang, Xixu Zhu, Yi Shen, Ke Wang, Xiubao Ren, Yiping Zhang, Shenglin Ma, Junping Zhang, Yong Song, Wenfeng Fang, Yuanzhi Lu","doi":"10.20892/j.issn.2095-3941.2024.0503","DOIUrl":"https://doi.org/10.20892/j.issn.2095-3941.2024.0503","url":null,"abstract":"<p><p>Alterations in the mesenchymal-epithelial transition factor (<i>MET</i>) gene are critical drivers of non-small cell lung cancer (NSCLC). In recent years advances in precision therapies targeting MET alterations have significantly expanded treatment options for NSCLC patients. These alterations include <i>MET</i> exon 14 skipping mutations (<i>MET</i> exon 14 skipping), <i>MET</i> gene amplifications, <i>MET</i> point mutations (primarily kinase domain mutations), and MET protein overexpression. Accurate identification of these alterations and appropriate selection of patient populations and targeted therapies are essential for improving clinical outcomes. The East China Lung Cancer Group, Youth Committee (ECLUNG YOUNG, Yangtze River Delta Lung Cancer Cooperation Group) has synthesized insights from China's innovative drug development landscape and clinical practice to formulate an expert consensus on the diagnosis and treatment of NSCLC patients with MET alterations. This consensus addresses key areas, such as optimal testing timing, testing methods, testing strategies, quality control measures, and treatment approaches. By offering standardized recommendations, this guidance aims to streamline diagnostic and therapeutic processes and enhance clinical decision-making for NSCLC with MET alterations.</p>","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":"22 3","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143810561","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MDM1 overexpression promotes p53 expression and cell apoptosis to enhance therapeutic sensitivity to chemoradiotherapy in patients with colorectal cancer.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-04-08 DOI: 10.20892/j.issn.2095-3941.2024.0540
Ningxin Ren, Hongxia Chen, Ying Huang, Jing Jin, Shaosen Zhang, Ruoqing Yan, Mengjie Li, Linlin Zheng, Shuangmei Zou, Yexiong Li, Wen Tan, Dongxin Lin

Objective: Identifying biomarkers that predict the efficacy and prognosis of chemoradiotherapy is important for individualized clinical treatment. We previously reported that high murine double minute 1 (MDM1) expression in patients with rectal cancer is linked to a favorable chemoradiation response. In this study the role of MDM1 in the chemoradiotherapy response in colorectal cancer (CRC) patients was evaluated.

Methods: Colony formation and cell proliferation assays as well as xenograft models were used to determine if MDM1 expression affects the sensitivity of CRC cells to chemoradiation. RNA sequencing revealed that MDM1 regulates tumor protein 53 (TP53) expression and apoptosis. A series of molecular biology experiments were performed to determine how MDM1 affects p53 expression. The effects of inhibitors targeting apoptosis on MDM1 knockout cells were evaluated.

Results: Gene expression profiling revealed that MDM1 is a potential chemoradiotherapy sensitivity marker. The sensitivity of CRC cells to chemoradiation treatment decreased after MDM1 knockout and increased after MDM1 overexpression. MDM1 affected p53 expression, thereby regulating apoptosis. MDM1 overexpression limited YBX1 binding to TP53 promoter, regulated TP53 expression, and rendered CRC cells more sensitive to chemoradiation. In CRC cells with low MDM1 expression, a combination of apoptosis-inducing inhibitors and chemoradiation treatment restored sensitivity to cancer therapy.

Conclusions: The current study showed that MDM1 expression influences the sensitivity of CRC cells to chemoradiation by influencing p53 and apoptosis pathways, which is the basis for the underlying molecular mechanism, and serves as a possible predictive marker for chemoradiotherapy prognosis.

{"title":"MDM1 overexpression promotes p53 expression and cell apoptosis to enhance therapeutic sensitivity to chemoradiotherapy in patients with colorectal cancer.","authors":"Ningxin Ren, Hongxia Chen, Ying Huang, Jing Jin, Shaosen Zhang, Ruoqing Yan, Mengjie Li, Linlin Zheng, Shuangmei Zou, Yexiong Li, Wen Tan, Dongxin Lin","doi":"10.20892/j.issn.2095-3941.2024.0540","DOIUrl":"https://doi.org/10.20892/j.issn.2095-3941.2024.0540","url":null,"abstract":"<p><strong>Objective: </strong>Identifying biomarkers that predict the efficacy and prognosis of chemoradiotherapy is important for individualized clinical treatment. We previously reported that high murine double minute 1 (<i>MDM1</i>) expression in patients with rectal cancer is linked to a favorable chemoradiation response. In this study the role of MDM1 in the chemoradiotherapy response in colorectal cancer (CRC) patients was evaluated.</p><p><strong>Methods: </strong>Colony formation and cell proliferation assays as well as xenograft models were used to determine if MDM1 expression affects the sensitivity of CRC cells to chemoradiation. RNA sequencing revealed that MDM1 regulates tumor protein 53 (<i>TP53</i>) expression and apoptosis. A series of molecular biology experiments were performed to determine how MDM1 affects p53 expression. The effects of inhibitors targeting apoptosis on MDM1 knockout cells were evaluated.</p><p><strong>Results: </strong>Gene expression profiling revealed that MDM1 is a potential chemoradiotherapy sensitivity marker. The sensitivity of CRC cells to chemoradiation treatment decreased after MDM1 knockout and increased after MDM1 overexpression. MDM1 affected p53 expression, thereby regulating apoptosis. MDM1 overexpression limited YBX1 binding to <i>TP53</i> promoter, regulated <i>TP53</i> expression, and rendered CRC cells more sensitive to chemoradiation. In CRC cells with low MDM1 expression, a combination of apoptosis-inducing inhibitors and chemoradiation treatment restored sensitivity to cancer therapy.</p><p><strong>Conclusions: </strong>The current study showed that MDM1 expression influences the sensitivity of CRC cells to chemoradiation by influencing p53 and apoptosis pathways, which is the basis for the underlying molecular mechanism, and serves as a possible predictive marker for chemoradiotherapy prognosis.</p>","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":"22 3","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143810564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Application and future prospects of bispecific antibodies in the treatment of non-small cell lung cancer.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-04-07 DOI: 10.20892/j.issn.2095-3941.2024.0470
Junxu Wen, Wenxing Cui, Xiaoyan Yin, Yu Chen, Ailing Liu, Qian Wang, Xiangjiao Meng

As the leading cause of cancer-related deaths, lung cancer remains a noteworthy threat to human health. Although immunotherapies, such as immune checkpoint inhibitors (ICIs), have significantly increased the efficacy of lung cancer treatment, a significant percentage of patients are not sensitive to immunotherapies and patients who initially respond to treatment can quickly develop acquired drug resistance. Bispecific antibodies (bsAbs) bind two different antigens or epitopes simultaneously and have been shown to enhance antitumor efficacy with suitable safety profiles, thus attracting increasing attention as novel antitumor therapies. At present, in addition to the approved bsAb, amivantamab, three novel bsAbs (KN046, AK112, and SHR-1701) are being evaluated in phase 3 clinical trials and many bsAbs are being evaluated in phase 1/2 clinical trials for patients with non-small cell lung cancer (NSCLC). Herein we present the structure, classification, and mechanism of action underlying bsAbs in NSCLC and introduce related clinical trials. Finally, we discuss challenges, potential solutions, and future prospects in the context of cancer treatment with bsAbs.

{"title":"Application and future prospects of bispecific antibodies in the treatment of non-small cell lung cancer.","authors":"Junxu Wen, Wenxing Cui, Xiaoyan Yin, Yu Chen, Ailing Liu, Qian Wang, Xiangjiao Meng","doi":"10.20892/j.issn.2095-3941.2024.0470","DOIUrl":"https://doi.org/10.20892/j.issn.2095-3941.2024.0470","url":null,"abstract":"<p><p>As the leading cause of cancer-related deaths, lung cancer remains a noteworthy threat to human health. Although immunotherapies, such as immune checkpoint inhibitors (ICIs), have significantly increased the efficacy of lung cancer treatment, a significant percentage of patients are not sensitive to immunotherapies and patients who initially respond to treatment can quickly develop acquired drug resistance. Bispecific antibodies (bsAbs) bind two different antigens or epitopes simultaneously and have been shown to enhance antitumor efficacy with suitable safety profiles, thus attracting increasing attention as novel antitumor therapies. At present, in addition to the approved bsAb, amivantamab, three novel bsAbs (KN046, AK112, and SHR-1701) are being evaluated in phase 3 clinical trials and many bsAbs are being evaluated in phase 1/2 clinical trials for patients with non-small cell lung cancer (NSCLC). Herein we present the structure, classification, and mechanism of action underlying bsAbs in NSCLC and introduce related clinical trials. Finally, we discuss challenges, potential solutions, and future prospects in the context of cancer treatment with bsAbs.</p>","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":" ","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143794687","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IL-33/ST2 signalling promotes tumor growth by regulating polarization of alternatively activated macrophages.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-03-27 DOI: 10.20892/j.issn.2095-3941.2024.0483
Liping Liu, Haoge Luo, Yingdong Xie, Ying Wang, Shiying Ren, Haiyang Sun, Dong Li

Objective: Suppression of tumorigenicity 2 (ST2), the receptor for interleukin (IL)-33, has a critical role in tumor growth, angiogenesis, metastasis, and immune modulation. The IL-33/ST2 pathway is known to influence the polarization and function of macrophages, which is integral to modulating the tumor microenvironment. However, the precise role of IL-33/ST2 in tumors, particularly non-small cell lung cancer (NSCLC), has not been established.

Methods: ST2 expression in NSCLC was analysed using a murine model and patient specimens. The effect of the IL-33/ST2 axis on macrophage polarization in NSCLC was determined.

Results: Elevated ST2 expression was correlated with aggressive tumor growth. Specifically, ST2 expression on macrophages was associated with lung cancer progression and the absence of ST2 on macrophages was associated with diminished tumor growth. IL-33 promoted polarization of alternatively activated macrophages in an ST2-dependent manner that was mediated via the PI3K/Akt signalling pathway. Moreover, IL-33 inhibited T-cell function by inducing the secretion of transforming growth factor β from alternatively activated macrophages.

Conclusions: Macrophages expressing ST2 can serve as promising therapeutic targets for NSCLC immunotherapy, highlighting the IL-33/ST2 axis as a potential target for future antitumor strategies.

{"title":"IL-33/ST2 signalling promotes tumor growth by regulating polarization of alternatively activated macrophages.","authors":"Liping Liu, Haoge Luo, Yingdong Xie, Ying Wang, Shiying Ren, Haiyang Sun, Dong Li","doi":"10.20892/j.issn.2095-3941.2024.0483","DOIUrl":"https://doi.org/10.20892/j.issn.2095-3941.2024.0483","url":null,"abstract":"<p><strong>Objective: </strong>Suppression of tumorigenicity 2 (ST2), the receptor for interleukin (IL)-33, has a critical role in tumor growth, angiogenesis, metastasis, and immune modulation. The IL-33/ST2 pathway is known to influence the polarization and function of macrophages, which is integral to modulating the tumor microenvironment. However, the precise role of IL-33/ST2 in tumors, particularly non-small cell lung cancer (NSCLC), has not been established.</p><p><strong>Methods: </strong>ST2 expression in NSCLC was analysed using a murine model and patient specimens. The effect of the IL-33/ST2 axis on macrophage polarization in NSCLC was determined.</p><p><strong>Results: </strong>Elevated <i>ST2</i> expression was correlated with aggressive tumor growth. Specifically, <i>ST2</i> expression on macrophages was associated with lung cancer progression and the absence of <i>ST2</i> on macrophages was associated with diminished tumor growth. IL-33 promoted polarization of alternatively activated macrophages in an ST2-dependent manner that was mediated <i>via</i> the PI3K/Akt signalling pathway. Moreover, IL-33 inhibited T-cell function by inducing the secretion of transforming growth factor β from alternatively activated macrophages.</p><p><strong>Conclusions: </strong>Macrophages expressing ST2 can serve as promising therapeutic targets for NSCLC immunotherapy, highlighting the IL-33/ST2 axis as a potential target for future antitumor strategies.</p>","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":" ","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143717986","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Current status of multiple markers in precision immunotherapy for colorectal cancer.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-03-26 DOI: 10.20892/j.issn.2095-3941.2025.0030
Chao Liu, Ya Lan, Hong Wang, Yanqiao Zhang
{"title":"Current status of multiple markers in precision immunotherapy for colorectal cancer.","authors":"Chao Liu, Ya Lan, Hong Wang, Yanqiao Zhang","doi":"10.20892/j.issn.2095-3941.2025.0030","DOIUrl":"10.20892/j.issn.2095-3941.2025.0030","url":null,"abstract":"","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":" ","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143708668","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advances and challenges in gastric cancer testing: the role of biomarkers.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-03-24 DOI: 10.20892/j.issn.2095-3941.2024.0386
Yu Sun, Pavitratha Puspanathan, Tony Lim, Dongmei Lin

Advances in the identification of molecular biomarkers and the development of targeted therapies have enhanced the prognosis of patients with advanced gastric cancer. Several established biomarkers have been widely integrated into routine clinical diagnostics of gastric cancer to guide personalized treatment. Human epidermal growth factor receptor 2 (HER2) was the first molecular biomarker to be used in gastric cancer with trastuzumab being the first approved targeted therapy for HER2-positive gastric cancer. Programmed death-ligand 1 positivity and microsatellite instability can guide the use of immunotherapies, such as pembrolizumab and nivolumab. More recently, zolbetuximab has been approved for patients with claudin 18.2-positive diseases in some countries. More targeted therapies, including savolitinib for MET-positive patients, are currently under clinical investigation. However, the clinical application of these diagnostic approaches could be hampered by many existing challenges, including invasive and costly sampling methods, variability in immunohistochemistry interpretation, high costs and long turnaround times for next-generation sequencing, the absence of standardized and clinically validated diagnostic cut-off values for some biomarkers, and tumor heterogeneity. Novel testing and analysis techniques, such as artificial intelligence-assisted image analysis and multiplex immunohistochemistry, and emerging therapeutic strategies, including combination therapies that integrate immune checkpoint inhibitors with targeted therapies, offer potential solutions to some of these challenges. This article reviews recent progress in gastric cancer testing, outlines current challenges, and explores future directions for biomarker testing and targeted therapy for gastric cancer.

{"title":"Advances and challenges in gastric cancer testing: the role of biomarkers.","authors":"Yu Sun, Pavitratha Puspanathan, Tony Lim, Dongmei Lin","doi":"10.20892/j.issn.2095-3941.2024.0386","DOIUrl":"10.20892/j.issn.2095-3941.2024.0386","url":null,"abstract":"<p><p>Advances in the identification of molecular biomarkers and the development of targeted therapies have enhanced the prognosis of patients with advanced gastric cancer. Several established biomarkers have been widely integrated into routine clinical diagnostics of gastric cancer to guide personalized treatment. Human epidermal growth factor receptor 2 (HER2) was the first molecular biomarker to be used in gastric cancer with trastuzumab being the first approved targeted therapy for HER2-positive gastric cancer. Programmed death-ligand 1 positivity and microsatellite instability can guide the use of immunotherapies, such as pembrolizumab and nivolumab. More recently, zolbetuximab has been approved for patients with claudin 18.2-positive diseases in some countries. More targeted therapies, including savolitinib for MET-positive patients, are currently under clinical investigation. However, the clinical application of these diagnostic approaches could be hampered by many existing challenges, including invasive and costly sampling methods, variability in immunohistochemistry interpretation, high costs and long turnaround times for next-generation sequencing, the absence of standardized and clinically validated diagnostic cut-off values for some biomarkers, and tumor heterogeneity. Novel testing and analysis techniques, such as artificial intelligence-assisted image analysis and multiplex immunohistochemistry, and emerging therapeutic strategies, including combination therapies that integrate immune checkpoint inhibitors with targeted therapies, offer potential solutions to some of these challenges. This article reviews recent progress in gastric cancer testing, outlines current challenges, and explores future directions for biomarker testing and targeted therapy for gastric cancer.</p>","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":" ","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143691236","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DB-1310, a HER3-targeting antibody-drug conjugate, has synergistic anti-tumor activity with trastuzumab in HER2- and HER3-expressing breast cancer.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-03-19 DOI: 10.20892/j.issn.2095-3941.2024.0586
Xi Li, Liwen Liang, Zhongyuan Zhu, Haiqing Hua, Yang Qiu
{"title":"DB-1310, a HER3-targeting antibody-drug conjugate, has synergistic anti-tumor activity with trastuzumab in HER2- and HER3-expressing breast cancer.","authors":"Xi Li, Liwen Liang, Zhongyuan Zhu, Haiqing Hua, Yang Qiu","doi":"10.20892/j.issn.2095-3941.2024.0586","DOIUrl":"10.20892/j.issn.2095-3941.2024.0586","url":null,"abstract":"","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":" ","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143662594","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Insights into the DNA damage response and tumor drug resistance.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-03-18 DOI: 10.20892/j.issn.2095-3941.2025.0020
Xiaolu Ma, Zina Cheng, Caixia Guo
{"title":"Insights into the DNA damage response and tumor drug resistance.","authors":"Xiaolu Ma, Zina Cheng, Caixia Guo","doi":"10.20892/j.issn.2095-3941.2025.0020","DOIUrl":"10.20892/j.issn.2095-3941.2025.0020","url":null,"abstract":"","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":" ","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-03-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143656158","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Continuous R-DA-EDOCH alternated with high-dose Ara-C induces deep remission and overcomes high-risk factors in young patients with newly diagnosed mantle cell lymphoma.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-03-12 DOI: 10.20892/j.issn.2095-3941.2024.0200
Yi Wang, Yuting Yan, Dandan Shan, Jiawen Chen, Wei Liu, Tingyu Wang, Gang An, Weiwei Sui, Wenyang Huang, Wenjie Xiong, Huimin Liu, Qi Sun, Huijun Wang, Zhijian Xiao, Jianxiang Wang, Lugui Qiu, Dehui Zou, Shuhua Yi

Objective: Our previous studies have indicated potentially higher proliferative activity of tumor cells in Chinese patients with mantle-cell lymphoma (MCL) than those in Western. Given the success and tolerability of R-DA-EDOCH immunochemotherapy in treating aggressive B-cell lymphomas, we designed a prospective, phase 3 trial to explore the efficacy and safety of alternating R-DA-EDOCH/R-DHAP induction therapy for young patients with newly diagnosed MCL. The primary endpoint was the complete remission rate (CRR) at the end of induction (EOI).

Methods: A total of 55 patients were enrolled. The CRR at the EOI was 89.1% [95% confidence interval (CI) 78%-96%], and the overall response rate was 98.1% (95% CI 90%-100%). Most patients with bone marrow involvement quickly attained minimal residual disease (MRD) negative status, with a 95.7% rate at the EOI.

Results: The 3-year progression-free survival (PFS) and overall survival rates were 66.3% and 83.2%, respectively. No patients discontinued treatment because of adverse events. Univariate analysis identified pathologic morphology and TP53 mutations as risk factors for PFS. However, high tumor proliferative activity and certain cytogenetic abnormalities showed no significant adverse prognostic significance.

Conclusions: Intensive therapy based on a high cytarabine dose and continuously administered EDOCH achieved a high MRD-negative rate and provides an optional induction choice for young patients with MCL with high-risk factors.

{"title":"Continuous R-DA-EDOCH alternated with high-dose Ara-C induces deep remission and overcomes high-risk factors in young patients with newly diagnosed mantle cell lymphoma.","authors":"Yi Wang, Yuting Yan, Dandan Shan, Jiawen Chen, Wei Liu, Tingyu Wang, Gang An, Weiwei Sui, Wenyang Huang, Wenjie Xiong, Huimin Liu, Qi Sun, Huijun Wang, Zhijian Xiao, Jianxiang Wang, Lugui Qiu, Dehui Zou, Shuhua Yi","doi":"10.20892/j.issn.2095-3941.2024.0200","DOIUrl":"10.20892/j.issn.2095-3941.2024.0200","url":null,"abstract":"<p><strong>Objective: </strong>Our previous studies have indicated potentially higher proliferative activity of tumor cells in Chinese patients with mantle-cell lymphoma (MCL) than those in Western. Given the success and tolerability of R-DA-EDOCH immunochemotherapy in treating aggressive B-cell lymphomas, we designed a prospective, phase 3 trial to explore the efficacy and safety of alternating R-DA-EDOCH/R-DHAP induction therapy for young patients with newly diagnosed MCL. The primary endpoint was the complete remission rate (CRR) at the end of induction (EOI).</p><p><strong>Methods: </strong>A total of 55 patients were enrolled. The CRR at the EOI was 89.1% [95% confidence interval (CI) 78%-96%], and the overall response rate was 98.1% (95% CI 90%-100%). Most patients with bone marrow involvement quickly attained minimal residual disease (MRD) negative status, with a 95.7% rate at the EOI.</p><p><strong>Results: </strong>The 3-year progression-free survival (PFS) and overall survival rates were 66.3% and 83.2%, respectively. No patients discontinued treatment because of adverse events. Univariate analysis identified pathologic morphology and TP53 mutations as risk factors for PFS. However, high tumor proliferative activity and certain cytogenetic abnormalities showed no significant adverse prognostic significance.</p><p><strong>Conclusions: </strong>Intensive therapy based on a high cytarabine dose and continuously administered EDOCH achieved a high MRD-negative rate and provides an optional induction choice for young patients with MCL with high-risk factors.</p>","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":"22 2","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-03-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11899595/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143603978","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Advances in gut microbiota-related treatment strategies for managing colorectal cancer in humans.
IF 5.6 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2025-03-12 DOI: 10.20892/j.issn.2095-3941.2024.0263
Bhaskar Roy, Kunfeng Cao, Chabungbam Orville Singh, Xiaodong Fang, Huanming Yang, Dong Wei

Colorectal cancer (CRC) is a major contributor to global cancer-related mortality with increasing incidence rates in both developed and developing regions. Therefore, CRC presents a significant challenge to global health. The development of innovative tools for enhancing early CRC screening and diagnosis, along with novel treatments and therapies for improved management, remains an urgent necessity. CRC is intricately associated with the gut microbiota, which is integral to food digestion, nutrient generation, drug metabolism, metabolite production, immune enhancement, endocrine regulation, neurogenesis modulation, and the maintenance of physiologic and psychological equilibrium. Dysbiosis or imbalances in the gut microbiome have been implicated in various disorders, including CRC. Emerging evidence highlights the critical role of the gut microbiome in CRC pathogenesis and treatment, which presents potential opportunities for early detection and diagnosis. Despite substantial advances in understanding the relationship between the gut microbiota and CRC, significant challenges persist. Gaining a deeper and more detailed understanding of the interactions between the human microbiota and cancer is essential to fully realize the potential of the microbiota in cancer management. Unlike genetic factors, the gut microbiome is subject to modification, offering a promising avenue for the development of CRC treatments and drug discovery. This review provides an overview of the interactions between the human gut microbiome and CRC, while examining prospects for precision management of CRC.

{"title":"Advances in gut microbiota-related treatment strategies for managing colorectal cancer in humans.","authors":"Bhaskar Roy, Kunfeng Cao, Chabungbam Orville Singh, Xiaodong Fang, Huanming Yang, Dong Wei","doi":"10.20892/j.issn.2095-3941.2024.0263","DOIUrl":"10.20892/j.issn.2095-3941.2024.0263","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is a major contributor to global cancer-related mortality with increasing incidence rates in both developed and developing regions. Therefore, CRC presents a significant challenge to global health. The development of innovative tools for enhancing early CRC screening and diagnosis, along with novel treatments and therapies for improved management, remains an urgent necessity. CRC is intricately associated with the gut microbiota, which is integral to food digestion, nutrient generation, drug metabolism, metabolite production, immune enhancement, endocrine regulation, neurogenesis modulation, and the maintenance of physiologic and psychological equilibrium. Dysbiosis or imbalances in the gut microbiome have been implicated in various disorders, including CRC. Emerging evidence highlights the critical role of the gut microbiome in CRC pathogenesis and treatment, which presents potential opportunities for early detection and diagnosis. Despite substantial advances in understanding the relationship between the gut microbiota and CRC, significant challenges persist. Gaining a deeper and more detailed understanding of the interactions between the human microbiota and cancer is essential to fully realize the potential of the microbiota in cancer management. Unlike genetic factors, the gut microbiome is subject to modification, offering a promising avenue for the development of CRC treatments and drug discovery. This review provides an overview of the interactions between the human gut microbiome and CRC, while examining prospects for precision management of CRC.</p>","PeriodicalId":9611,"journal":{"name":"Cancer Biology & Medicine","volume":"22 2","pages":""},"PeriodicalIF":5.6,"publicationDate":"2025-03-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11899591/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143604021","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer Biology & Medicine
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