Yuting Liu, Shing Chun Tang, Chi Han Li, Ka Fai To, Bo Li, Stephen Lam Chan, Chi Hin Wong, Yangchao Chen
KRAS gene mutations are common in pancreatic ductal adenocarcinoma (PDAC), but targeting mutant KRAS is still challenging. Here, an endoribonuclease-prepared small interfering RNA (esiRNA) library was used to screen new kinases that play critical roles in PDAC driven by KRAS gene mutations, and serine/threonine kinase 31 (STK31) was identified and characterized as a potential therapeutic target for KRAS-mutant PDAC. Our results showed that STK31 was upregulated in KRAS-mutant PDAC patients with poor survival and highly expressed in PDAC cell lines with KRASG12D mutation. Inhibition of STK31 in KRAS-mutant cell lines significantly reduced PDAC cell growth in vitro and hindered tumor growth in vivo. Gain and loss of function experiments revealed that STK31 is a downstream target of KRAS in PDAC. A pharmacological inhibition assay showed MAPK/ERK signaling involved in STK31 regulation. The further mechanistic study validated that c-Jun, regulated by KRAS/MAPK signaling, directly modulates the transcription level of STK31 by binding to its promoter region. Through RNA sequencing, we found that the cell cycle regulators CCNB1 and CDC25C are downstream targets of STK31. Taken together, our results indicate that STK31, which is the downstream target of the KRAS/MAPK/ERK/c-Jun signaling pathway in KRAS-mutant PDAC, promotes PDAC cell growth by modulating the expression of the cell cycle regulators CCNB1 and CDC25C.
{"title":"The molecular mechanism underlying KRAS regulation on STK31 expression in pancreatic ductal adenocarcinoma","authors":"Yuting Liu, Shing Chun Tang, Chi Han Li, Ka Fai To, Bo Li, Stephen Lam Chan, Chi Hin Wong, Yangchao Chen","doi":"10.1111/cas.16286","DOIUrl":"10.1111/cas.16286","url":null,"abstract":"<p>KRAS gene mutations are common in pancreatic ductal adenocarcinoma (PDAC), but targeting mutant KRAS is still challenging. Here, an endoribonuclease-prepared small interfering RNA (esiRNA) library was used to screen new kinases that play critical roles in PDAC driven by KRAS gene mutations, and serine/threonine kinase 31 (STK31) was identified and characterized as a potential therapeutic target for KRAS-mutant PDAC. Our results showed that STK31 was upregulated in KRAS-mutant PDAC patients with poor survival and highly expressed in PDAC cell lines with KRAS<sup>G12D</sup> mutation. Inhibition of STK31 in KRAS-mutant cell lines significantly reduced PDAC cell growth in vitro and hindered tumor growth in vivo. Gain and loss of function experiments revealed that STK31 is a downstream target of KRAS in PDAC. A pharmacological inhibition assay showed MAPK/ERK signaling involved in STK31 regulation. The further mechanistic study validated that c-Jun, regulated by KRAS/MAPK signaling, directly modulates the transcription level of STK31 by binding to its promoter region. Through RNA sequencing, we found that the cell cycle regulators CCNB1 and CDC25C are downstream targets of STK31. Taken together, our results indicate that STK31, which is the downstream target of the KRAS/MAPK/ERK/c-Jun signaling pathway in KRAS-mutant PDAC, promotes PDAC cell growth by modulating the expression of the cell cycle regulators CCNB1 and CDC25C.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3288-3304"},"PeriodicalIF":4.5,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447899/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141761975","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}
Yan Kuang, Hong Xu, Fangfang Lu, Jiahua Meng, Yeye Yi, Huilan Yang, Hairui Hou, Hao Wei, Shanheng Su. Inhibition of microRNA let-7b expression by KDM2B promotes cancer progression by targeting EZH2 in ovarian cancer. Cancer Science. 2021;112:231–242.
We apologize for these errors.
匡艳 徐红 卢芳芳 孟佳华 易烨 杨慧兰 侯海瑞 魏浩 苏善恒KDM2B靶向EZH2抑制卵巢癌microRNA let-7b表达促进癌症进展癌症科学2021;112:231-242.We apologize for these errors.
{"title":"Correction to inhibition of microRNA let-7b expression by KDM2B promotes cancer progression by targeting EZH2 in ovarian cancer","authors":"","doi":"10.1111/cas.16291","DOIUrl":"10.1111/cas.16291","url":null,"abstract":"<p>Yan Kuang, Hong Xu, Fangfang Lu, Jiahua Meng, Yeye Yi, Huilan Yang, Hairui Hou, Hao Wei, Shanheng Su. Inhibition of microRNA let-7b expression by KDM2B promotes cancer progression by targeting EZH2 in ovarian cancer. Cancer Science. 2021;112:231–242.</p><p>We apologize for these errors.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3492-3494"},"PeriodicalIF":4.5,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447914/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141753076","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}
Cancer cells are generally exposed to numerous extrinsic stimulations in the tumor microenvironment. In this environment, cancer cells change their expression profiles to fight against circumstantial stresses, allowing their progression in the challenging tissue space. Technological advancements of spatial omics have had substantial influence on cancer genomics. This technical progress, especially that occurring in the spatial transcriptome, has been drastic and rapid. Here, we describe the latest spatial analytical technologies that have allowed omics feature characterization to retain their spatial and histopathological information in cancer tissues. Several spatial omics platforms have been launched, and the latest platforms finally attained single-cell level or even higher subcellular level resolution. We discuss several key papers elucidating the initial utility of the spatial analysis. In fact, spatial transcriptome analyses reveal comprehensive omics characteristics not only in cancer cells but also their surrounding cells, such as tumor infiltrating immune cells and cancer-associated fibroblasts. We also introduce several spatial omics platforms. We describe our own attempts to investigate molecular events associated with cancer progression. Furthermore, we discuss the next challenges in analyzing the multiomics status of cells, including their morphology and location. These novel technologies, in conjunction with spatial transcriptome analysis and, more importantly, with histopathology, will elucidate even novel key aspects of the intratumor heterogeneity of cancers. Such enhanced knowledge is expected to open a new path for overcoming therapeutic resistance and eventually to precisely stratify patients.
{"title":"Spatial omics technologies for understanding molecular status associated with cancer progression","authors":"Satoi Nagasawa, Junko Zenkoh, Yutaka Suzuki, Ayako Suzuki","doi":"10.1111/cas.16283","DOIUrl":"10.1111/cas.16283","url":null,"abstract":"<p>Cancer cells are generally exposed to numerous extrinsic stimulations in the tumor microenvironment. In this environment, cancer cells change their expression profiles to fight against circumstantial stresses, allowing their progression in the challenging tissue space. Technological advancements of spatial omics have had substantial influence on cancer genomics. This technical progress, especially that occurring in the spatial transcriptome, has been drastic and rapid. Here, we describe the latest spatial analytical technologies that have allowed omics feature characterization to retain their spatial and histopathological information in cancer tissues. Several spatial omics platforms have been launched, and the latest platforms finally attained single-cell level or even higher subcellular level resolution. We discuss several key papers elucidating the initial utility of the spatial analysis. In fact, spatial transcriptome analyses reveal comprehensive omics characteristics not only in cancer cells but also their surrounding cells, such as tumor infiltrating immune cells and cancer-associated fibroblasts. We also introduce several spatial omics platforms. We describe our own attempts to investigate molecular events associated with cancer progression. Furthermore, we discuss the next challenges in analyzing the multiomics status of cells, including their morphology and location. These novel technologies, in conjunction with spatial transcriptome analysis and, more importantly, with histopathology, will elucidate even novel key aspects of the intratumor heterogeneity of cancers. Such enhanced knowledge is expected to open a new path for overcoming therapeutic resistance and eventually to precisely stratify patients.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3208-3217"},"PeriodicalIF":4.5,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447966/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141753077","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}
This single-arm confirmatory study (JCOG1305) aimed to evaluate the utility of interim positron emission tomography (iPET)-guided therapy for newly diagnosed advanced-stage classic Hodgkin lymphoma (cHL). Patients aged 16–60 years with cHL received two cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) and then underwent an iPET scan (PET2), which was centrally reviewed using a five-point Deauville scale. PET2-negative patients continued an additional four cycles of ABVD, whereas PET2-positive patients switched to six cycles of escalated bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone (eBEACOPP). The co-primary endpoints were 2-year progression-free survival (PFS) among all eligible and PET2-positive patients. Ninety-three patients were enrolled between January 2016 and December 2019. One patient was ineligible because of a diagnostic error. The median age of the 92 eligible patients was 35 (interquartile range, 28–48) years. Forty (43%) patients had stage III disease, and 43 (47%) had stage IV disease. The remaining nine (10%) patients had stage IIB disease with risk factors. Nineteen PET2-positive (21%) patients received eBEACOPP, 18 completed six cycles of eBEACOPP, 73 PET2-negative (79%) patients continued ABVD, and 70 completed an additional four cycles of ABVD. With a median follow-up period of 41.1 months, the 2-year PFS of 92 eligible patients and 19 PET2-positive patients were 84.8% (80% confidence interval [CI], 79.2–88.9) and 84.2% (80% CI, 69.7–92.1), respectively. Both primary endpoints were met at the prespecified threshold. This study demonstrates that iPET-guided therapy is a useful treatment option for younger patients with newly diagnosed advanced-stage cHL. Registration number: jRCTs031180218.
{"title":"Interim PET-guided ABVD or ABVD/escalated BEACOPP for newly diagnosed advanced-stage classic Hodgkin lymphoma (JCOG1305)","authors":"Shigeru Kusumoto, Wataru Munakata, Ryunosuke Machida, Takashi Terauchi, Hiroaki Onaya, Masahiko Oguchi, Shinsuke Iida, Kisato Nosaka, Yasuhiro Suzuki, Yasuhiko Harada, Kana Miyazaki, Masaki Maruta, Noriko Fukuhara, Tomomi Toubai, Nobuko Kubota, Ken Ohmachi, Toko Saito, Shinya Rai, Ishikazu Mizuno, Suguru Fukuhara, Mai Takeuchi, Ukihide Tateishi, Dai Maruyama, Kunihiro Tsukasaki, Hirokazu Nagai","doi":"10.1111/cas.16281","DOIUrl":"10.1111/cas.16281","url":null,"abstract":"<p>This single-arm confirmatory study (JCOG1305) aimed to evaluate the utility of interim positron emission tomography (iPET)-guided therapy for newly diagnosed advanced-stage classic Hodgkin lymphoma (cHL). Patients aged 16–60 years with cHL received two cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) and then underwent an iPET scan (PET2), which was centrally reviewed using a five-point Deauville scale. PET2-negative patients continued an additional four cycles of ABVD, whereas PET2-positive patients switched to six cycles of escalated bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone (eBEACOPP). The co-primary endpoints were 2-year progression-free survival (PFS) among all eligible and PET2-positive patients. Ninety-three patients were enrolled between January 2016 and December 2019. One patient was ineligible because of a diagnostic error. The median age of the 92 eligible patients was 35 (interquartile range, 28–48) years. Forty (43%) patients had stage III disease, and 43 (47%) had stage IV disease. The remaining nine (10%) patients had stage IIB disease with risk factors. Nineteen PET2-positive (21%) patients received eBEACOPP, 18 completed six cycles of eBEACOPP, 73 PET2-negative (79%) patients continued ABVD, and 70 completed an additional four cycles of ABVD. With a median follow-up period of 41.1 months, the 2-year PFS of 92 eligible patients and 19 PET2-positive patients were 84.8% (80% confidence interval [CI], 79.2–88.9) and 84.2% (80% CI, 69.7–92.1), respectively. Both primary endpoints were met at the prespecified threshold. This study demonstrates that iPET-guided therapy is a useful treatment option for younger patients with newly diagnosed advanced-stage cHL. Registration number: jRCTs031180218.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3384-3393"},"PeriodicalIF":4.5,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447878/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141735375","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}
Satoko Ugai, Qian Yao, Yasutoshi Takashima, Yuxue Zhong, Kosuke Matsuda, Hidetaka Kawamura, Yu Imamura, Kazuo Okadome, Kosuke Mima, Kota Arima, Keisuke Kosumi, Mingyang Song, Jeffrey A. Meyerhardt, Marios Giannakis, Jonathan A. Nowak, Tomotaka Ugai, Shuji Ogino
Evidence indicates that combinations of anti-EGFR antibodies and KRAS p.G12C (c.34G>T) inhibitors can be an effective treatment strategy for advanced colorectal cancer. We hypothesized that KRAS c.34G>T (p.G12C)-mutated colorectal carcinoma might be a distinct tumor subtype. We utilized a prospective cohort incident tumor biobank (including 1347 colorectal carcinomas) and detected KRAS c.34G>T (p.G12C) mutation in 43 cases (3.2%) and other KRAS mutations (in codon 12, 13, 61, or 146) in 467 cases (35%). The CpG island methylator phenotype (CIMP)-low prevalence was similarly higher in KRAS c.34G>T mutants (52%) and other KRAS mutants (49%) than in KRAS-wild-type tumors (31%). KRAS c.34G>T mutants showed higher CIMP-high prevalence (14%) and lower CIMP-negative prevalence (33%) compared with other KRAS mutants (6% and 45%, respectively; p = 0.0036). Similar to other KRAS mutants, KRAS c.34G>T-mutated tumors were associated with cecal location, non-microsatellite instability (MSI)-high status, BRAF wild type, and PIK3CA mutation when compared with KRAS-wild-type tumors. Compared with BRAF-mutated tumors, KRAS c.34G>T mutants showed more frequent LINE-1 hypomethylation, a biomarker for early-onset colorectal carcinoma. KRAS c.34G>T mutants were not associated with other features, including the tumor tissue abundance of Fusobacterium nucleatum (F. animalis), pks+Escherichia coli, Bifidobacterium, or (enterotoxigenic) Bacteroides fragilis. Among 1122 BRAF-wild-type colorectal carcinomas, compared with KRAS-wild-type tumors, multivariable-adjusted colorectal cancer-specific mortality hazard ratios (95% confidence interval) were 1.82 (1.05–3.17) in KRAS c.34G>T (p.G12C)-mutated tumors (p = 0.035) and 1.57 (1.22–2.02) in other KRAS-mutated tumors (p = 0.0004). Our study provides novel evidence for clinical and tumor characteristics of KRAS c.34G>T (p.G12C)-mutated colorectal carcinoma.
{"title":"Clinicopathological, molecular, and prognostic features of colorectal carcinomas with KRAS c.34G>T (p.G12C) mutation","authors":"Satoko Ugai, Qian Yao, Yasutoshi Takashima, Yuxue Zhong, Kosuke Matsuda, Hidetaka Kawamura, Yu Imamura, Kazuo Okadome, Kosuke Mima, Kota Arima, Keisuke Kosumi, Mingyang Song, Jeffrey A. Meyerhardt, Marios Giannakis, Jonathan A. Nowak, Tomotaka Ugai, Shuji Ogino","doi":"10.1111/cas.16262","DOIUrl":"10.1111/cas.16262","url":null,"abstract":"<p>Evidence indicates that combinations of anti-EGFR antibodies and KRAS p.G12C (c.34G>T) inhibitors can be an effective treatment strategy for advanced colorectal cancer. We hypothesized that <i>KRAS</i> c.34G>T (p.G12C)-mutated colorectal carcinoma might be a distinct tumor subtype. We utilized a prospective cohort incident tumor biobank (including 1347 colorectal carcinomas) and detected <i>KRAS</i> c.34G>T (p.G12C) mutation in 43 cases (3.2%) and other <i>KRAS</i> mutations (in codon 12, 13, 61, or 146) in 467 cases (35%). The CpG island methylator phenotype (CIMP)-low prevalence was similarly higher in <i>KRAS</i> c.34G>T mutants (52%) and other <i>KRAS</i> mutants (49%) than in <i>KRAS</i>-wild-type tumors (31%). <i>KRAS</i> c.34G>T mutants showed higher CIMP-high prevalence (14%) and lower CIMP-negative prevalence (33%) compared with other <i>KRAS</i> mutants (6% and 45%, respectively; <i>p</i> = 0.0036). Similar to other <i>KRAS</i> mutants, <i>KRAS</i> c.34G>T-mutated tumors were associated with cecal location, non-microsatellite instability (MSI)-high status, <i>BRAF</i> wild type, and <i>PIK3CA</i> mutation when compared with <i>KRAS</i>-wild-type tumors. Compared with <i>BRAF</i>-mutated tumors, <i>KRAS</i> c.34G>T mutants showed more frequent LINE-1 hypomethylation, a biomarker for early-onset colorectal carcinoma. <i>KRAS</i> c.34G>T mutants were not associated with other features, including the tumor tissue abundance of <i>Fusobacterium nucleatum</i> (<i>F. animalis</i>), <i>pks</i><sup>+</sup> <i>Escherichia coli</i>, <i>Bifidobacterium</i>, or (enterotoxigenic) <i>Bacteroides fragilis</i>. Among 1122 <i>BRAF</i>-wild-type colorectal carcinomas, compared with <i>KRAS</i>-wild-type tumors, multivariable-adjusted colorectal cancer-specific mortality hazard ratios (95% confidence interval) were 1.82 (1.05–3.17) in <i>KRAS</i> c.34G>T (p.G12C)-mutated tumors (<i>p</i> = 0.035) and 1.57 (1.22–2.02) in other <i>KRAS</i>-mutated tumors (<i>p</i> = 0.0004). Our study provides novel evidence for clinical and tumor characteristics of <i>KRAS</i> c.34G>T (p.G12C)-mutated colorectal carcinoma.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3455-3465"},"PeriodicalIF":4.5,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448363/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141749394","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}
The molecular mechanisms driving the development of cervical adenocarcinoma (CADC) and optimal patient management strategies remain elusive. In this study, we have identified circMAN1A2_009 as an oncogenic circular RNA (circRNA) in CADC. Clinically, circMAN1A2_009 showed significant upregulation in CADC tissues, with an impressive area under the curve value of 0.8075 for detecting CADC. Functional studies, involving both gain-of-function and loss-of-function experiments, revealed that circMAN1A2_009 suppressed reactive oxygen species accumulation and apoptosis, and boosted cell viability in CADC cells. Conversely, silencing circMAN1A2_009 reversed these effects. Further mechanistic investigations indicated that circMAN1A2_009 interacted with YBX1, facilitating the phosphorylation levels of YBX1 at serine 102 (p-YBX1S102) and facilitating YBX1 nuclear localization through sequence 245–251. This interaction subsequently increased the activity of the glyoxalase 1 (GLO1) promoter, leading to the activation of GLO1 expression. Consistently, inhibition of either YBX1 or GLO1 mirrored the biological effects of circMAN1A2_009 in CADC cells. Additionally, knockdown of YBX1 or GLO1 partially reversed the oncogenic behaviors induced by circMAN1A2_009. In conclusion, our findings propose circMAN1A2_009 as a potential oncogene and a promising indicator for diagnosing and guiding therapy in CADC patients.
{"title":"CircMAN1A2_009 facilitates YBX1 nuclear localization to induce GLO1 activation for cervical adenocarcinoma cell growth","authors":"Yongjie Huang, Xinyi Wei, Mengyan Tu, Weiguo Lu, Junfen Xu","doi":"10.1111/cas.16264","DOIUrl":"10.1111/cas.16264","url":null,"abstract":"<p>The molecular mechanisms driving the development of cervical adenocarcinoma (CADC) and optimal patient management strategies remain elusive. In this study, we have identified circMAN1A2_009 as an oncogenic circular RNA (circRNA) in CADC. Clinically, circMAN1A2_009 showed significant upregulation in CADC tissues, with an impressive area under the curve value of 0.8075 for detecting CADC. Functional studies, involving both gain-of-function and loss-of-function experiments, revealed that circMAN1A2_009 suppressed reactive oxygen species accumulation and apoptosis, and boosted cell viability in CADC cells. Conversely, silencing circMAN1A2_009 reversed these effects. Further mechanistic investigations indicated that circMAN1A2_009 interacted with YBX1, facilitating the phosphorylation levels of YBX1 at serine 102 (p-YBX1<sup>S102</sup>) and facilitating YBX1 nuclear localization through sequence 245–251. This interaction subsequently increased the activity of the glyoxalase 1 (GLO1) promoter, leading to the activation of GLO1 expression. Consistently, inhibition of either YBX1 or GLO1 mirrored the biological effects of circMAN1A2_009 in CADC cells. Additionally, knockdown of YBX1 or GLO1 partially reversed the oncogenic behaviors induced by circMAN1A2_009. In conclusion, our findings propose circMAN1A2_009 as a potential oncogene and a promising indicator for diagnosing and guiding therapy in CADC patients.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3273-3287"},"PeriodicalIF":4.5,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447891/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141749393","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}
Lazertinib, a novel third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), demonstrates marked efficacy in EGFR-mutant lung cancer. However, resistance commonly develops, prompting consideration of therapeutic strategies to overcome initial drug resistance mechanisms. This study aimed to elucidate the adaptive resistance to lazertinib and advocate novel combination treatments that demonstrate efficacy in preventing resistance as a first-line treatment for EGFR mutation-positive NSCLC. We found that AXL knockdown significantly inhibited lung cancer cell viability in the presence of lazertinib, indicating that AXL activation contributes to lazertinib resistance. However, long-term culture with a combination of lazertinib and AXL inhibitors led to residual cell proliferation and increased the MCL-1 expression level, which was mediated by the nuclear translocation of the transcription factor YAP. Triple therapy with an MCL-1 or YAP inhibitor in combination with lazertinib and an AXL inhibitor significantly reduced cell viability and increased the apoptosis rate. These results demonstrate that AXL and YAP/MCL-1 signals contribute to adaptive lazertinib resistance in EGFR-mutant lung cancer cells, suggesting that the initial dual inhibition of AXL and YAP/MCL-1 might be a highly effective strategy in eliminating lazertinib-resistant cells.
{"title":"Initial AXL and MCL-1 inhibition contributes to abolishing lazertinib tolerance in EGFR-mutant lung cancer cells","authors":"Yohei Matsui, Tadaaki Yamada, Yuki Katayama, Soichi Hirai, Ryo Sawada, Yusuke Tachibana, Masaki Ishida, Hayato Kawachi, Ryota Nakamura, Naoya Nishioka, Kenji Morimoto, Masahiro Iwasaku, Mano Horinaka, Toshiyuki Sakai, Shinsaku Tokuda, Koichi Takayama","doi":"10.1111/cas.16292","DOIUrl":"10.1111/cas.16292","url":null,"abstract":"<p>Lazertinib, a novel third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), demonstrates marked efficacy in EGFR-mutant lung cancer. However, resistance commonly develops, prompting consideration of therapeutic strategies to overcome initial drug resistance mechanisms. This study aimed to elucidate the adaptive resistance to lazertinib and advocate novel combination treatments that demonstrate efficacy in preventing resistance as a first-line treatment for EGFR mutation-positive NSCLC. We found that AXL knockdown significantly inhibited lung cancer cell viability in the presence of lazertinib, indicating that AXL activation contributes to lazertinib resistance. However, long-term culture with a combination of lazertinib and AXL inhibitors led to residual cell proliferation and increased the MCL-1 expression level, which was mediated by the nuclear translocation of the transcription factor YAP. Triple therapy with an MCL-1 or YAP inhibitor in combination with lazertinib and an AXL inhibitor significantly reduced cell viability and increased the apoptosis rate. These results demonstrate that AXL and YAP/MCL-1 signals contribute to adaptive lazertinib resistance in EGFR-mutant lung cancer cells, suggesting that the initial dual inhibition of AXL and YAP/MCL-1 might be a highly effective strategy in eliminating lazertinib-resistant cells.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3333-3345"},"PeriodicalIF":4.5,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447890/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141749395","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}
Jian Qi, Bo Hong, Shujie Wang, Jingyun Wang, Jinman Fang, Ruifang Sun, Jinfu Nie, Hongzhi Wang
Early detection plays a critical role in mitigating mortality rates linked to gastric cancer. However, current clinical screening methods exhibit suboptimal efficacy. Methylation alterations identified from cell-free DNA (cfDNA) present a promising biomarker for early cancer detection. Our study focused on identifying gastric cancer-specific markers from cfDNA methylation to facilitate early detection. We enrolled 150 gastric cancer patients and 100 healthy controls in this study, and undertook genome-wide methylation profiling of cfDNA using cell-free methylated DNA immunoprecipitation and high-throughput sequencing. We identified 21 differentially methylated regions (DMRs) between the gastric tumor and nontumor groups using multiple algorithms. Subsequently, using the 21 DMRs, we developed a gastric cancer detection model by random forest algorithm in the discovery set, and validated the model in an independent set. The model was able to accurately discriminate gastric cancer with a sensitivity and specificity of 93.90% and 95.15% in the discovery set, respectively, and 88.38% and 94.23% in the validation set, respectively. These results underscore the efficacy and accuracy of cfDNA-derived methylation markers in distinguishing early stage gastric cancer. This study highlighted the significance of cfDNA methylation alterations in early gastric cancer detection.
{"title":"Plasma cell-free DNA methylome-based liquid biopsy for accurate gastric cancer detection","authors":"Jian Qi, Bo Hong, Shujie Wang, Jingyun Wang, Jinman Fang, Ruifang Sun, Jinfu Nie, Hongzhi Wang","doi":"10.1111/cas.16284","DOIUrl":"10.1111/cas.16284","url":null,"abstract":"<p>Early detection plays a critical role in mitigating mortality rates linked to gastric cancer. However, current clinical screening methods exhibit suboptimal efficacy. Methylation alterations identified from cell-free DNA (cfDNA) present a promising biomarker for early cancer detection. Our study focused on identifying gastric cancer-specific markers from cfDNA methylation to facilitate early detection. We enrolled 150 gastric cancer patients and 100 healthy controls in this study, and undertook genome-wide methylation profiling of cfDNA using cell-free methylated DNA immunoprecipitation and high-throughput sequencing. We identified 21 differentially methylated regions (DMRs) between the gastric tumor and nontumor groups using multiple algorithms. Subsequently, using the 21 DMRs, we developed a gastric cancer detection model by random forest algorithm in the discovery set, and validated the model in an independent set. The model was able to accurately discriminate gastric cancer with a sensitivity and specificity of 93.90% and 95.15% in the discovery set, respectively, and 88.38% and 94.23% in the validation set, respectively. These results underscore the efficacy and accuracy of cfDNA-derived methylation markers in distinguishing early stage gastric cancer. This study highlighted the significance of cfDNA methylation alterations in early gastric cancer detection.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3426-3438"},"PeriodicalIF":4.5,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447983/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141749397","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}
Retraction: T.-Q. Yang, X.-J. Lu, T.-F. Wu, D.-D. Ding, Z.-H. Zhao, G.-L. Chen, X.-S. Xie, B. Li, Y.-X. Wei, L.-C. Guo, Y. Zhang, Y.-L. Huang, Y.-X. Zhou, and Z.-W. Du, “MicroRNA-16 inhibits glioma cell growth and invasion through suppression of BCL2 and the nuclear factor-κB1/MMP9 signaling pathway,” Cancer Science 105 no. 3 (2014): 265–271. https://doi.org/10.1111/cas.12351.
The above article, published online on 13 January 2014 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the journal Editor-in-Chief, Masanori Hatakeyama; the Japanese Cancer Association; and John Wiley and Sons Ltd. The journal had previously published a correction regarding duplicated portions of Figure 4a on 24 May 2022. Following publication of this correction, the journal was made aware that not all duplicated portions of Figure 4a had been replaced. Additionally, concerns were raised of splicing in Figure 2d and image duplications in Figures 2d, 3d, and 4c. The authors did not respond to inquiries from the publisher regarding these concerns. and did not provide the requested original images and data. Therefore, the data reported in this article are considered unreliable and the journal must issue a retraction. The authors did not respond to our notice of retraction.
撤回:T.-Q.Yang, X.-J. Lu, T.-F.Wu, D.-D. Ding, Z.-H.Ding, Z.-H. Zhao, G.-L. Chen, X.-S.Zhao, G.-L. Chen, X.-S.Xie, B. Li, Y.-X.Wei, L.-C. Guo, Y. Zhang, Y.-X.Guo, Y. Zhang, Y.-L. Huang, Y.-X.Zhou,and Z.-W. Du,"MicroRNA-16 inhibited MicroRNADu, "MicroRNA-16 inhibits glioma cell growth and invasion through suppression of BCL2 and the nuclear factor-κB1/MMP9 signaling pathway," Cancer Science 105 no. 3 (2014): 265-271. https://doi.org/10.1111/cas.12351。上述文章于 2014 年 1 月 13 日在线发表于 Wiley Online Library (wileyonlinelibrary.com),经杂志主编 Masanori Hatakeyama、日本癌症协会和 John Wiley and Sons Ltd.(约翰-威利父子有限公司)协商,该文章已被撤回。该期刊曾于 2022 年 5 月 24 日就图 4a 中重复的部分发表了更正。更正发表后,本刊了解到图 4a 的重复部分并未全部替换。此外,图 2d 中的拼接以及图 2d、3d 和 4c 中的图像重复也引起了关注。作者没有回复出版商关于这些问题的询问,也没有提供所要求的原始图像和数据。因此,这篇文章中报告的数据被认为是不可靠的,期刊必须发表撤稿声明。作者没有回应我们的撤稿通知。
{"title":"RETRACTION: MicroRNA-16 inhibits glioma cell growth and invasion through suppression of BCL2 and the nuclear factor-κB1/MMP9 signaling pathway","authors":"","doi":"10.1111/cas.16269","DOIUrl":"10.1111/cas.16269","url":null,"abstract":"<p><b>Retraction</b>: T.-Q. Yang, X.-J. Lu, T.-F. Wu, D.-D. Ding, Z.-H. Zhao, G.-L. Chen, X.-S. Xie, B. Li, Y.-X. Wei, L.-C. Guo, Y. Zhang, Y.-L. Huang, Y.-X. Zhou, and Z.-W. Du, “MicroRNA-16 inhibits glioma cell growth and invasion through suppression of BCL2 and the nuclear factor-κB1/MMP9 signaling pathway,” <i>Cancer Science</i> 105 no. 3 (2014): 265–271. https://doi.org/10.1111/cas.12351.</p><p>The above article, published online on 13 January 2014 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the journal Editor-in-Chief, Masanori Hatakeyama; the Japanese Cancer Association; and John Wiley and Sons Ltd. The journal had previously published a correction regarding duplicated portions of Figure 4a on 24 May 2022. Following publication of this correction, the journal was made aware that not all duplicated portions of Figure 4a had been replaced. Additionally, concerns were raised of splicing in Figure 2d and image duplications in Figures 2d, 3d, and 4c. The authors did not respond to inquiries from the publisher regarding these concerns. and did not provide the requested original images and data. Therefore, the data reported in this article are considered unreliable and the journal must issue a retraction. The authors did not respond to our notice of retraction.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 9","pages":"3200"},"PeriodicalIF":4.5,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.16269","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141735376","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}
Ziyi Xue, Haijing Xie, Ying Shan, Lin Zhang, Lin Cheng, Wenyue Chen, Rui Zhu, Kaiwen Zhang, Haosheng Ni, Zhenxin Zhang, Yiwen You, Bo You
Sorafenib, an anticancer drug, has been shown to induce ferroptosis in cancer cells. However, resistance to sorafenib greatly limits its therapeutic efficacy, and the exact mechanism of resistance is not fully understood. This study investigated the role of N-Acetyltransferase 10 (NAT10) in influencing the anticancer activity of sorafenib in nasopharyngeal carcinoma (NPC) and its molecular mechanism. NAT10 expression was significantly upregulated in NPC. Mechanistically, NAT10 promotes proteins of solute carrier family 7 member 11 (SLC7A11) expression through ac4C acetylation, inhibiting sorafenib-induced ferroptosis in NPC cells. The combined application of sorafenib and the NAT10 inhibitor remodelin significantly inhibits SLC7A11 expression and promotes ferroptosis in NPC cells. In vivo knockout of NAT10 inhibited the growth of sorafenib-resistant NPC. Our findings suggest that NAT10 inhibition might be a promising therapeutic approach to enhance the anticancer activity of sorafenib.
{"title":"NAT10 inhibition promotes ac4C-dependent ferroptosis to counteract sorafenib resistance in nasopharyngeal carcinoma","authors":"Ziyi Xue, Haijing Xie, Ying Shan, Lin Zhang, Lin Cheng, Wenyue Chen, Rui Zhu, Kaiwen Zhang, Haosheng Ni, Zhenxin Zhang, Yiwen You, Bo You","doi":"10.1111/cas.16249","DOIUrl":"10.1111/cas.16249","url":null,"abstract":"<p>Sorafenib, an anticancer drug, has been shown to induce ferroptosis in cancer cells. However, resistance to sorafenib greatly limits its therapeutic efficacy, and the exact mechanism of resistance is not fully understood. This study investigated the role of N-Acetyltransferase 10 (NAT10) in influencing the anticancer activity of sorafenib in nasopharyngeal carcinoma (NPC) and its molecular mechanism. NAT10 expression was significantly upregulated in NPC. Mechanistically, NAT10 promotes proteins of solute carrier family 7 member 11 (SLC7A11) expression through ac4C acetylation, inhibiting sorafenib-induced ferroptosis in NPC cells. The combined application of sorafenib and the NAT10 inhibitor remodelin significantly inhibits SLC7A11 expression and promotes ferroptosis in NPC cells. In vivo knockout of NAT10 inhibited the growth of sorafenib-resistant NPC. Our findings suggest that NAT10 inhibition might be a promising therapeutic approach to enhance the anticancer activity of sorafenib.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"115 10","pages":"3256-3272"},"PeriodicalIF":4.5,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11447888/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141749396","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}