首页 > 最新文献

Cancer research最新文献

英文 中文
A Pan-RAS Inhibitor with a Unique Mechanism of Action Blocks Tumor Growth and Induces Antitumor Immunity in Gastrointestinal Cancer
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-19 DOI: 10.1158/0008-5472.can-24-0323
Jeremy B. Foote, Tyler E. Mattox, Adam B. Keeton, Xi Chen, Forrest T. Smith, Kristy Berry, Thomas W. Holmes, Junwei Wang, Chung-hui Huang, Antonio Ward, AMIT K. Mitra, Veronica Ramirez-Alcantara, Cherlene Hardy, Karianne G. Fleten, Kjersti Flatmark, Karina J. Yoon, Sujith Sarvesh, Ganji P. Nagaraju, Dhana Sekhar Reddy Bandi, Yulia Y. Maxuitenko, Jacaob Valiyaveettil, Julienne L. Carstens, Donald J. Buchsbaum, Jennifer Yang, Gang Zhou, Elmar Nurmemmedov, Ivan Babic, Vadim Gaponeko, Hazem Abdelkarim, Michael R. Boyd, Greg Gorman, Upender Manne, Sejong Bae, Bassel F. El-Rayes, Gary A. Piazza
RAS is a common driver of cancer that was considered undruggable for decades. Recent advances have enabled the development of RAS inhibitors, but the efficacy of these inhibitors remains limited by resistance. Here, we developed a pan-RAS inhibitor, ADT-007, that binds nucleotide-free RAS to block GTP activation of effector interactions and MAPK/AKT signaling, resulting in mitotic arrest and apoptosis. ADT-007 potently inhibited the growth of RAS mutant cancer cells irrespective of the RAS mutation or isozyme, and RASWT cancer cells with GTP-activated RAS from upstream mutations were equally sensitive. Conversely, RASWT cancer cells harboring downstream BRAF mutations and normal cells were essentially insensitive to ADT-007. Sensitivity of cancer cells to ADT-007 required activated RAS and dependence on RAS for proliferation, while insensitivity was attributed to metabolic deactivation by UDP-glucuronosyltransferases that were expressed in RASWT and normal cells but repressed in RAS mutant cancer cells. ADT-007 displayed unique advantages over KRAS mutant-specific, pan-KRAS, and pan-RAS inhibitors that could impact in vivo antitumor efficacy by escaping compensatory mechanisms that lead to resistance. Local administration of ADT-007 showed robust antitumor activity in syngeneic immune-competent and xenogeneic immune-deficient mouse models of colorectal and pancreatic cancer. The antitumor activity of ADT-007 was associated with the suppression of MAPK signaling and activation of innate and adaptive immunity in the tumor immune microenvironment. Oral administration of ADT-007 prodrug also inhibited tumor growth. Thus, ADT-007 has the potential to address the complex RAS mutational landscape of many human cancers and to improve treatment of RAS-driven tumors.
{"title":"A Pan-RAS Inhibitor with a Unique Mechanism of Action Blocks Tumor Growth and Induces Antitumor Immunity in Gastrointestinal Cancer","authors":"Jeremy B. Foote, Tyler E. Mattox, Adam B. Keeton, Xi Chen, Forrest T. Smith, Kristy Berry, Thomas W. Holmes, Junwei Wang, Chung-hui Huang, Antonio Ward, AMIT K. Mitra, Veronica Ramirez-Alcantara, Cherlene Hardy, Karianne G. Fleten, Kjersti Flatmark, Karina J. Yoon, Sujith Sarvesh, Ganji P. Nagaraju, Dhana Sekhar Reddy Bandi, Yulia Y. Maxuitenko, Jacaob Valiyaveettil, Julienne L. Carstens, Donald J. Buchsbaum, Jennifer Yang, Gang Zhou, Elmar Nurmemmedov, Ivan Babic, Vadim Gaponeko, Hazem Abdelkarim, Michael R. Boyd, Greg Gorman, Upender Manne, Sejong Bae, Bassel F. El-Rayes, Gary A. Piazza","doi":"10.1158/0008-5472.can-24-0323","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-0323","url":null,"abstract":"RAS is a common driver of cancer that was considered undruggable for decades. Recent advances have enabled the development of RAS inhibitors, but the efficacy of these inhibitors remains limited by resistance. Here, we developed a pan-RAS inhibitor, ADT-007, that binds nucleotide-free RAS to block GTP activation of effector interactions and MAPK/AKT signaling, resulting in mitotic arrest and apoptosis. ADT-007 potently inhibited the growth of RAS mutant cancer cells irrespective of the RAS mutation or isozyme, and RASWT cancer cells with GTP-activated RAS from upstream mutations were equally sensitive. Conversely, RASWT cancer cells harboring downstream BRAF mutations and normal cells were essentially insensitive to ADT-007. Sensitivity of cancer cells to ADT-007 required activated RAS and dependence on RAS for proliferation, while insensitivity was attributed to metabolic deactivation by UDP-glucuronosyltransferases that were expressed in RASWT and normal cells but repressed in RAS mutant cancer cells. ADT-007 displayed unique advantages over KRAS mutant-specific, pan-KRAS, and pan-RAS inhibitors that could impact in vivo antitumor efficacy by escaping compensatory mechanisms that lead to resistance. Local administration of ADT-007 showed robust antitumor activity in syngeneic immune-competent and xenogeneic immune-deficient mouse models of colorectal and pancreatic cancer. The antitumor activity of ADT-007 was associated with the suppression of MAPK signaling and activation of innate and adaptive immunity in the tumor immune microenvironment. Oral administration of ADT-007 prodrug also inhibited tumor growth. Thus, ADT-007 has the potential to address the complex RAS mutational landscape of many human cancers and to improve treatment of RAS-driven tumors.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"29 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142858409","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
CLK1 Activates YAP to Promote Intrahepatic Cholangiocarcinogenesis
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-18 DOI: 10.1158/0008-5472.can-24-0147
Shuai Xue, Xiangzheng Chen, Guoteng Qiu, Haotian Liao, Zeyuan Qiang, Zheng Zhang, Xuping Feng, Lin Xu, Rui Xie, Hongyu Zhou, Jiwei Huang, Yong Zeng, Haichuan Wang
Cdc2-like kinase 1 (CLK1) has dual-specificity kinase ability to phosphorylate tyrosine and serine/threonine protein residues. CLK1 regulates many physiological processes and has been shown to contribute to multiple types of cancer. Here, we investigated the functional role of CLK1 during intrahepatic cholangiocarcinoma (ICC) development. The expression of CLK1 was elevated in ICC tumors, and patients with high expression of CLK1 demonstrated poor prognosis. In hydrodynamically transfected mouse models, CLK1 alone was insufficient to induce ICC, whereas CLK1 cooperated with AKT (AKT/CLK1) to trigger ICC initiation. In addition, overexpression of CLK1 in ICC cells facilitated proliferation in vitro and tumor growth in vivo, while loss of CLK1 elicited the opposite effects. Moreover, RNAseq analysis indicated that high levels of CLK1 corresponded with activation of the Hippo-YAP signaling pathway. Consistently, the AKT/CLK1 murine tumors displayed upregulation of YAP as well as its downstream targets. Furthermore, loss or pharmacological inhibition YAP in ICC cells inhibited CLK1-induced growth, and deletion of Yap completely retarded the induction of AKT/CLK1 tumors. Mechanistically, 4D-label free mass spectrometry and co-immunoprecipitation assays revealed WWC2 as a potential mediator of CLK1-YAP cascade. Collectively, the current findings identify a critical role for CLK1 in promoting ICC development and indicate that inhibiting YAP might be an effective approach for perturbing CLK1-mediated tumorigenesis.
{"title":"CLK1 Activates YAP to Promote Intrahepatic Cholangiocarcinogenesis","authors":"Shuai Xue, Xiangzheng Chen, Guoteng Qiu, Haotian Liao, Zeyuan Qiang, Zheng Zhang, Xuping Feng, Lin Xu, Rui Xie, Hongyu Zhou, Jiwei Huang, Yong Zeng, Haichuan Wang","doi":"10.1158/0008-5472.can-24-0147","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-0147","url":null,"abstract":"Cdc2-like kinase 1 (CLK1) has dual-specificity kinase ability to phosphorylate tyrosine and serine/threonine protein residues. CLK1 regulates many physiological processes and has been shown to contribute to multiple types of cancer. Here, we investigated the functional role of CLK1 during intrahepatic cholangiocarcinoma (ICC) development. The expression of CLK1 was elevated in ICC tumors, and patients with high expression of CLK1 demonstrated poor prognosis. In hydrodynamically transfected mouse models, CLK1 alone was insufficient to induce ICC, whereas CLK1 cooperated with AKT (AKT/CLK1) to trigger ICC initiation. In addition, overexpression of CLK1 in ICC cells facilitated proliferation in vitro and tumor growth in vivo, while loss of CLK1 elicited the opposite effects. Moreover, RNAseq analysis indicated that high levels of CLK1 corresponded with activation of the Hippo-YAP signaling pathway. Consistently, the AKT/CLK1 murine tumors displayed upregulation of YAP as well as its downstream targets. Furthermore, loss or pharmacological inhibition YAP in ICC cells inhibited CLK1-induced growth, and deletion of Yap completely retarded the induction of AKT/CLK1 tumors. Mechanistically, 4D-label free mass spectrometry and co-immunoprecipitation assays revealed WWC2 as a potential mediator of CLK1-YAP cascade. Collectively, the current findings identify a critical role for CLK1 in promoting ICC development and indicate that inhibiting YAP might be an effective approach for perturbing CLK1-mediated tumorigenesis.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"109 21 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142849170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Antibody-Drug Conjugates Targeting the EGFR Ligand Epiregulin Elicit Robust Anti-Tumor Activity in Colorectal Cancer
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-18 DOI: 10.1158/0008-5472.can-24-0798
Joan Jacob, Yasuaki Anami, Peyton C. High, Zhengdong Liang, Shraddha Subramanian, Sukhen C. Ghosh, Solmaz AghaAmiri, Cara Guernsey-Biddle, Ha Tran, Julie Rowe, Ali Azhdarinia, Kyoji Tsuchikama, Kendra S. Carmon
As colorectal cancer (CRC) remains a leading cause of cancer-related death, identifying therapeutic targets and approaches is essential to improve patient outcomes. The EGFR ligand epiregulin (EREG) is highly expressed in RAS wildtype and mutant CRC with minimal expression in normal tissues, making it an attractive target for antibody-drug conjugate (ADC) development. In this study, we produced and purified an EREG monoclonal antibody (mAb), H231, that had high specificity and affinity for human and mouse EREG. H231 also internalized to lysosomes, which is important for ADC payload release. ImmunoPET and ex vivo biodistribution studies showed significant tumor uptake of 89Zr-labeled H231 with minimal uptake in normal tissues. H231 was conjugated to either cleavable dipeptide or tripeptide chemical linkers attached to the DNA-alkylating payload duocarmycin DM, and cytotoxicity of EREG ADCs was assessed in a panel of CRC cell lines. EREG ADCs incorporating tripeptide linkers demonstrated the highest potency in EREG-expressing CRC cells irrespective of RAS mutations. Preclinical safety and efficacy studies showed EREG ADCs were well-tolerated, neutralized EGFR pathway activity, caused significant tumor growth inhibition or regression, and increased survival in CRC cell line and patient-derived xenograft models. These data suggest EREG is a promising target for the development of ADCs for treating CRC and other cancer types that express high levels of EREG. While the efficacy of clinically approved anti-EGFR mAbs are largely limited by RAS mutational status, EREG ADCs may show promise for both RAS mutant and wildtype patients, thus improving existing treatment options.
{"title":"Antibody-Drug Conjugates Targeting the EGFR Ligand Epiregulin Elicit Robust Anti-Tumor Activity in Colorectal Cancer","authors":"Joan Jacob, Yasuaki Anami, Peyton C. High, Zhengdong Liang, Shraddha Subramanian, Sukhen C. Ghosh, Solmaz AghaAmiri, Cara Guernsey-Biddle, Ha Tran, Julie Rowe, Ali Azhdarinia, Kyoji Tsuchikama, Kendra S. Carmon","doi":"10.1158/0008-5472.can-24-0798","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-0798","url":null,"abstract":"As colorectal cancer (CRC) remains a leading cause of cancer-related death, identifying therapeutic targets and approaches is essential to improve patient outcomes. The EGFR ligand epiregulin (EREG) is highly expressed in RAS wildtype and mutant CRC with minimal expression in normal tissues, making it an attractive target for antibody-drug conjugate (ADC) development. In this study, we produced and purified an EREG monoclonal antibody (mAb), H231, that had high specificity and affinity for human and mouse EREG. H231 also internalized to lysosomes, which is important for ADC payload release. ImmunoPET and ex vivo biodistribution studies showed significant tumor uptake of 89Zr-labeled H231 with minimal uptake in normal tissues. H231 was conjugated to either cleavable dipeptide or tripeptide chemical linkers attached to the DNA-alkylating payload duocarmycin DM, and cytotoxicity of EREG ADCs was assessed in a panel of CRC cell lines. EREG ADCs incorporating tripeptide linkers demonstrated the highest potency in EREG-expressing CRC cells irrespective of RAS mutations. Preclinical safety and efficacy studies showed EREG ADCs were well-tolerated, neutralized EGFR pathway activity, caused significant tumor growth inhibition or regression, and increased survival in CRC cell line and patient-derived xenograft models. These data suggest EREG is a promising target for the development of ADCs for treating CRC and other cancer types that express high levels of EREG. While the efficacy of clinically approved anti-EGFR mAbs are largely limited by RAS mutational status, EREG ADCs may show promise for both RAS mutant and wildtype patients, thus improving existing treatment options.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"260 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142849173","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Selective Enhancer Gain-of-Function Deregulates MYC Expression in Multiple Myeloma. 选择性增强子功能增益可解除多发性骨髓瘤中 MYC 的表达。
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-16 DOI: 10.1158/0008-5472.CAN-24-1440
Mahshid Rahmat, Kendell Clement, Jean-Baptiste Alberge, Romanos Sklavenitis-Pistofidis, Rohan Kodgule, Charles P Fulco, Daniel Heilpern-Mallory, Katarina Nilsson, David Dorfman, Jesse M Engreitz, Gad Getz, Luca Pinello, Russell J H Ryan, Irene M Ghobrial

MYC deregulation occurs in the majority of multiple myeloma cases and is associated with progression and worse prognosis. Enhanced MYC expression occurs in about 70% of patients with multiple myeloma, but it is known to be driven by translocation or amplification events in only ∼40% of myelomas. Here, we used CRISPR interference to uncover an epigenetic mechanism of MYC regulation whereby increased accessibility of a plasma cell-type-specific enhancer leads to increased MYC expression. This native enhancer activity was not associated with enhancer hijacking events but led to specific binding of cMAF, IRF4, and SPIB transcription factors that activated MYC expression in the absence of known genetic aberrations. In addition, focal amplification was another mechanism of activation of this enhancer in approximately 3.4% of patients with multiple myeloma. Together, these findings define an epigenetic mechanism of MYC deregulation in multiple myeloma beyond known translocations or amplifications and point to the importance of noncoding regulatory elements and their associated transcription factor networks as drivers of multiple myeloma progression. Significance: The discovery of a native developmental enhancer that sustains the expression of MYC in a subset of myelomas could help identify predictive biomarkers and therapeutic targets to improve the outcomes of patients with multiple myeloma.

MYC失调发生在大多数多发性骨髓瘤(MM)病例中,与病情进展和预后恶化有关。约70%的MM患者会出现MYC表达增强,但已知只有约40%的骨髓瘤是由易位或扩增事件驱动的。在这里,我们利用CRISPR干扰(CRISPRi)发现了MYC调控的表观遗传学机制,即浆细胞型特异性增强子的可及性增加导致MYC表达增加。这种原生增强子活性与增强子劫持事件无关,但会导致 c-MAF、IRF4 和 SPIB 转录因子的特异性结合,从而在没有已知基因畸变的情况下激活 MYC 的表达。此外,在约 3.4% 的 MM 患者中,病灶扩增是激活该增强子的另一种机制。这些发现共同确定了MM中MYC失调的表观遗传学机制,而不是已知的易位或扩增,并指出了非编码调控元件及其相关转录因子网络作为MM进展驱动因素的重要性。
{"title":"Selective Enhancer Gain-of-Function Deregulates MYC Expression in Multiple Myeloma.","authors":"Mahshid Rahmat, Kendell Clement, Jean-Baptiste Alberge, Romanos Sklavenitis-Pistofidis, Rohan Kodgule, Charles P Fulco, Daniel Heilpern-Mallory, Katarina Nilsson, David Dorfman, Jesse M Engreitz, Gad Getz, Luca Pinello, Russell J H Ryan, Irene M Ghobrial","doi":"10.1158/0008-5472.CAN-24-1440","DOIUrl":"10.1158/0008-5472.CAN-24-1440","url":null,"abstract":"<p><p>MYC deregulation occurs in the majority of multiple myeloma cases and is associated with progression and worse prognosis. Enhanced MYC expression occurs in about 70% of patients with multiple myeloma, but it is known to be driven by translocation or amplification events in only ∼40% of myelomas. Here, we used CRISPR interference to uncover an epigenetic mechanism of MYC regulation whereby increased accessibility of a plasma cell-type-specific enhancer leads to increased MYC expression. This native enhancer activity was not associated with enhancer hijacking events but led to specific binding of cMAF, IRF4, and SPIB transcription factors that activated MYC expression in the absence of known genetic aberrations. In addition, focal amplification was another mechanism of activation of this enhancer in approximately 3.4% of patients with multiple myeloma. Together, these findings define an epigenetic mechanism of MYC deregulation in multiple myeloma beyond known translocations or amplifications and point to the importance of noncoding regulatory elements and their associated transcription factor networks as drivers of multiple myeloma progression. Significance: The discovery of a native developmental enhancer that sustains the expression of MYC in a subset of myelomas could help identify predictive biomarkers and therapeutic targets to improve the outcomes of patients with multiple myeloma.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"4173-4183"},"PeriodicalIF":12.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11649448/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280502","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Targeting SHCBP1 Inhibits Tumor Progression by Restoring Ciliogenesis in Ductal Carcinoma. 靶向 SHCBP1 可通过恢复乳腺导管癌中的纤毛生成抑制肿瘤进展
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-16 DOI: 10.1158/0008-5472.CAN-24-1095
Wengui Shi, Lianshun Li, Huiming Zhao, Zhengyang Li, Zhijian Ma, Qianlin Gu, Huili Ye, Xiangyan Jiang, Yuman Dong, Long Qin, Huinian Zhou, Zeyuan Yu, Zuoyi Jiao

Primary cilia detect and transmit environmental signals into cells. Primary cilia are absent in a subset of ductal carcinomas characterized by distinctive biological activities, and recovery of cilia with normal functionality has been shown to have therapeutic potential in some cancer types. Therefore, elucidation of the underlying mechanism and clinical significance of ciliary loss in ductal carcinomas could help develop effective treatment strategies. Here, we identified a link between Shc1-binding protein (SHCBP1) and cilia in ductal carcinomas. Shcbp1 knockout in transgenic mice profoundly impeded tumor progression and metastasis, prolonging survival. Single-cell transcriptome analysis revealed a functional connection between SHCBP1 deficiency and increased tumor ciliogenesis. SHCBP1 ablation restored ciliogenesis in unciliated ductal carcinoma by promoting the proximity between the midbody remnant (MBR) and centrosome through enhanced Rab8 GTPase activity and Rab8GTP positioning within the MBR. Inhibition of tumor progression by SHCBP1 loss relied on the recovery of ciliogenesis. Analysis of a large cohort of patients with ductal carcinoma revealed a negative correlation between SHCBP1-induced ciliary loss and patient prognosis. Restoring ciliogenesis via SHCBP1 ablation elicited therapeutic effects in patient-derived xenograft models. Together, this study delineates that induction of MBR-centrosome proximity through SHCBP1-deficiency reactivates ciliogenesis, offering unique opportunities for the treatment of unciliated ductal carcinomas. Significance: SHCBP1 depletion rescues tumor ciliogenesis by enhancing Rab8 GTPase activity to restore the proximity of the  midbody remnant to the centrosome, which impedes progression of ductal carcinomas and suggests potential therapeutic strategies.

原生纤毛能探测环境信号并将其传递到细胞中。原发性纤毛缺失存在于一部分具有独特生物活性的导管癌中,而恢复纤毛的正常功能已被证明在某些癌症类型中具有治疗潜力。因此,阐明导管癌中纤毛缺失的内在机制和临床意义有助于制定有效的治疗策略。在这里,我们发现了导管癌中SHCBP1与纤毛之间的联系。在转基因小鼠中敲除 Shcbp1 能显著抑制肿瘤的发展和转移,延长生存期。单细胞转录组分析揭示了 SHCBP1 缺乏与肿瘤纤毛生成增加之间的功能性联系。SHCBP1 消融通过增强 Rab8 GTPase 活性和 Rab8GTP 在中体残基(MBR)内的定位,促进中体残基与中心体之间的接近,从而恢复无纤毛导管癌的纤毛生成。SHCBP1 缺失对肿瘤进展的抑制依赖于纤毛生成的恢复。对一大批导管癌患者的分析表明,SHCBP1 诱导的纤毛缺失与患者的预后呈负相关。通过消融 SHCBP1 恢复纤毛生成在患者异种移植模型中产生了治疗效果。总之,这项研究阐明了通过 SHCBP1 缺失诱导 MBR-中心体接近可重新激活纤毛生成,为治疗无纤毛导管癌提供了独特的机会。
{"title":"Targeting SHCBP1 Inhibits Tumor Progression by Restoring Ciliogenesis in Ductal Carcinoma.","authors":"Wengui Shi, Lianshun Li, Huiming Zhao, Zhengyang Li, Zhijian Ma, Qianlin Gu, Huili Ye, Xiangyan Jiang, Yuman Dong, Long Qin, Huinian Zhou, Zeyuan Yu, Zuoyi Jiao","doi":"10.1158/0008-5472.CAN-24-1095","DOIUrl":"10.1158/0008-5472.CAN-24-1095","url":null,"abstract":"<p><p>Primary cilia detect and transmit environmental signals into cells. Primary cilia are absent in a subset of ductal carcinomas characterized by distinctive biological activities, and recovery of cilia with normal functionality has been shown to have therapeutic potential in some cancer types. Therefore, elucidation of the underlying mechanism and clinical significance of ciliary loss in ductal carcinomas could help develop effective treatment strategies. Here, we identified a link between Shc1-binding protein (SHCBP1) and cilia in ductal carcinomas. Shcbp1 knockout in transgenic mice profoundly impeded tumor progression and metastasis, prolonging survival. Single-cell transcriptome analysis revealed a functional connection between SHCBP1 deficiency and increased tumor ciliogenesis. SHCBP1 ablation restored ciliogenesis in unciliated ductal carcinoma by promoting the proximity between the midbody remnant (MBR) and centrosome through enhanced Rab8 GTPase activity and Rab8GTP positioning within the MBR. Inhibition of tumor progression by SHCBP1 loss relied on the recovery of ciliogenesis. Analysis of a large cohort of patients with ductal carcinoma revealed a negative correlation between SHCBP1-induced ciliary loss and patient prognosis. Restoring ciliogenesis via SHCBP1 ablation elicited therapeutic effects in patient-derived xenograft models. Together, this study delineates that induction of MBR-centrosome proximity through SHCBP1-deficiency reactivates ciliogenesis, offering unique opportunities for the treatment of unciliated ductal carcinomas. Significance: SHCBP1 depletion rescues tumor ciliogenesis by enhancing Rab8 GTPase activity to restore the proximity of the  midbody remnant to the centrosome, which impedes progression of ductal carcinomas and suggests potential therapeutic strategies.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"4156-4172"},"PeriodicalIF":12.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280504","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
The Innate Immune System and the TRAIL-Bcl-XL Axis Mediate a Sex Bias in Lung Cancer and Confer a Therapeutic Vulnerability in Females. 先天性免疫系统和TRAIL-BCL-XL轴介导肺癌的性别差异并使女性更易接受治疗
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-16 DOI: 10.1158/0008-5472.CAN-24-0585
Lauren May, Bin Hu, Preksha Jerajani, Akash Jagdeesh, Ohud Alhawiti, Lillian Cai, Nina Semenova, Chunqing Guo, Madison Isbell, Xiaoyan Deng, Anthony C Faber, Raghavendra Pillappa, Dipankar Bandyopadhyay, Xiang-Yang Wang, Alexander Neuwelt, Jennifer Koblinski, Paula D Bos, Howard Li, Rebecca Martin, Joseph W Landry

There is a significant sex bias in lung cancer, with males showing increased mortality compared with females. A better mechanistic understanding of these differences could help identify therapeutic targets to personalize cancer therapies to each sex. After observing a clear sex bias in humanized mice, with male patient-derived xenograft lung tumors being more progressive and deadlier than female patient-derived xenograft lung tumors, we identified mouse tumor models of lung cancer with the same sex bias. This sex bias was not observed in models of breast, colon, melanoma, and renal cancers. In vivo, the sex bias in growth and lethality required intact ovaries, functional innate NK cells and monocytes/macrophages, and the activating receptor NKG2D. Ex vivo cell culture models were sensitized to the anticancer effects of NKG2D-mediated NK cell and macrophage killing through the TRAIL-Bcl-XL axis when cultured with serum from female mice with intact ovaries. In both flank and orthotopic models, the Bcl-XL inhibitor navitoclax (ABT-263) improved tumor growth control in female mice and required NK cells, macrophages, and the TRAIL signaling pathway. This research suggests that navitoclax and TRAIL pathway agonists could be used as a personalized therapy to improve outcomes in women with lung cancer. Significance: Lung cancers in females are more susceptible to killing through a TRAIL-Bcl-XL axis, indicating that targeting this axis therapeutically could represent a personalized approach to treat female patients with lung cancer.

肺癌有明显的性别差异,男性死亡率高于女性。从机理上更好地了解这些差异有助于确定治疗靶点,从而针对不同性别的患者制定个性化的癌症疗法。在人源化小鼠中观察到明显的性别偏倚后,我们确定了具有相同性别偏倚的肺癌小鼠肿瘤模型。而在乳腺癌、结肠癌、黑色素瘤和肾癌模型中则没有观察到这种性别偏倚。在体内,生长和致死的性别偏倚需要完整的卵巢、功能性先天性自然杀伤(NK)细胞和单核细胞/巨噬细胞以及激活受体 NKG2D。用卵巢完好的雌性小鼠血清培养体外细胞培养模型时,NKG2D通过TRAIL-BCL-XL轴介导的NK细胞和巨噬细胞杀伤作用对其抗癌效果具有敏感性。在侧腹和正位模型中,BCL-XL抑制剂navitoclax(ABT-263)改善了雌性小鼠的肿瘤生长控制,并且需要NK细胞、巨噬细胞和TRAIL信号通路。这项研究表明,navitoclax和TRAIL通路激动剂可作为一种个性化疗法来改善女性肺癌患者的预后。
{"title":"The Innate Immune System and the TRAIL-Bcl-XL Axis Mediate a Sex Bias in Lung Cancer and Confer a Therapeutic Vulnerability in Females.","authors":"Lauren May, Bin Hu, Preksha Jerajani, Akash Jagdeesh, Ohud Alhawiti, Lillian Cai, Nina Semenova, Chunqing Guo, Madison Isbell, Xiaoyan Deng, Anthony C Faber, Raghavendra Pillappa, Dipankar Bandyopadhyay, Xiang-Yang Wang, Alexander Neuwelt, Jennifer Koblinski, Paula D Bos, Howard Li, Rebecca Martin, Joseph W Landry","doi":"10.1158/0008-5472.CAN-24-0585","DOIUrl":"10.1158/0008-5472.CAN-24-0585","url":null,"abstract":"<p><p>There is a significant sex bias in lung cancer, with males showing increased mortality compared with females. A better mechanistic understanding of these differences could help identify therapeutic targets to personalize cancer therapies to each sex. After observing a clear sex bias in humanized mice, with male patient-derived xenograft lung tumors being more progressive and deadlier than female patient-derived xenograft lung tumors, we identified mouse tumor models of lung cancer with the same sex bias. This sex bias was not observed in models of breast, colon, melanoma, and renal cancers. In vivo, the sex bias in growth and lethality required intact ovaries, functional innate NK cells and monocytes/macrophages, and the activating receptor NKG2D. Ex vivo cell culture models were sensitized to the anticancer effects of NKG2D-mediated NK cell and macrophage killing through the TRAIL-Bcl-XL axis when cultured with serum from female mice with intact ovaries. In both flank and orthotopic models, the Bcl-XL inhibitor navitoclax (ABT-263) improved tumor growth control in female mice and required NK cells, macrophages, and the TRAIL signaling pathway. This research suggests that navitoclax and TRAIL pathway agonists could be used as a personalized therapy to improve outcomes in women with lung cancer. Significance: Lung cancers in females are more susceptible to killing through a TRAIL-Bcl-XL axis, indicating that targeting this axis therapeutically could represent a personalized approach to treat female patients with lung cancer.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"4140-4155"},"PeriodicalIF":12.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11649478/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280505","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Pharmacologic Blockade of a Pioneer Transcription Factor. 药理阻断先锋转录因子。
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-16 DOI: 10.1158/0008-5472.CAN-24-3957
Katerina Cermakova, H Courtney Hodges

Cancers frequently co-opt lineage-specific transcription factors (TF) utilized in normal development to sustain proliferation. However, the effects of these TFs on tumor development depend considerably on where in the genome they bind. A new article by Taylor and colleagues expands on previously developed diamidine compounds that obstruct the DNA binding sites of the pioneer TF PU.1 (SPI1) in acute myeloid leukemia. Immobilization and sequencing of genomic DNA targeted by these compounds revealed that these inhibitors alter the genomic binding patterns of PU.1. The authors report that their strategy constrains the genomic binding preferences of PU.1, leading to redistribution of PU.1 to promoters and other gene-proximal regions with elevated guanine/cytosine content. In this study, we discuss recent developments for targeting PU.1 in hematologic malignancies. We also explore the shared functional roles of PU.1 and SWI/SNF ATP-dependent chromatin remodeling complexes, which not only work together to sustain the enhancer landscape needed for tumor cell proliferation but also play key roles in nontumor settings.

癌症经常通过利用正常发育过程中的特异性转录因子(TFs)来维持增殖。然而,这些转录因子对肿瘤发生的影响在很大程度上取决于它们在基因组中的结合位点。泰勒及其同事的一篇新论文扩展了之前开发的二脒化合物,这些化合物阻碍了急性髓性白血病(AML)中先锋转录因子 PU.1 (SPI1)的 DNA 结合位点。对这些化合物靶向的基因组 DNA 进行固定和测序后发现,这些抑制剂改变了 PU.1 的基因组结合模式。作者报告说,他们的策略限制了 PU.1 的基因组结合偏好,导致 PU.1 重新分布到启动子和其他 G/C 含量升高的基因近端区域。在此,我们讨论了在血液恶性肿瘤中靶向 PU.1 的最新进展。我们还探讨了 PU.1 和 SWI/SNF ATP 依赖性染色质重塑复合物的共同功能作用,它们共同维持肿瘤细胞增殖所需的增强子景观,但在非肿瘤环境中也有关键作用。
{"title":"Pharmacologic Blockade of a Pioneer Transcription Factor.","authors":"Katerina Cermakova, H Courtney Hodges","doi":"10.1158/0008-5472.CAN-24-3957","DOIUrl":"10.1158/0008-5472.CAN-24-3957","url":null,"abstract":"<p><p>Cancers frequently co-opt lineage-specific transcription factors (TF) utilized in normal development to sustain proliferation. However, the effects of these TFs on tumor development depend considerably on where in the genome they bind. A new article by Taylor and colleagues expands on previously developed diamidine compounds that obstruct the DNA binding sites of the pioneer TF PU.1 (SPI1) in acute myeloid leukemia. Immobilization and sequencing of genomic DNA targeted by these compounds revealed that these inhibitors alter the genomic binding patterns of PU.1. The authors report that their strategy constrains the genomic binding preferences of PU.1, leading to redistribution of PU.1 to promoters and other gene-proximal regions with elevated guanine/cytosine content. In this study, we discuss recent developments for targeting PU.1 in hematologic malignancies. We also explore the shared functional roles of PU.1 and SWI/SNF ATP-dependent chromatin remodeling complexes, which not only work together to sustain the enhancer landscape needed for tumor cell proliferation but also play key roles in nontumor settings.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"4124-4125"},"PeriodicalIF":12.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142543886","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Correction: Single-Cell Transcriptomic Heterogeneity in Invasive Ductal and Lobular Breast Cancer Cells. 更正:浸润性乳腺导管癌和乳腺叶癌细胞的单细胞转录组异质性。
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-16 DOI: 10.1158/0008-5472.CAN-24-3940
Fangyuan Chen, Kai Ding, Nolan Priedigkeit, Ashuvinee Elangovan, Kevin M Levine, Neil Carleton, Laura Savariau, Jennifer M Atkinson, Steffi Oesterreich, Adrian V Lee
{"title":"Correction: Single-Cell Transcriptomic Heterogeneity in Invasive Ductal and Lobular Breast Cancer Cells.","authors":"Fangyuan Chen, Kai Ding, Nolan Priedigkeit, Ashuvinee Elangovan, Kevin M Levine, Neil Carleton, Laura Savariau, Jennifer M Atkinson, Steffi Oesterreich, Adrian V Lee","doi":"10.1158/0008-5472.CAN-24-3940","DOIUrl":"https://doi.org/10.1158/0008-5472.CAN-24-3940","url":null,"abstract":"","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"84 24","pages":"4298"},"PeriodicalIF":12.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142827493","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Induction of the TEAD Coactivator VGLL1 by Estrogen Receptor-Targeted Therapy Drives Resistance in Breast Cancer. 雌激素受体靶向疗法诱导 TEAD 协同激活因子 VGLL1 引发乳腺癌抗药性
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-16 DOI: 10.1158/0008-5472.CAN-24-0013
Carolina Gemma, Chun-Fui Lai, Anup K Singh, Antonino Belfiore, Neil Portman, Heloisa Z Milioli, Manikandan Periyasamy, Sara Raafat, Alyssa J Nicholls, Claire M Davies, Naina R Patel, Georgia M Simmons, Hailing Fan, Van T M Nguyen, Luca Magnani, Emad Rakha, Lesley-Ann Martin, Elgene Lim, R Charles Coombes, Giancarlo Pruneri, Laki Buluwela, Simak Ali

Resistance to endocrine therapies (ET) is common in estrogen receptor (ER)-positive breast cancer, and most relapsed patients die with ET-resistant disease. Although genetic mutations provide explanations for some relapses, mechanisms of resistance remain undefined in many cases. Drug-induced epigenetic reprogramming has been shown to provide possible routes to resistance. By analyzing histone H3 lysine 27 acetylation profiles and transcriptional reprogramming in models of ET resistance, we discovered that selective ER degraders, such as fulvestrant, promote expression of vestigial-like 1 (VGLL1), a coactivator for TEF-1 and AbaA domain (TEAD) transcription factors. VGLL1, acting via TEADs, promoted the expression of genes that drive the growth of fulvestrant-resistant breast cancer cells. Pharmacological disruption of VGLL1-TEAD4 interaction inhibited VGLL1/TEAD-induced transcriptional programs to prevent the growth of resistant cells. EGFR was among the VGLL1/TEAD-regulated genes, and VGLL1-directed EGFR upregulation sensitized fulvestrant-resistant breast cancer cells to EGFR inhibitors. Taken together, these findings identify VGLL1 as a transcriptional driver in ET resistance and advance therapeutic possibilities for relapsed ER+ breast cancer patients. Significance: Transcriptional reprogramming mediated by the upregulation of the TEAD coactivator VGLL1 confers resistance to estrogen receptor degraders in breast cancer but provides alternative therapeutic options for this clinically important patient group.

雌激素受体(ER)阳性乳腺癌对内分泌疗法(ET)的耐药性很常见,大多数复发患者都死于 ET 耐药性疾病。虽然基因突变可以解释某些复发,但在许多情况下,耐药机制仍未确定。药物诱导的表观遗传学重编程已被证明提供了可能的抗药性途径。通过分析ET耐药模型中组蛋白H3赖氨酸27乙酰化(H3K27ac)谱和转录重编程,我们发现选择性ER降解剂(SERDs),如氟维司群,可促进TEAD转录因子的共激活剂VGLL1的表达。VGLL1 通过 TEAD 起作用,促进了驱动氟维司群抗性乳腺癌细胞生长的基因的表达。药物破坏 VGLL1/TEAD4 的相互作用抑制了 VGLL1/TEAD 诱导的转录程序,从而阻止了耐药细胞的生长。表皮生长因子受体(EGFR)是 VGLL1/TEAD 调控基因之一,VGLL1 引导的表皮生长因子受体(EGFR)上调可使对氟维司群有耐药性的乳腺癌细胞对表皮生长因子受体(EGFR)抑制剂敏感。综上所述,这些发现确定了 VGLL1 是 ET 抗性的转录驱动因子,为复发性 ER+ 乳腺癌患者的治疗提供了更多可能性。
{"title":"Induction of the TEAD Coactivator VGLL1 by Estrogen Receptor-Targeted Therapy Drives Resistance in Breast Cancer.","authors":"Carolina Gemma, Chun-Fui Lai, Anup K Singh, Antonino Belfiore, Neil Portman, Heloisa Z Milioli, Manikandan Periyasamy, Sara Raafat, Alyssa J Nicholls, Claire M Davies, Naina R Patel, Georgia M Simmons, Hailing Fan, Van T M Nguyen, Luca Magnani, Emad Rakha, Lesley-Ann Martin, Elgene Lim, R Charles Coombes, Giancarlo Pruneri, Laki Buluwela, Simak Ali","doi":"10.1158/0008-5472.CAN-24-0013","DOIUrl":"10.1158/0008-5472.CAN-24-0013","url":null,"abstract":"<p><p>Resistance to endocrine therapies (ET) is common in estrogen receptor (ER)-positive breast cancer, and most relapsed patients die with ET-resistant disease. Although genetic mutations provide explanations for some relapses, mechanisms of resistance remain undefined in many cases. Drug-induced epigenetic reprogramming has been shown to provide possible routes to resistance. By analyzing histone H3 lysine 27 acetylation profiles and transcriptional reprogramming in models of ET resistance, we discovered that selective ER degraders, such as fulvestrant, promote expression of vestigial-like 1 (VGLL1), a coactivator for TEF-1 and AbaA domain (TEAD) transcription factors. VGLL1, acting via TEADs, promoted the expression of genes that drive the growth of fulvestrant-resistant breast cancer cells. Pharmacological disruption of VGLL1-TEAD4 interaction inhibited VGLL1/TEAD-induced transcriptional programs to prevent the growth of resistant cells. EGFR was among the VGLL1/TEAD-regulated genes, and VGLL1-directed EGFR upregulation sensitized fulvestrant-resistant breast cancer cells to EGFR inhibitors. Taken together, these findings identify VGLL1 as a transcriptional driver in ET resistance and advance therapeutic possibilities for relapsed ER+ breast cancer patients. Significance: Transcriptional reprogramming mediated by the upregulation of the TEAD coactivator VGLL1 confers resistance to estrogen receptor degraders in breast cancer but provides alternative therapeutic options for this clinically important patient group.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"4283-4297"},"PeriodicalIF":12.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7616691/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142364507","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
C-Reactive Protein Facilitates Premetastatic Niche Formation in the Lungs.
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-16 DOI: 10.1158/0008-5472.CAN-24-3394
Jonas Saal, Niklas Klümper, Michael Hölzel

C-reactive protein (CRP) has long been recognized as a marker of inflammation, but its evolving role in immunomodulation and cancer has increasingly been recognized. In recent years, multiple studies have explored CRP as a biomarker for prognosis and therapy response, particularly in the context of cancer immunotherapy. In this issue of Cancer Research, Feng and colleagues investigate the role of CRP in the development of lung metastasis. They provide evidence for a direct role of CRP acting together with commensal bacteria to instruct an immune-tolerant state of pulmonary macrophages through Fc gamma receptor IIb signaling. By suppressing immune surveillance in the lungs, CRP facilitates the formation of a premetastatic niche, allowing circulating tumor cells to establish metastases. See related article by Feng et al., p. 4184.

{"title":"C-Reactive Protein Facilitates Premetastatic Niche Formation in the Lungs.","authors":"Jonas Saal, Niklas Klümper, Michael Hölzel","doi":"10.1158/0008-5472.CAN-24-3394","DOIUrl":"https://doi.org/10.1158/0008-5472.CAN-24-3394","url":null,"abstract":"<p><p>C-reactive protein (CRP) has long been recognized as a marker of inflammation, but its evolving role in immunomodulation and cancer has increasingly been recognized. In recent years, multiple studies have explored CRP as a biomarker for prognosis and therapy response, particularly in the context of cancer immunotherapy. In this issue of Cancer Research, Feng and colleagues investigate the role of CRP in the development of lung metastasis. They provide evidence for a direct role of CRP acting together with commensal bacteria to instruct an immune-tolerant state of pulmonary macrophages through Fc gamma receptor IIb signaling. By suppressing immune surveillance in the lungs, CRP facilitates the formation of a premetastatic niche, allowing circulating tumor cells to establish metastases. See related article by Feng et al., p. 4184.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"84 24","pages":"4121-4123"},"PeriodicalIF":12.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142827496","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
期刊
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