首页 > 最新文献

Discover. Oncology最新文献

英文 中文
Upregulation of CD19 by low-dose chidamide promotes CAR T cells functionality in B-cell non-Hodgkin lymphoma.
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-24 DOI: 10.1007/s12672-025-01810-1
Ying Ni, Qun Zhang, Xiaochen Tang, Xiuchun Li, Shiguang Ye, Yan Lu, Aibin Liang, Ping Li

B-cell non-Hodgkin lymphoma (B-NHL) is a highly heterogeneous group of lymphopoietic malignancies that account for 85% to 90% of all non-Hodgkin lymphomas. In recent years, CD19 Chimeric antigen receptor T (CAR T) cell immunotherapy has significantly improved the cure rate of B-NHL patients, but there are still some patients who cannot achieve remission after treatment, or relapse after remission. Therefore, it is of great importance to overcome the drug resistance of CD19 CAR T cells after B-NHL treatment and reduce the recurrence rate of CD19 CAR T cells after B-NHL treatment. We found that low concentrations of chidamide did not enhance the ability of CD19 CAR T cells to kill B-NHL cells during and after preparation. B-NHL cells pretreated with chidamide were more likely to be killed by CD19 CAR T cells. CD19 CAR T cells secreted more cytokines (IL-2, TNF-α, and IFN-γ) after co-culture with B-NHL cells pretreated with chidamide. At the same time, the expression of CD19 on B-NHL cell surface was increased by chidamide. In vivo experiments showed that infusion of CD19 CAR T cells after chidamide bridging intervention can enhance the therapeutic effect of B-NHL and prolong the overall survival of mice. This study provides a new direction and theoretical foundation for CD19 CAR T cell therapy in B-NHL.

{"title":"Upregulation of CD19 by low-dose chidamide promotes CAR T cells functionality in B-cell non-Hodgkin lymphoma.","authors":"Ying Ni, Qun Zhang, Xiaochen Tang, Xiuchun Li, Shiguang Ye, Yan Lu, Aibin Liang, Ping Li","doi":"10.1007/s12672-025-01810-1","DOIUrl":"https://doi.org/10.1007/s12672-025-01810-1","url":null,"abstract":"<p><p>B-cell non-Hodgkin lymphoma (B-NHL) is a highly heterogeneous group of lymphopoietic malignancies that account for 85% to 90% of all non-Hodgkin lymphomas. In recent years, CD19 Chimeric antigen receptor T (CAR T) cell immunotherapy has significantly improved the cure rate of B-NHL patients, but there are still some patients who cannot achieve remission after treatment, or relapse after remission. Therefore, it is of great importance to overcome the drug resistance of CD19 CAR T cells after B-NHL treatment and reduce the recurrence rate of CD19 CAR T cells after B-NHL treatment. We found that low concentrations of chidamide did not enhance the ability of CD19 CAR T cells to kill B-NHL cells during and after preparation. B-NHL cells pretreated with chidamide were more likely to be killed by CD19 CAR T cells. CD19 CAR T cells secreted more cytokines (IL-2, TNF-α, and IFN-γ) after co-culture with B-NHL cells pretreated with chidamide. At the same time, the expression of CD19 on B-NHL cell surface was increased by chidamide. In vivo experiments showed that infusion of CD19 CAR T cells after chidamide bridging intervention can enhance the therapeutic effect of B-NHL and prolong the overall survival of mice. This study provides a new direction and theoretical foundation for CD19 CAR T cell therapy in B-NHL.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"84"},"PeriodicalIF":2.8,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of PANoptosis associated lncRNAs associated with clinical prognosis and immune infiltration microenvironment in colon adenocarcinoma.
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-24 DOI: 10.1007/s12672-025-01838-3
Yangyang Wang, Shihui Zhao, Songtao Du, Tianyi Xia, Liqiang Song, Mingyu Xia, Bomiao Zhang

Early diagnosis and disease management based on risk stratification have a very positive impact on colon adenocarcinoma (COAD) prognosis. It is of positive significance to further explore risk stratification of COAD patients and identify predictive molecular biomarkers. PANoptosis is defined as a form of inflammatory cell death regulated by PANoptosome, with common features of pyroptosis, apoptosis and necroptosis. The role of PANoptosis in COAD has not been fully studied. In this study, we analyzed significant differences in the expression of PANoptosis-related gene (PRG) features in COAD. Subsequently, the PANoptosis associated lncRNAs (PALs) associated with PRGs were analyzed by LASSO algorithm and multivariate Cox analysis, and PALs related to the prognosis of COAD were selected. Based on the expression patterns of prognostic PAL features, we performed unsupervised consensus cluster analysis to categorize COAD samples into distinct PAL molecular subtypes and investigate their associated immune infiltration characteristics. We subsequently constructed PAL score model based on prognostic characteristics and verified its independent prognostic value for COAD. The nomogram diagnostic model was established to confirm the prognostic value of PAL scoring system again. Pathway enrichment analysis, somatic mutation profiling, and drug sensitivity analysis were employed to comprehensively assess the clinical value of the PAL score. Additionally, qRT-PCR was used to further validate the abnormal expression of the selected targets in COAD. Our results provide a new idea for clinical risk stratification and new evidence for the role of PANoptosis in COAD.

{"title":"Identification of PANoptosis associated lncRNAs associated with clinical prognosis and immune infiltration microenvironment in colon adenocarcinoma.","authors":"Yangyang Wang, Shihui Zhao, Songtao Du, Tianyi Xia, Liqiang Song, Mingyu Xia, Bomiao Zhang","doi":"10.1007/s12672-025-01838-3","DOIUrl":"https://doi.org/10.1007/s12672-025-01838-3","url":null,"abstract":"<p><p>Early diagnosis and disease management based on risk stratification have a very positive impact on colon adenocarcinoma (COAD) prognosis. It is of positive significance to further explore risk stratification of COAD patients and identify predictive molecular biomarkers. PANoptosis is defined as a form of inflammatory cell death regulated by PANoptosome, with common features of pyroptosis, apoptosis and necroptosis. The role of PANoptosis in COAD has not been fully studied. In this study, we analyzed significant differences in the expression of PANoptosis-related gene (PRG) features in COAD. Subsequently, the PANoptosis associated lncRNAs (PALs) associated with PRGs were analyzed by LASSO algorithm and multivariate Cox analysis, and PALs related to the prognosis of COAD were selected. Based on the expression patterns of prognostic PAL features, we performed unsupervised consensus cluster analysis to categorize COAD samples into distinct PAL molecular subtypes and investigate their associated immune infiltration characteristics. We subsequently constructed PAL score model based on prognostic characteristics and verified its independent prognostic value for COAD. The nomogram diagnostic model was established to confirm the prognostic value of PAL scoring system again. Pathway enrichment analysis, somatic mutation profiling, and drug sensitivity analysis were employed to comprehensively assess the clinical value of the PAL score. Additionally, qRT-PCR was used to further validate the abnormal expression of the selected targets in COAD. Our results provide a new idea for clinical risk stratification and new evidence for the role of PANoptosis in COAD.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"83"},"PeriodicalIF":2.8,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143032585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Single-cell RNA-seq analysis reveals microenvironmental infiltration of myeloid cells and pancreatic prognostic markers in PDAC.
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-23 DOI: 10.1007/s12672-025-01830-x
Yanying Fan, Lili Wu, Xinyu Qiu, Han Shi, Longhang Wu, Juan Lin, Jie Lin, Tianhong Teng

Background: Pancreatic ductal adenocarcinoma (PDAC) has a heterogeneous make-up of myeloid cells that influences the therapeutic response and prognosis. However, understanding the myeloid cell at both a genetic and cellular level remains a significant challenge.

Methods: Single-cell RNA sequencing (scRNA-seq) data were downloaded from t the Tumor Immune Single-cell Hub and gene expression data were retrieved from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database. Gene set variation analysis (GSVA) was used to estimate the relative proportions of each cell type based on the signatures identified by scRNA-seq or previous literature. Cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) was performed to evaluate the abundance of immune infiltrating cells. For further analysis, LASSO and Cox analyses were used to construct a risk model using univariate Cox regression.

Results: Using the scRNA-seq dataset, we identified 7 clusters of myeloid cells, and these clusters were assigned a cell type based on their marker genes. In addition, the results of the CellChat analysis and SCENIC analysis indicate that TAM-spp1 cells may promote the migration of pancreatic tumor cells on different levels. Moreover, the TAM-spp1 cell is most closely related to poor prognoses. An 8-gene risk model was constructed by using univariate Cox and LASSO analyses. In the GEO cohorts, this risk model demonstrated excellent predictive abilities for prognosis. Further, patients with high-risk scores had a lower likelihood of benefiting from immunotherapy.

Conclusion: Using bulk RNA-seq and single-cell RNA-seq, we analyzed myeloid heterogeneity at the single-cell level, and we developed an 8-gene model that predicts survival outcomes and immunotherapy response in PADC.

{"title":"Single-cell RNA-seq analysis reveals microenvironmental infiltration of myeloid cells and pancreatic prognostic markers in PDAC.","authors":"Yanying Fan, Lili Wu, Xinyu Qiu, Han Shi, Longhang Wu, Juan Lin, Jie Lin, Tianhong Teng","doi":"10.1007/s12672-025-01830-x","DOIUrl":"10.1007/s12672-025-01830-x","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic ductal adenocarcinoma (PDAC) has a heterogeneous make-up of myeloid cells that influences the therapeutic response and prognosis. However, understanding the myeloid cell at both a genetic and cellular level remains a significant challenge.</p><p><strong>Methods: </strong>Single-cell RNA sequencing (scRNA-seq) data were downloaded from t the Tumor Immune Single-cell Hub and gene expression data were retrieved from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database. Gene set variation analysis (GSVA) was used to estimate the relative proportions of each cell type based on the signatures identified by scRNA-seq or previous literature. Cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) was performed to evaluate the abundance of immune infiltrating cells. For further analysis, LASSO and Cox analyses were used to construct a risk model using univariate Cox regression.</p><p><strong>Results: </strong>Using the scRNA-seq dataset, we identified 7 clusters of myeloid cells, and these clusters were assigned a cell type based on their marker genes. In addition, the results of the CellChat analysis and SCENIC analysis indicate that TAM-spp1 cells may promote the migration of pancreatic tumor cells on different levels. Moreover, the TAM-spp1 cell is most closely related to poor prognoses. An 8-gene risk model was constructed by using univariate Cox and LASSO analyses. In the GEO cohorts, this risk model demonstrated excellent predictive abilities for prognosis. Further, patients with high-risk scores had a lower likelihood of benefiting from immunotherapy.</p><p><strong>Conclusion: </strong>Using bulk RNA-seq and single-cell RNA-seq, we analyzed myeloid heterogeneity at the single-cell level, and we developed an 8-gene model that predicts survival outcomes and immunotherapy response in PADC.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"81"},"PeriodicalIF":2.8,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022331","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Association between serum levels of 12 different cytokines and short-term efficacy of chemoradiotherapy in esophageal squamous cell carcinoma.
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-23 DOI: 10.1007/s12672-025-01823-w
Yaping Zhang, Qiufeng Qi, Ming Zhu, Yun Peng, Yanqing Bao, Jun Liu, Yanzhi Bi, Min Xiao, Shaohua Chi, Yongping Liu

Background: Esophageal squamous cell carcinoma (ESCC) has a poor prognosis, with chemoradiotherapy (CRT) being a key treatment method. This study focused on circulating cytokines as potential predictors of treatment response and prognosis in patients with ESCC.

Materials and methods: Serum samples were collected from 36 ESCC patients, and 12 different cytokines were quantified using a multiplex immunofluorescence assay. We used non-parametric Wilcoxon unpaired rank tests to examine the relationship between cytokine concentrations and clinical outcomes. The duration of progression-free survival was assessed through imaging studies and telephone follow-ups. Kaplan-Meier survival plots, analyzed with the log-rank test, were utilized to depict survival trends.

Results: Pre-treatment serum IL-8 levels were significantly elevated in patients with lymphoid metastases (p = 0.036). Lower initial levels of IL-8 and IL-1β were observed in patients with partial response group compared to those with stable disease (p = 0.002, p = 0.01). Elevated baseline levels of IL-8 and interferon-gamma (IFN-γ) were correlated with a poorer prognosis. Higher levels of IL-5 and IFN-γ levels following therapy were associated with worse outcomes.

Conclusions: Our findings indicate that IL-8, IL-1β, IL-5, and IFN-γ may serve as potential biomarkers for treatment efficacy and prognosis in ESCC. Patients with low levels of IL-8 and IL-1β demonstrate a favorable response to CRT. Elevated serum levels of IL-8, IL-1β, IFN-γ, and IL-5 may predict poorer clinical outcomes.

{"title":"Association between serum levels of 12 different cytokines and short-term efficacy of chemoradiotherapy in esophageal squamous cell carcinoma.","authors":"Yaping Zhang, Qiufeng Qi, Ming Zhu, Yun Peng, Yanqing Bao, Jun Liu, Yanzhi Bi, Min Xiao, Shaohua Chi, Yongping Liu","doi":"10.1007/s12672-025-01823-w","DOIUrl":"10.1007/s12672-025-01823-w","url":null,"abstract":"<p><strong>Background: </strong>Esophageal squamous cell carcinoma (ESCC) has a poor prognosis, with chemoradiotherapy (CRT) being a key treatment method. This study focused on circulating cytokines as potential predictors of treatment response and prognosis in patients with ESCC.</p><p><strong>Materials and methods: </strong>Serum samples were collected from 36 ESCC patients, and 12 different cytokines were quantified using a multiplex immunofluorescence assay. We used non-parametric Wilcoxon unpaired rank tests to examine the relationship between cytokine concentrations and clinical outcomes. The duration of progression-free survival was assessed through imaging studies and telephone follow-ups. Kaplan-Meier survival plots, analyzed with the log-rank test, were utilized to depict survival trends.</p><p><strong>Results: </strong>Pre-treatment serum IL-8 levels were significantly elevated in patients with lymphoid metastases (p = 0.036). Lower initial levels of IL-8 and IL-1β were observed in patients with partial response group compared to those with stable disease (p = 0.002, p = 0.01). Elevated baseline levels of IL-8 and interferon-gamma (IFN-γ) were correlated with a poorer prognosis. Higher levels of IL-5 and IFN-γ levels following therapy were associated with worse outcomes.</p><p><strong>Conclusions: </strong>Our findings indicate that IL-8, IL-1β, IL-5, and IFN-γ may serve as potential biomarkers for treatment efficacy and prognosis in ESCC. Patients with low levels of IL-8 and IL-1β demonstrate a favorable response to CRT. Elevated serum levels of IL-8, IL-1β, IFN-γ, and IL-5 may predict poorer clinical outcomes.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"80"},"PeriodicalIF":2.8,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754553/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Survival outcomes of surgery and adjuvant chemotherapy in early-stage small cell and large cell lung cancer: a novel focus on tumors less than 1 cm.
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-23 DOI: 10.1007/s12672-025-01777-z
Jorge Raul Vazquez-Urrutia, Junjia Zhu, Shinkichi Takamori, Max Greenberg, Priyanka Bhatia, Takefumi Komiya

Background: The role of adjuvant chemotherapy in early-stage small cell lung cancer (SCLC) and large cell neuroendocrine carcinoma (LCNEC) remains unclear, particularly for small tumors. This study assesses the survival benefits of adjuvant chemotherapy after surgical resection with a novel focus on tumors less than 1 cm.

Materials and methods: Data from the National Cancer Database (NCDB) was extracted for patients with SCLC (n = 11,962) and LCNEC (n = 6821) who underwent surgical resection between 2004 and 2020. Exclusion criteria were limited survival (< 30 days), positive lymph nodes, distant metastases, large tumors (> 5 cm), residual microscopic disease, and neoadjuvant therapy. The primary outcome was overall survival (OS) from diagnosis, which was evaluated using Kaplan-Meier methods and multivariate Cox regression analyses. A propensity score matching (PSM) analysis was performed to compare outcomes in patients with SCLC and tumors ≤ 1 cm who received adjuvant chemotherapy versus surgery alone.

Results: The study involved 4114 SCLC and 3954 LCNEC patients. Adjuvant chemotherapy was associated with a significant increase median OS in both SCLC (6.26 vs. 4.18 years; p < 0.001) and LCNEC (7.02 years vs. 4.89 years; p < 0.001), while also being an independent predictor of better OS in SCLC (HR: 0.74) and LCNEC (HR: 0.75) (p < 0.001). The benefit was more noticeable in tumors ≤ 1 cm, showing a significant OS increase after PSM (median OS 7.34 vs. 5.02 years; p = 0.0048).

Conclusion: Adjuvant chemotherapy after surgery is associated with improved overall survival in stage I SCLC and LCNEC, particularly in SCLC tumors of 1 cm or less.

{"title":"Survival outcomes of surgery and adjuvant chemotherapy in early-stage small cell and large cell lung cancer: a novel focus on tumors less than 1 cm.","authors":"Jorge Raul Vazquez-Urrutia, Junjia Zhu, Shinkichi Takamori, Max Greenberg, Priyanka Bhatia, Takefumi Komiya","doi":"10.1007/s12672-025-01777-z","DOIUrl":"10.1007/s12672-025-01777-z","url":null,"abstract":"<p><strong>Background: </strong>The role of adjuvant chemotherapy in early-stage small cell lung cancer (SCLC) and large cell neuroendocrine carcinoma (LCNEC) remains unclear, particularly for small tumors. This study assesses the survival benefits of adjuvant chemotherapy after surgical resection with a novel focus on tumors less than 1 cm.</p><p><strong>Materials and methods: </strong>Data from the National Cancer Database (NCDB) was extracted for patients with SCLC (n = 11,962) and LCNEC (n = 6821) who underwent surgical resection between 2004 and 2020. Exclusion criteria were limited survival (< 30 days), positive lymph nodes, distant metastases, large tumors (> 5 cm), residual microscopic disease, and neoadjuvant therapy. The primary outcome was overall survival (OS) from diagnosis, which was evaluated using Kaplan-Meier methods and multivariate Cox regression analyses. A propensity score matching (PSM) analysis was performed to compare outcomes in patients with SCLC and tumors ≤ 1 cm who received adjuvant chemotherapy versus surgery alone.</p><p><strong>Results: </strong>The study involved 4114 SCLC and 3954 LCNEC patients. Adjuvant chemotherapy was associated with a significant increase median OS in both SCLC (6.26 vs. 4.18 years; p < 0.001) and LCNEC (7.02 years vs. 4.89 years; p < 0.001), while also being an independent predictor of better OS in SCLC (HR: 0.74) and LCNEC (HR: 0.75) (p < 0.001). The benefit was more noticeable in tumors ≤ 1 cm, showing a significant OS increase after PSM (median OS 7.34 vs. 5.02 years; p = 0.0048).</p><p><strong>Conclusion: </strong>Adjuvant chemotherapy after surgery is associated with improved overall survival in stage I SCLC and LCNEC, particularly in SCLC tumors of 1 cm or less.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"82"},"PeriodicalIF":2.8,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022332","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Convergence of nanotechnology and artificial intelligence in the fight against liver cancer: a comprehensive review. 纳米技术与人工智能在对抗肝癌中的结合:综合综述。
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-22 DOI: 10.1007/s12672-025-01821-y
Manjusha Bhange, Darshan Telange

Liver cancer is one of the most challenging malignancies, often associated with poor prognosis and limited treatment options. Recent advancements in nanotechnology and artificial intelligence (AI) have opened new frontiers in the fight against this disease. Nanotechnology enables precise, targeted drug delivery, enhancing the efficacy of therapeutics while minimizing off-target effects. Simultaneously, AI contributes to improved diagnostic accuracy, predictive modeling, and the development of personalized treatment strategies. This review explores the convergence of nanotechnology and AI in liver cancer treatment, evaluating current progress, identifying existing research gaps, and discussing future directions. We highlight how AI-powered algorithms can optimize nanocarrier design, facilitate real-time monitoring of treatment efficacy, and enhance clinical decision-making. By integrating AI with nanotechnology, clinicians can achieve more accurate patient stratification and treatment personalization, ultimately improving patient outcomes. This convergence holds significant promise for transforming liver cancer therapy into a more precise, individualized, and efficient process. However, data privacy, regulatory hurdles, and the need for large-scale clinical validation remain. Addressing these issues will be essential to fully realizing the potential of these technologies in oncology.

肝癌是最具挑战性的恶性肿瘤之一,通常与预后不良和治疗选择有限有关。纳米技术和人工智能(AI)的最新进展为防治这种疾病开辟了新的领域。纳米技术能够精确、有针对性地给药,提高治疗效果,同时最大限度地减少脱靶效应。同时,人工智能有助于提高诊断准确性、预测建模和制定个性化治疗策略。这篇综述探讨了纳米技术和人工智能在肝癌治疗中的融合,评估了目前的进展,确定了现有的研究差距,并讨论了未来的方向。我们重点介绍了人工智能算法如何优化纳米载体设计,促进治疗效果的实时监测,并增强临床决策。通过将人工智能与纳米技术相结合,临床医生可以实现更准确的患者分层和个性化治疗,最终改善患者的治疗效果。这种融合为肝癌治疗转变为更精确、个性化和高效的过程带来了重大希望。然而,数据隐私、监管障碍和大规模临床验证的需求仍然存在。解决这些问题对于充分发挥这些技术在肿瘤学中的潜力至关重要。
{"title":"Convergence of nanotechnology and artificial intelligence in the fight against liver cancer: a comprehensive review.","authors":"Manjusha Bhange, Darshan Telange","doi":"10.1007/s12672-025-01821-y","DOIUrl":"10.1007/s12672-025-01821-y","url":null,"abstract":"<p><p>Liver cancer is one of the most challenging malignancies, often associated with poor prognosis and limited treatment options. Recent advancements in nanotechnology and artificial intelligence (AI) have opened new frontiers in the fight against this disease. Nanotechnology enables precise, targeted drug delivery, enhancing the efficacy of therapeutics while minimizing off-target effects. Simultaneously, AI contributes to improved diagnostic accuracy, predictive modeling, and the development of personalized treatment strategies. This review explores the convergence of nanotechnology and AI in liver cancer treatment, evaluating current progress, identifying existing research gaps, and discussing future directions. We highlight how AI-powered algorithms can optimize nanocarrier design, facilitate real-time monitoring of treatment efficacy, and enhance clinical decision-making. By integrating AI with nanotechnology, clinicians can achieve more accurate patient stratification and treatment personalization, ultimately improving patient outcomes. This convergence holds significant promise for transforming liver cancer therapy into a more precise, individualized, and efficient process. However, data privacy, regulatory hurdles, and the need for large-scale clinical validation remain. Addressing these issues will be essential to fully realizing the potential of these technologies in oncology.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"77"},"PeriodicalIF":2.8,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754566/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143001694","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
A Golgi apparatus‑related subtype and risk signature predicts prognosis and evaluates immunotherapy response in gastric cancer. 高尔基体相关亚型和风险标志预测胃癌的预后和评估免疫治疗反应。
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-22 DOI: 10.1007/s12672-025-01827-6
Ruyue Chen, Zengwu Yao, Lixin Jiang, Jinchen Hu

Background: Gastric cancer (GC) remains a significant health burden, calling for the discovery of novel biomarkers. Golgi apparatus, a crucial cellular organelle involved in tumorigenesis, remains underexplored in GC research. A comprehensive understanding of its role and associated mechanisms is urgently needed.

Materials and methods: Utilizing the TCGA-STAD dataset as the training cohort and GSE84433 as the validation cohort, we explored potential associations between Golgi apparatus-related genes (GARG) and GC clinical risk. We aimed to decipher the prognostic significance and underlying biological mechanisms of these genes via consistent clustering, differential expression analysis, enrichment analyses, and immune infiltration profiling. To assess the relationship between risk stratification and survival outcomes, drug sensitivity, and immune infiltration, we developed the Golgi Apparatus-Related Risk Signature (GARRS). The reliability of GARRS was further corroborated using immunohistochemical staining.

Results: Consensus clustering based on 17 GARG identified two patient subgroups, C1 and C2, exhibiting differential survival, immune scores, and immune cell infiltration. We developed a GARRS using Cox-Lasso regression analysis, accurately stratifying patients into high- and low-risk groups. GARRS' validity was confirmed in the validation set and immunohistochemical staining. Our findings underline the Golgi apparatus' significance in the GC immune microenvironment and GARRS' utility in predicting GC survival outcomes.

Conclusion: This study underscores the association between Golgi apparatus subtypes and GC immunotumor microenvironment. GARRS, validated for its prognostic, immune infiltration, and drug sensitivity predictive abilities, offers new insights into gastric cancer treatment strategies.

背景:胃癌(GC)仍然是一个重要的健康负担,需要发现新的生物标志物。高尔基体是参与肿瘤发生的重要细胞器,在GC研究中尚未得到充分的探索。迫切需要全面了解其作用和相关机制。材料和方法:利用TCGA-STAD数据集作为训练队列,GSE84433作为验证队列,我们探讨了高尔基器械相关基因(GARG)与GC临床风险之间的潜在关联。我们旨在通过一致的聚类分析、差异表达分析、富集分析和免疫浸润分析来破译这些基因的预后意义和潜在的生物学机制。为了评估风险分层与生存结果、药物敏感性和免疫浸润之间的关系,我们开发了高尔基器械相关风险特征(GARRS)。免疫组织化学染色进一步证实了GARRS的可靠性。结果:基于17 GARG的共识聚类确定了两个患者亚组,C1和C2,表现出不同的生存、免疫评分和免疫细胞浸润。我们使用Cox-Lasso回归分析开发了GARRS,准确地将患者分为高危组和低危组。验证集和免疫组化染色证实了GARRS的有效性。我们的研究结果强调了高尔基体在胃癌免疫微环境中的重要性以及GARRS在预测胃癌生存结果方面的实用性。结论:本研究强调高尔基体亚型与GC免疫肿瘤微环境的相关性。GARRS因其预后、免疫浸润和药物敏感性预测能力而得到验证,为胃癌治疗策略提供了新的见解。
{"title":"A Golgi apparatus‑related subtype and risk signature predicts prognosis and evaluates immunotherapy response in gastric cancer.","authors":"Ruyue Chen, Zengwu Yao, Lixin Jiang, Jinchen Hu","doi":"10.1007/s12672-025-01827-6","DOIUrl":"10.1007/s12672-025-01827-6","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) remains a significant health burden, calling for the discovery of novel biomarkers. Golgi apparatus, a crucial cellular organelle involved in tumorigenesis, remains underexplored in GC research. A comprehensive understanding of its role and associated mechanisms is urgently needed.</p><p><strong>Materials and methods: </strong>Utilizing the TCGA-STAD dataset as the training cohort and GSE84433 as the validation cohort, we explored potential associations between Golgi apparatus-related genes (GARG) and GC clinical risk. We aimed to decipher the prognostic significance and underlying biological mechanisms of these genes via consistent clustering, differential expression analysis, enrichment analyses, and immune infiltration profiling. To assess the relationship between risk stratification and survival outcomes, drug sensitivity, and immune infiltration, we developed the Golgi Apparatus-Related Risk Signature (GARRS). The reliability of GARRS was further corroborated using immunohistochemical staining.</p><p><strong>Results: </strong>Consensus clustering based on 17 GARG identified two patient subgroups, C1 and C2, exhibiting differential survival, immune scores, and immune cell infiltration. We developed a GARRS using Cox-Lasso regression analysis, accurately stratifying patients into high- and low-risk groups. GARRS' validity was confirmed in the validation set and immunohistochemical staining. Our findings underline the Golgi apparatus' significance in the GC immune microenvironment and GARRS' utility in predicting GC survival outcomes.</p><p><strong>Conclusion: </strong>This study underscores the association between Golgi apparatus subtypes and GC immunotumor microenvironment. GARRS, validated for its prognostic, immune infiltration, and drug sensitivity predictive abilities, offers new insights into gastric cancer treatment strategies.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"76"},"PeriodicalIF":2.8,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754555/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143001339","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Bibliometric and visual analysis of chronic stress in cancer research from 2014 to 2024.
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-22 DOI: 10.1007/s12672-025-01744-8
Zhuheng Wei, Anxia Li, Ling Su, Bo Zhang, Yuanyuan Yan

Objective: In today's fast-paced society, stress has become a widespread phenomenon, garnering increasing attention for its impact on cancer. This study aims to investigate the current status and research hotspots of chronic stress in cancer research from 2014 to 2024, with the goal of providing valuable insights for future studies.

Methods: We retrieved 618 articles published between 2014 and 2024 from the Web of Science database and analyzed them using R software, VOSviewer, and CiteSpace.

Results: There is an overall upward trend in chronic stress-related cancer research, with China leading in publications, followed by the United States, India, Australia, and Italy. The journal most cited is Brain Behavior and Immunity. Key themes identified include 'inflammation', 'breast cancer', 'anxiety', 'psychological stress', and 'oxidative stress'. The primary focus of the research is the impact of chronic stress on various cancer types, the underlying molecular mechanisms, and the implications of chronic stress-related treatments on cancer outcomes.

Conclusion: Chronic stress is increasingly recognized as a Carcinogenic factors. This study provides a comprehensive analysis of chronic stress-related cancer research from 2014 to 2024, offering valuable guidance for future research in this field.

{"title":"Bibliometric and visual analysis of chronic stress in cancer research from 2014 to 2024.","authors":"Zhuheng Wei, Anxia Li, Ling Su, Bo Zhang, Yuanyuan Yan","doi":"10.1007/s12672-025-01744-8","DOIUrl":"10.1007/s12672-025-01744-8","url":null,"abstract":"<p><strong>Objective: </strong>In today's fast-paced society, stress has become a widespread phenomenon, garnering increasing attention for its impact on cancer. This study aims to investigate the current status and research hotspots of chronic stress in cancer research from 2014 to 2024, with the goal of providing valuable insights for future studies.</p><p><strong>Methods: </strong>We retrieved 618 articles published between 2014 and 2024 from the Web of Science database and analyzed them using R software, VOSviewer, and CiteSpace.</p><p><strong>Results: </strong>There is an overall upward trend in chronic stress-related cancer research, with China leading in publications, followed by the United States, India, Australia, and Italy. The journal most cited is Brain Behavior and Immunity. Key themes identified include 'inflammation', 'breast cancer', 'anxiety', 'psychological stress', and 'oxidative stress'. The primary focus of the research is the impact of chronic stress on various cancer types, the underlying molecular mechanisms, and the implications of chronic stress-related treatments on cancer outcomes.</p><p><strong>Conclusion: </strong>Chronic stress is increasingly recognized as a Carcinogenic factors. This study provides a comprehensive analysis of chronic stress-related cancer research from 2014 to 2024, offering valuable guidance for future research in this field.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"79"},"PeriodicalIF":2.8,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754581/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Identification of KRT16 and ANXA10 as cell cycle regulation genes for lung adenocarcinoma based on self-transcriptome sequencing of surgical samples and TCGA public data mining. 基于手术样本自转录组测序和TCGA公开数据挖掘的肺腺癌细胞周期调控基因KRT16和ANXA10的鉴定
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-22 DOI: 10.1007/s12672-024-01707-5
Wen-Jian Liu, Jia-Pan Shen, Ren-Quan Zhang, Xiao-Yun Fan

Aim: This study aimed to identify the genes associated with the development of lung adenocarcinoma (LUAD) and potential therapeutic targets.

Methods: Differentially expressed genes (DEGs) were identified by self-transcriptome sequencing of tumor tissues and paracancerous tissues resected during surgery and combined with The Cancer Genome Atlas (TCGA) data to screen for the genes associated with LUAD prognosis. The expression was validated at mRNA and protein levels, and the gene knockdown was used to examine the impact and underlying mechanisms on lung cancer cells.

Results: A total of 227 DEGs were identified by transcriptome sequencing, and the 20 DEGs with the most significant differences were used for co-analysis with TCGA data. The findings suggested that KRT16 and ANXA10 might have an important role in the development of LUAD after validating the mRNA and protein expression levels at the cellular level. The knockdown of KRT16 and ANXA10 inhibited the proliferation of lung cancer cells, and the cell cycle was blocked in the G1 phase. The expression of the G1/S-phase cell cycle checkpoint-related proteins cyclin D1 and cyclin E was inhibited by KRT16 and ANXA10 knockdown, respectively. The tumor formation ability decreased after KRT16 or ANXA10 knockdown in vivo.

Conclusions: KRT16 and ANXA10 are potential genes regulating the development of LUAD. Also, they may be potential targets for the targeted therapy of LUAD by inhibiting the proliferation of lung cancer cells and blocking the cell cycle by affecting key protein expression levels at cell cycle checkpoints.

目的:本研究旨在鉴定与肺腺癌(LUAD)发展相关的基因和潜在的治疗靶点。方法:通过手术切除的肿瘤组织和癌旁组织的自转录组测序,鉴定差异表达基因(differential expressed genes, DEGs),并结合癌症基因组图谱(Cancer Genome Atlas, TCGA)数据,筛选与LUAD预后相关的基因。在mRNA和蛋白水平上验证了表达,并使用基因敲低来检测对肺癌细胞的影响及其潜在机制。结果:通过转录组测序共鉴定出227个基因片段,将差异最大的20个基因片段与TCGA数据进行共分析。在细胞水平上验证了KRT16和ANXA10 mRNA和蛋白的表达水平,提示KRT16和ANXA10可能在LUAD的发生发展中发挥重要作用。KRT16和ANXA10的下调抑制了肺癌细胞的增殖,细胞周期被阻断在G1期。下调KRT16和ANXA10分别抑制G1/ s期细胞周期检查点相关蛋白cyclin D1和cyclin E的表达。体内敲低KRT16或ANXA10后,肿瘤形成能力下降。结论:KRT16和ANXA10是调节LUAD发生的潜在基因。此外,它们可能是LUAD靶向治疗的潜在靶点,通过影响细胞周期检查点的关键蛋白表达水平来抑制肺癌细胞的增殖和阻断细胞周期。
{"title":"Identification of KRT16 and ANXA10 as cell cycle regulation genes for lung adenocarcinoma based on self-transcriptome sequencing of surgical samples and TCGA public data mining.","authors":"Wen-Jian Liu, Jia-Pan Shen, Ren-Quan Zhang, Xiao-Yun Fan","doi":"10.1007/s12672-024-01707-5","DOIUrl":"10.1007/s12672-024-01707-5","url":null,"abstract":"<p><strong>Aim: </strong>This study aimed to identify the genes associated with the development of lung adenocarcinoma (LUAD) and potential therapeutic targets.</p><p><strong>Methods: </strong>Differentially expressed genes (DEGs) were identified by self-transcriptome sequencing of tumor tissues and paracancerous tissues resected during surgery and combined with The Cancer Genome Atlas (TCGA) data to screen for the genes associated with LUAD prognosis. The expression was validated at mRNA and protein levels, and the gene knockdown was used to examine the impact and underlying mechanisms on lung cancer cells.</p><p><strong>Results: </strong>A total of 227 DEGs were identified by transcriptome sequencing, and the 20 DEGs with the most significant differences were used for co-analysis with TCGA data. The findings suggested that KRT16 and ANXA10 might have an important role in the development of LUAD after validating the mRNA and protein expression levels at the cellular level. The knockdown of KRT16 and ANXA10 inhibited the proliferation of lung cancer cells, and the cell cycle was blocked in the G1 phase. The expression of the G1/S-phase cell cycle checkpoint-related proteins cyclin D1 and cyclin E was inhibited by KRT16 and ANXA10 knockdown, respectively. The tumor formation ability decreased after KRT16 or ANXA10 knockdown in vivo.</p><p><strong>Conclusions: </strong>KRT16 and ANXA10 are potential genes regulating the development of LUAD. Also, they may be potential targets for the targeted therapy of LUAD by inhibiting the proliferation of lung cancer cells and blocking the cell cycle by affecting key protein expression levels at cell cycle checkpoints.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"78"},"PeriodicalIF":2.8,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754560/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143001730","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Maytansinoids in cancer therapy: advancements in antibody-drug conjugates and nanotechnology-enhanced drug delivery systems. 癌症治疗中的美坦素:抗体-药物偶联物和纳米技术增强的药物传递系统的进展。
IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM Pub Date : 2025-01-21 DOI: 10.1007/s12672-025-01820-z
Matteo Perra, Ines Castangia, Matteo Aroffu, Federica Fulgheri, Rita Abi-Rached, Maria Letizia Manca, Hernán Cortés, María Luisa Del Prado-Audelo, Carla Nomura-Contreras, Alejandra Romero-Montero, Dietrich Büsselberg, Gerardo Leyva-Gómez, Javad Sharifi-Rad, Daniela Calina

Cancer remains the second leading cause of death globally, driving the need for innovative therapies. Among natural compounds, maytansinoids have shown significant promise, contributing to nearly 25% of recently approved anticancer drugs. Despite their potential, early clinical trials faced challenges due to severe side effects, prompting advancements in delivery systems such as antibody-maytansinoid conjugates (AMCs). This review highlights the anticancer activity of maytansinoids, with a focus on AMCs designed to target cancer cells specifically. Preclinical and clinical studies show that AMCs, including FDA-approved drugs like Kadcyla and Elahere, effectively inhibit tumor growth while reducing systemic toxicity. Key developments include improved synthesis methods, linker chemistry and payload design. Ongoing research aims to enhance the safety and efficacy of AMCs, integrate nanotechnology for drug delivery, and identify novel therapeutic targets. These advancements hold potential to transform maytansinoid-based cancer treatments in the future.

癌症仍然是全球第二大死亡原因,推动了对创新疗法的需求。在天然化合物中,美黄素类化合物已显示出巨大的前景,占最近批准的抗癌药物的近25%。尽管具有潜力,但由于严重的副作用,早期临床试验面临挑战,促使诸如抗体-美坦素缀合物(AMCs)等递送系统取得进展。本文综述了美黄素类化合物的抗癌活性,重点介绍了针对癌细胞设计的AMCs。临床前和临床研究表明,包括fda批准的Kadcyla和Elahere等药物在内的amc可以有效抑制肿瘤生长,同时降低全身毒性。关键的发展包括改进的合成方法、连接剂化学和有效载荷设计。目前正在进行的研究旨在提高amc的安全性和有效性,整合纳米技术用于药物传递,并确定新的治疗靶点。这些进步有可能在未来改变以美坦素为基础的癌症治疗。
{"title":"Maytansinoids in cancer therapy: advancements in antibody-drug conjugates and nanotechnology-enhanced drug delivery systems.","authors":"Matteo Perra, Ines Castangia, Matteo Aroffu, Federica Fulgheri, Rita Abi-Rached, Maria Letizia Manca, Hernán Cortés, María Luisa Del Prado-Audelo, Carla Nomura-Contreras, Alejandra Romero-Montero, Dietrich Büsselberg, Gerardo Leyva-Gómez, Javad Sharifi-Rad, Daniela Calina","doi":"10.1007/s12672-025-01820-z","DOIUrl":"10.1007/s12672-025-01820-z","url":null,"abstract":"<p><p>Cancer remains the second leading cause of death globally, driving the need for innovative therapies. Among natural compounds, maytansinoids have shown significant promise, contributing to nearly 25% of recently approved anticancer drugs. Despite their potential, early clinical trials faced challenges due to severe side effects, prompting advancements in delivery systems such as antibody-maytansinoid conjugates (AMCs). This review highlights the anticancer activity of maytansinoids, with a focus on AMCs designed to target cancer cells specifically. Preclinical and clinical studies show that AMCs, including FDA-approved drugs like Kadcyla and Elahere, effectively inhibit tumor growth while reducing systemic toxicity. Key developments include improved synthesis methods, linker chemistry and payload design. Ongoing research aims to enhance the safety and efficacy of AMCs, integrate nanotechnology for drug delivery, and identify novel therapeutic targets. These advancements hold potential to transform maytansinoid-based cancer treatments in the future.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"73"},"PeriodicalIF":2.8,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11751265/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143001776","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Discover. Oncology
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