首页 > 最新文献

Translational cancer research最新文献

英文 中文
LncRNA CBR3-AS1 is associated with the BCR::ABL1 kinase independent mechanism of tyrosine kinase inhibitor (TKI) resistance in chronic myeloid leukemia patients. LncRNA CBR3-AS1 与慢性髓性白血病患者酪氨酸激酶抑制剂(TKI)耐药的 BCR::ABL1 激酶独立机制有关。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-24 DOI: 10.21037/tcr-24-281
Wuqiang Lin, Xiuli Chen, Heyong Zheng, Zhenjie Cai, Linjun Xie, Beibei Zhang, Rongrong Zheng

Background: It is difficult for chronic myeloid leukemia (CML) patients with BCR::ABL1 independent drug resistance to achieve optimal efficacy. The aim of this study is to investigate the BCR::ABL1 kinase independent mechanism of tyrosine kinase inhibitor (TKI) resistance in CML patients to develop targeted therapeutic strategy.

Methods: Herein, we analyzed the long non-coding RNA (lncRNA) and messenger RNA (mRNA) expression profiles of patients who achieved sustained deep molecular response (DMR) after TKI treatment and patients with non-DMR using RNA-seqencing. Furthermore, the differentially expressed lncRNAs and mRNAs were identified. The expression of chosen lncRNA was validated in an expanded cohort, and bioinformatics analysis was performed to analyze the function of selected mRNA.

Results: LncRNA data analysis indicated the diversity lncRNA profiles among healthy individuals, CML patients with non-DMR, and CML patients with DMR. Differential expression analysis and Veen plot of up-regulated lncRNAs in patients with non-DMR (compared with healthy individuals) and down-regulated lncRNAs in patients with DMR (compared to patients with non-DMR) revealed that lncRNA CBR3-AS1 overexpression might be related to BCR::ABL1 independent TKI resistance of CML patients. The expression of CBR3-AS1 was then verified in an expanded cohort, suggesting that, compared with control group, there was no statistical difference of CBR3-AS1 expression in DMR group, whereas, CBR3-AS1 was up-regulated in non-DMR group. Moreover, the mRNA data analysis of RNA-sequencing was performed. We considered genes that up-regulated in non-DMR group (compared with control group), down-regulated in DMR group (compared with non-DMR group), showed no statistical difference between control and DMR group as the potential genes that associated with TKI resistance of CML patients. A total of 55 corresponding mRNAs were obtained including KCNA6, a target gene of CBR3-AS1. Further bioinformatics analysis showed that the major interacted genes of KCNA6 were enriched in several resistance-associated pathways including interleukin -17 signaling pathway and cyclic adenosine monophosphate signaling pathway.

Conclusions: In conclusion, this work indicates that CBR3-AS1 might be involved in BCR::ABL1 independent TKI resistance of CML patients through targeting KCNA6, providing a novel target for intervention treatment of CML patients with BCR::ABL1 independent TKI resistance.

背景:具有BCR::ABL1独立耐药性的慢性髓性白血病(CML)患者很难获得最佳疗效。方法:我们利用RNA-seqencing技术分析了TKI治疗后获得持续深度分子反应(DMR)的患者和非DMR患者的长非编码RNA(lncRNA)和信使RNA(mRNA)表达谱。此外,还鉴定了差异表达的lncRNA和mRNA。在扩大的队列中验证了所选 lncRNA 的表达,并进行了生物信息学分析以分析所选 mRNA 的功能:结果:LncRNA数据分析显示,健康人、非DMR的CML患者和DMR的CML患者的lncRNA谱具有多样性。非DMR患者(与健康人相比)中上调的lncRNAs和DMR患者(与非DMR患者相比)中下调的lncRNAs的差异表达分析和Veen图显示,lncRNA CBR3-AS1的过表达可能与CML患者的BCR::ABL1独立TKI耐药有关。随后,CBR3-AS1的表达在扩大的队列中得到了验证,结果表明,与对照组相比,CBR3-AS1在DMR组中的表达没有统计学差异,而在非DMR组中,CBR3-AS1被上调。此外,我们还对 RNA 序列的 mRNA 数据进行了分析。我们将非DMR组(与对照组相比)上调、DMR组(与非DMR组相比)下调、对照组与DMR组之间无统计学差异的基因视为与CML患者TKI耐药相关的潜在基因。共获得 55 个相应的 mRNA,其中包括 CBR3-AS1 的靶基因 KCNA6。进一步的生物信息学分析表明,KCNA6的主要相互作用基因富集在几个耐药性相关通路中,包括白细胞介素-17信号通路和环磷酸腺苷信号通路:总之,这项研究表明CBR3-AS1可能通过靶向KCNA6参与了CML患者的BCR::ABL1独立TKI耐药,为干预治疗BCR::ABL1独立TKI耐药的CML患者提供了一个新的靶点。
{"title":"LncRNA <i>CBR3-AS1</i> is associated with the BCR::ABL1 kinase independent mechanism of tyrosine kinase inhibitor (TKI) resistance in chronic myeloid leukemia patients.","authors":"Wuqiang Lin, Xiuli Chen, Heyong Zheng, Zhenjie Cai, Linjun Xie, Beibei Zhang, Rongrong Zheng","doi":"10.21037/tcr-24-281","DOIUrl":"10.21037/tcr-24-281","url":null,"abstract":"<p><strong>Background: </strong>It is difficult for chronic myeloid leukemia (CML) patients with BCR::ABL1 independent drug resistance to achieve optimal efficacy. The aim of this study is to investigate the BCR::ABL1 kinase independent mechanism of tyrosine kinase inhibitor (TKI) resistance in CML patients to develop targeted therapeutic strategy.</p><p><strong>Methods: </strong>Herein, we analyzed the long non-coding RNA (lncRNA) and messenger RNA (mRNA) expression profiles of patients who achieved sustained deep molecular response (DMR) after TKI treatment and patients with non-DMR using RNA-seqencing. Furthermore, the differentially expressed lncRNAs and mRNAs were identified. The expression of chosen lncRNA was validated in an expanded cohort, and bioinformatics analysis was performed to analyze the function of selected mRNA.</p><p><strong>Results: </strong>LncRNA data analysis indicated the diversity lncRNA profiles among healthy individuals, CML patients with non-DMR, and CML patients with DMR. Differential expression analysis and Veen plot of up-regulated lncRNAs in patients with non-DMR (compared with healthy individuals) and down-regulated lncRNAs in patients with DMR (compared to patients with non-DMR) revealed that lncRNA <i>CBR3-AS1</i> overexpression might be related to BCR::ABL1 independent TKI resistance of CML patients. The expression of <i>CBR3-AS1</i> was then verified in an expanded cohort, suggesting that, compared with control group, there was no statistical difference of <i>CBR3-AS1</i> expression in DMR group, whereas, <i>CBR3-AS1</i> was up-regulated in non-DMR group. Moreover, the mRNA data analysis of RNA-sequencing was performed. We considered genes that up-regulated in non-DMR group (compared with control group), down-regulated in DMR group (compared with non-DMR group), showed no statistical difference between control and DMR group as the potential genes that associated with TKI resistance of CML patients. A total of 55 corresponding mRNAs were obtained including <i>KCNA6</i>, a target gene of <i>CBR3-AS1</i>. Further bioinformatics analysis showed that the major interacted genes of <i>KCNA6</i> were enriched in several resistance-associated pathways including interleukin -17 signaling pathway and cyclic adenosine monophosphate signaling pathway.</p><p><strong>Conclusions: </strong>In conclusion, this work indicates that <i>CBR3-AS1</i> might be involved in BCR::ABL1 independent TKI resistance of CML patients through targeting <i>KCNA6</i>, providing a novel target for intervention treatment of CML patients with BCR::ABL1 independent TKI resistance.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319989/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983289","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 and analysis of prognostic factors for fibroblastic osteosarcoma patients: a population-based study. 纤维组织骨肉瘤患者的生存期和预后因素分析:一项基于人群的研究。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-02 DOI: 10.21037/tcr-24-126
Dongsheng Zhu, Wen Zheng, Han Qi, Feng Chen, Xiaodong Wang

Background: Osteosarcoma is the most common mesenchymal cell malignancy, 10% of which is fibroblastic osteosarcoma (FOS). Due to the low incidence of osteosarcoma, the impact of many pathological factors on survival is still unclear, especially FOS. The goal of this study was to assess the latest survival rates for FOS and the risk factors affecting survival using the Surveillance, Epidemiology, and End Results (SEER) database.

Methods: Age, sex, race, SEER stage, surgery, radiation, chemotherapy, site of FOS, and survival time were collected from the SEER database for survival and prognostic factor analysis. The patients were randomly assigned to either the training cohort or the testing cohort. The overall survival (OS) curves were obtained by Kaplan-Meier according to different factors. A multivariate Cox regression model and a predictive nomogram have also been constructed.

Results: The study enrolled a total of 120 patients. OS at 1, 3, and 5 years for all patients was 90.83%, 79.17%, and 70.83%, respectively. In the 5-year survival analysis, in distant of SEER stage (P<0.01), radiation (P=0.03), and no surgery (P<0.01) were associated with a worse prognosis in patients with FOS. Multivariate analysis showed that age, and in distant of SEER stage were independent indicators of unfavorable prognosis. A nomogram was used to predict the prognosis of FOS and a calibration curve was used to validate the nomogram prediction against the actual observed survival outcomes.

Conclusions: In summary, older age, and worse SEER stage were associated with poorer OS. The nomogram effectively predicted the probabilities of 1-, 3-, and 5-year OS, demonstrating strong concordance with the actual observed outcomes.

背景:骨肉瘤是最常见的间叶细胞恶性肿瘤,其中10%为成纤维骨肉瘤(FOS)。由于骨肉瘤的发病率较低,许多病理因素对生存率的影响仍不明确,尤其是成纤维骨肉瘤。本研究的目的是利用监测、流行病学和最终结果(SEER)数据库评估FOS的最新存活率以及影响存活率的风险因素:方法:从SEER数据库中收集年龄、性别、种族、SEER分期、手术、放疗、化疗、FOS部位和存活时间,进行存活率和预后因素分析。患者被随机分配到训练组或测试组。根据不同的因素,通过卡普兰-梅耶(Kaplan-Meier)获得总生存期(OS)曲线。此外,还构建了多变量考克斯回归模型和预测提名图:研究共纳入了 120 名患者。所有患者的 1、3 和 5 年生存率分别为 90.83%、79.17% 和 70.83%。在 5 年生存率分析中,SEER 分期(PConclusions.PCR)的远期生存率分别为 90.83%、79.17% 和 70.83%:总之,年龄越大、SEER 分期越晚,患者的生存期越短。提名图有效预测了1年、3年和5年的OS概率,与实际观察结果非常吻合。
{"title":"Survival and analysis of prognostic factors for fibroblastic osteosarcoma patients: a population-based study.","authors":"Dongsheng Zhu, Wen Zheng, Han Qi, Feng Chen, Xiaodong Wang","doi":"10.21037/tcr-24-126","DOIUrl":"10.21037/tcr-24-126","url":null,"abstract":"<p><strong>Background: </strong>Osteosarcoma is the most common mesenchymal cell malignancy, 10% of which is fibroblastic osteosarcoma (FOS). Due to the low incidence of osteosarcoma, the impact of many pathological factors on survival is still unclear, especially FOS. The goal of this study was to assess the latest survival rates for FOS and the risk factors affecting survival using the Surveillance, Epidemiology, and End Results (SEER) database.</p><p><strong>Methods: </strong>Age, sex, race, SEER stage, surgery, radiation, chemotherapy, site of FOS, and survival time were collected from the SEER database for survival and prognostic factor analysis. The patients were randomly assigned to either the training cohort or the testing cohort. The overall survival (OS) curves were obtained by Kaplan-Meier according to different factors. A multivariate Cox regression model and a predictive nomogram have also been constructed.</p><p><strong>Results: </strong>The study enrolled a total of 120 patients. OS at 1, 3, and 5 years for all patients was 90.83%, 79.17%, and 70.83%, respectively. In the 5-year survival analysis, in distant of SEER stage (P<0.01), radiation (P=0.03), and no surgery (P<0.01) were associated with a worse prognosis in patients with FOS. Multivariate analysis showed that age, and in distant of SEER stage were independent indicators of unfavorable prognosis. A nomogram was used to predict the prognosis of FOS and a calibration curve was used to validate the nomogram prediction against the actual observed survival outcomes.</p><p><strong>Conclusions: </strong>In summary, older age, and worse SEER stage were associated with poorer OS. The nomogram effectively predicted the probabilities of 1-, 3-, and 5-year OS, demonstrating strong concordance with the actual observed outcomes.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319962/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983294","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
Combination approach using neoadjuvant therapy with radical prostatectomy for improving oncological outcomes of high-risk prostate cancer: a narrative review. 采用新辅助治疗与前列腺癌根治术相结合的方法改善高危前列腺癌的肿瘤治疗效果:综述。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-08 DOI: 10.21037/tcr-23-2394
Keita Nakane, Makoto Kawase, Daiki Kato, Koji Iinuma, Kota Kawase, Shinichi Takeuchi, Yuki Tobisawa, Takayasu Ito, Takuya Koie

Background and objective: Prostate cancer (PCa) is the most common cancer in men. High-risk PCa is associated with an increased risk of PCa-related death. The combined use of androgen deprivation therapy (ADT) is essential to improve oncological outcomes in patients with high-risk PCa, and relatively long-term ADT administration is preferred when radiotherapy is performed. Meanwhile, whether neoadjuvant therapy for radical prostatectomy (RP) improves oncological outcomes remains controversial. This study aimed to review the oncological outcomes of RP in high-risk PCa and emphasize the significance of neoadjuvant therapy including neoadjuvant hormonal therapy (NHT) and neoadjuvant chemohormonal therapy (NCHT) followed by RP for managing high-risk PCa.

Methods: We searched for articles published in the PubMed and Scopus databases from January 1, 2005 to March 30, 2023 using the medical subject headings (MeSH) terms: prostate cancer, prostatectomy, radiation therapy, neoadjuvant therapy, and treatment outcome.

Key content and findings: The study on NHT before RP for high-risk PCa found that NHT was associated with reduced adverse pathological features, such as pT3, positive surgical margins (PSM), and lymph node involvement. However, despite shorter operative times and improved surgical outcomes, NHT did not significantly enhance biochemical recurrence (BCR) or other oncological outcomes. The combination therapy using ADT and androgen receptor signaling inhibitors (ARSI) showed varying results. Another investigation explored NCHT with taxane-based agents, indicating acceptable treatment benefits and improved BCR-free survival rates in high-risk PCa patients, demonstrating potential feasibility for this approach. Ongoing trials, like the PROTEUS trial, aim to further evaluate the therapeutic efficacy of neoadjuvant therapy in high-risk PCa.

Conclusions: NHT for high-risk PCa does not contribute to improved oncological outcome and should not be administered easily for downstaging or PSM reduction. NHT in combination with ARSI has the potential advantage of improving the oncological outcome of high-risk PCa compared to RP alone, but the results are currently unsatisfactory, and the development of individualized treatment strategies using several different therapeutic approaches is needed.

背景和目的:前列腺癌(PCa)是男性最常见的癌症。高危 PCa 与 PCa 相关死亡风险增加有关。联合使用雄激素剥夺疗法(ADT)对改善高危 PCa 患者的肿瘤预后至关重要,在进行放射治疗时,应首选相对长期的 ADT 治疗。同时,根治性前列腺切除术(RP)的新辅助治疗是否能改善肿瘤预后仍存在争议。本研究旨在回顾前列腺癌根治术(RP)在高危PCa中的肿瘤治疗效果,并强调新辅助治疗(包括新辅助激素治疗(NHT)和新辅助化疗(NCHT))在前列腺癌根治术后治疗高危PCa中的重要意义:我们检索了2005年1月1日至2023年3月30日期间发表在PubMed和Scopus数据库中的文章,检索时使用了医学主题词(MeSH):前列腺癌、前列腺切除术、放射治疗、新辅助治疗和治疗结果:对高危前列腺癌RP术前进行新辅助治疗的研究发现,新辅助治疗与pT3、手术切缘阳性(PSM)和淋巴结受累等不良病理特征的减少有关。然而,尽管缩短了手术时间并改善了手术效果,NHT 并未显著提高生化复发(BCR)或其他肿瘤结果。使用 ADT 和雄激素受体信号转导抑制剂(ARSI)进行联合治疗的结果各不相同。另一项研究探讨了 NCHT 与以类固醇类药物为主的治疗方法,结果显示高危 PCa 患者的治疗效果可以接受,无 BCR 生存率也有所提高,这表明这种方法具有潜在的可行性。目前正在进行的试验,如PROTEUS试验,旨在进一步评估新辅助治疗在高危PCa中的疗效:结论:NHT治疗高危PCa无助于改善肿瘤预后,不应轻易为降低分期或减少PSM而进行NHT治疗。与单纯RP相比,NHT联合ARSI具有改善高危PCa肿瘤预后的潜在优势,但目前的结果并不令人满意,因此需要采用多种不同的治疗方法制定个体化治疗策略。
{"title":"Combination approach using neoadjuvant therapy with radical prostatectomy for improving oncological outcomes of high-risk prostate cancer: a narrative review.","authors":"Keita Nakane, Makoto Kawase, Daiki Kato, Koji Iinuma, Kota Kawase, Shinichi Takeuchi, Yuki Tobisawa, Takayasu Ito, Takuya Koie","doi":"10.21037/tcr-23-2394","DOIUrl":"10.21037/tcr-23-2394","url":null,"abstract":"<p><strong>Background and objective: </strong>Prostate cancer (PCa) is the most common cancer in men. High-risk PCa is associated with an increased risk of PCa-related death. The combined use of androgen deprivation therapy (ADT) is essential to improve oncological outcomes in patients with high-risk PCa, and relatively long-term ADT administration is preferred when radiotherapy is performed. Meanwhile, whether neoadjuvant therapy for radical prostatectomy (RP) improves oncological outcomes remains controversial. This study aimed to review the oncological outcomes of RP in high-risk PCa and emphasize the significance of neoadjuvant therapy including neoadjuvant hormonal therapy (NHT) and neoadjuvant chemohormonal therapy (NCHT) followed by RP for managing high-risk PCa.</p><p><strong>Methods: </strong>We searched for articles published in the PubMed and Scopus databases from January 1, 2005 to March 30, 2023 using the medical subject headings (MeSH) terms: prostate cancer, prostatectomy, radiation therapy, neoadjuvant therapy, and treatment outcome.</p><p><strong>Key content and findings: </strong>The study on NHT before RP for high-risk PCa found that NHT was associated with reduced adverse pathological features, such as pT3, positive surgical margins (PSM), and lymph node involvement. However, despite shorter operative times and improved surgical outcomes, NHT did not significantly enhance biochemical recurrence (BCR) or other oncological outcomes. The combination therapy using ADT and androgen receptor signaling inhibitors (ARSI) showed varying results. Another investigation explored NCHT with taxane-based agents, indicating acceptable treatment benefits and improved BCR-free survival rates in high-risk PCa patients, demonstrating potential feasibility for this approach. Ongoing trials, like the PROTEUS trial, aim to further evaluate the therapeutic efficacy of neoadjuvant therapy in high-risk PCa.</p><p><strong>Conclusions: </strong>NHT for high-risk PCa does not contribute to improved oncological outcome and should not be administered easily for downstaging or PSM reduction. NHT in combination with ARSI has the potential advantage of improving the oncological outcome of high-risk PCa compared to RP alone, but the results are currently unsatisfactory, and the development of individualized treatment strategies using several different therapeutic approaches is needed.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319982/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983321","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
Stratifying osteosarcoma patients using an epigenetic modification-related prognostic signature: implications for immunotherapy and chemotherapy selection. 利用表观遗传修饰相关预后特征对骨肉瘤患者进行分层:对免疫疗法和化疗选择的影响。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-22 DOI: 10.21037/tcr-23-2300
Zhichao Li, Yong Xue, Xianxing Huang, Gang Xiao

Background: Osteosarcoma (OS) poses significant challenges in treatment and lacks reliable prognostic markers. Epigenetic alterations play a crucial role in disease progression. This study aimed to develop an accurate prognostic signature for OS using epigenetic modification genes (EMGs).

Methods: The Therapeutically Applicable Research to Generate Effective Treatments (TARGET)-OS cohort was analyzed. Univariate Cox analysis identified survival-associated EMGs. Based on least absolute shrinkage and selection operator (LASSO) regression and multivariate analysis, a 6-gene prognostic signature termed the epigenetic modification-related prognostic signature (EMRPS) was derived in the testing cohort. Kaplan-Meier and receiver operating characteristic (ROC) curve analysis confirmed predictive accuracy through internal and external validation (GEO accession GSE21257). A prognostic nomogram incorporating EMRPS and clinical features was constructed. Transcriptomic analysis including differential gene expression, Gene Ontology (GO), gene set enrichment analysis (GSEA), and immune infiltration analysis was conducted to explore mechanisms linking EMRPS to OS prognosis. Additionally, EMRPS impact on drug sensitivity was predicted.

Results: A 6-gene EMRPS comprising DDX24, DNAJC1, HDAC4, SIRT7, SP140 and UHRF2 was successfully developed. The high-risk group showed significantly shorter survival, consistently observed in both internal and external validation. EMRPS demonstrated high predictive efficacy for 1-, 3-, and 5-year overall survival, with area under curve (AUC) >0.85 in training and ~0.7 in testing. The nomogram integrating age, gender, metastasis status, and EMRPS exhibited high predictive performance based on concordance index analysis. Mechanistic analysis indicated the low-risk group had increased immune infiltration and activity with higher immune checkpoint expression, reflecting an immune-activated tumor microenvironment (TME) suitable for immunotherapy. Drug sensitivity analysis revealed the low-risk group had increased sensitivity to cisplatin, a first-line OS chemotherapy.

Conclusions: Our study successfully established an efficient EMRPS and nomogram, highlighting their potential as novel prognostic markers and indicators for selecting appropriate immunotherapy and chemotherapy candidates in OS treatment.

背景:骨肉瘤(Osteosarcoma,OS)给治疗带来了巨大挑战,而且缺乏可靠的预后标志物。表观遗传学改变在疾病进展中起着至关重要的作用。本研究旨在利用表观遗传修饰基因(EMGs)为骨肉瘤建立准确的预后特征:方法:分析了 "产生有效治疗的治疗性应用研究(TARGET)-OS队列"。单变量 Cox 分析确定了与生存相关的 EMGs。基于最小绝对收缩和选择算子(LASSO)回归和多变量分析,在测试队列中得出了一个6个基因的预后特征,称为表观遗传修饰相关预后特征(EMRPS)。Kaplan-Meier和接收者操作特征(ROC)曲线分析通过内部和外部验证证实了预测的准确性(GEO accession GSE21257)。结合 EMRPS 和临床特征构建了预后提名图。转录组分析包括差异基因表达、基因本体(GO)、基因组富集分析(GSEA)和免疫浸润分析,以探索EMRPS与OS预后的关联机制。此外,还预测了EMRPS对药物敏感性的影响:结果:由DDX24、DNAJC1、HDAC4、SIRT7、SP140和UHRF2组成的6基因EMRPS被成功开发出来。高风险组的生存期明显较短,这在内部和外部验证中都得到了一致观察。EMRPS对1年、3年和5年总生存率具有很高的预测效力,其曲线下面积(AUC)在训练中大于0.85,在测试中约为0.7。根据一致性指数分析,整合了年龄、性别、转移状态和 EMRPS 的提名图显示出较高的预测性能。机理分析表明,低风险组的免疫浸润和活性增加,免疫检查点表达较高,反映了适合免疫疗法的免疫激活肿瘤微环境(TME)。药物敏感性分析显示,低风险组对顺铂的敏感性增加,而顺铂是一线OS化疗药物:我们的研究成功建立了高效的EMRPS和提名图,凸显了它们作为新型预后标志物和指标的潜力,可用于在OS治疗中选择合适的免疫疗法和化疗候选方案。
{"title":"Stratifying osteosarcoma patients using an epigenetic modification-related prognostic signature: implications for immunotherapy and chemotherapy selection.","authors":"Zhichao Li, Yong Xue, Xianxing Huang, Gang Xiao","doi":"10.21037/tcr-23-2300","DOIUrl":"10.21037/tcr-23-2300","url":null,"abstract":"<p><strong>Background: </strong>Osteosarcoma (OS) poses significant challenges in treatment and lacks reliable prognostic markers. Epigenetic alterations play a crucial role in disease progression. This study aimed to develop an accurate prognostic signature for OS using epigenetic modification genes (EMGs).</p><p><strong>Methods: </strong>The Therapeutically Applicable Research to Generate Effective Treatments (TARGET)-OS cohort was analyzed. Univariate Cox analysis identified survival-associated EMGs. Based on least absolute shrinkage and selection operator (LASSO) regression and multivariate analysis, a 6-gene prognostic signature termed the epigenetic modification-related prognostic signature (EMRPS) was derived in the testing cohort. Kaplan-Meier and receiver operating characteristic (ROC) curve analysis confirmed predictive accuracy through internal and external validation (GEO accession GSE21257). A prognostic nomogram incorporating EMRPS and clinical features was constructed. Transcriptomic analysis including differential gene expression, Gene Ontology (GO), gene set enrichment analysis (GSEA), and immune infiltration analysis was conducted to explore mechanisms linking EMRPS to OS prognosis. Additionally, EMRPS impact on drug sensitivity was predicted.</p><p><strong>Results: </strong>A 6-gene EMRPS comprising <i>DDX24</i>, <i>DNAJC1</i>, <i>HDAC4</i>, <i>SIRT7</i>, <i>SP140</i> and <i>UHRF2</i> was successfully developed. The high-risk group showed significantly shorter survival, consistently observed in both internal and external validation. EMRPS demonstrated high predictive efficacy for 1-, 3-, and 5-year overall survival, with area under curve (AUC) >0.85 in training and ~0.7 in testing. The nomogram integrating age, gender, metastasis status, and EMRPS exhibited high predictive performance based on concordance index analysis. Mechanistic analysis indicated the low-risk group had increased immune infiltration and activity with higher immune checkpoint expression, reflecting an immune-activated tumor microenvironment (TME) suitable for immunotherapy. Drug sensitivity analysis revealed the low-risk group had increased sensitivity to cisplatin, a first-line OS chemotherapy.</p><p><strong>Conclusions: </strong>Our study successfully established an efficient EMRPS and nomogram, highlighting their potential as novel prognostic markers and indicators for selecting appropriate immunotherapy and chemotherapy candidates in OS treatment.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319966/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983293","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
The role of dysregulated metabolism and associated genes in gastric cancer initiation and development. 代谢失调及相关基因在胃癌诱发和发展中的作用。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-22 DOI: 10.21037/tcr-23-2244
Zhengyan Jiang, Zhengrong Gu, Xianyan Lu, Wei Wen

The review delves into the intricate interplay between metabolic dysregulation and the onset and progression of gastric cancer (GC), shedding light on a pivotal aspect of this prevalent malignancy. GC stands as one of the leading causes of cancer-related mortality worldwide, its trajectory influenced by a multitude of factors, among which metabolic dysregulation and aberrant gene expression play significant roles. The article navigates through the fundamental roles of metabolic dysregulation in the genesis of GC, unveiling phenomena such as aberrant glycolysis, epitomized by the Warburg effect, alongside anomalies in lipid and amino acid metabolism. It delineates how these disruptions fuel the cancerous process, facilitating uncontrolled cell proliferation and survival. Furthermore, the intricate nexus between metabolism and the vitality of GC cells is elucidated, underscoring the profound influence of metabolic reprogramming on tumor energy dynamics and the accrual of metabolic by-products, which further perpetuate malignant growth. A pivotal segment of the review entails an exploration of key metabolic-related genes implicated in GC pathogenesis. MYC and TP53 are spotlighted among others, delineating their pivotal roles in driving tumorigenesis through metabolic pathway modulation. These genetic pathways serve as critical nodes in the intricate network orchestrating GC development, providing valuable targets for therapeutic intervention. This review embarks on a forward-looking trajectory, delineating the potential therapeutic avenues stemming from insights into metabolic dysregulation in GC. It underscores the promise of targeted therapies directed towards specific metabolic pathways implicated in tumor progression, alongside the burgeoning potential of combination therapy strategies leveraging both metabolic and conventional anti-cancer modalities. In essence, this comprehensive review serves as a beacon, illuminating the intricate landscape of metabolic dysregulation in GC pathogenesis. Through its nuanced exploration of metabolic aberrations and their genetic underpinnings, it not only enriches our understanding of GC biology but also unveils novel therapeutic vistas poised to revolutionize its clinical management.

这篇综述深入探讨了新陈代谢失调与胃癌(GC)的发生和发展之间错综复杂的相互作用,揭示了这种流行性恶性肿瘤的一个关键方面。胃癌是导致全球癌症相关死亡的主要原因之一,其发病轨迹受多种因素影响,其中代谢失调和异常基因表达起着重要作用。文章介绍了代谢失调在 GC 发病过程中的基本作用,揭示了以沃伯格效应为代表的糖酵解异常现象,以及脂质和氨基酸代谢异常。它描述了这些紊乱是如何助长癌症进程、促进细胞失控增殖和存活的。此外,还阐明了新陈代谢与 GC 细胞活力之间错综复杂的关系,强调了新陈代谢重编程对肿瘤能量动态和新陈代谢副产物累积的深远影响,这些副产物会进一步延续恶性肿瘤的生长。这篇综述的一个重要部分是探讨与 GC 发病机制有关的关键代谢相关基因。其中重点介绍了 MYC 和 TP53,阐明了它们在通过调节代谢途径驱动肿瘤发生方面的关键作用。这些基因通路是协调 GC 发展的复杂网络中的关键节点,为治疗干预提供了有价值的靶点。这篇综述以前瞻性的视角描绘了源自对 GC 代谢失调的深入了解的潜在治疗途径。它强调了针对与肿瘤进展有关的特定代谢通路的靶向治疗的前景,以及利用代谢和传统抗癌模式的联合治疗策略的巨大潜力。从本质上讲,这篇综合性综述就像一盏明灯,照亮了 GC 发病机制中代谢失调的复杂图景。通过对代谢畸变及其遗传基础的深入探讨,它不仅丰富了我们对 GC 生物学的认识,而且还揭示了有望彻底改变临床治疗的新的治疗前景。
{"title":"The role of dysregulated metabolism and associated genes in gastric cancer initiation and development.","authors":"Zhengyan Jiang, Zhengrong Gu, Xianyan Lu, Wei Wen","doi":"10.21037/tcr-23-2244","DOIUrl":"10.21037/tcr-23-2244","url":null,"abstract":"<p><p>The review delves into the intricate interplay between metabolic dysregulation and the onset and progression of gastric cancer (GC), shedding light on a pivotal aspect of this prevalent malignancy. GC stands as one of the leading causes of cancer-related mortality worldwide, its trajectory influenced by a multitude of factors, among which metabolic dysregulation and aberrant gene expression play significant roles. The article navigates through the fundamental roles of metabolic dysregulation in the genesis of GC, unveiling phenomena such as aberrant glycolysis, epitomized by the Warburg effect, alongside anomalies in lipid and amino acid metabolism. It delineates how these disruptions fuel the cancerous process, facilitating uncontrolled cell proliferation and survival. Furthermore, the intricate nexus between metabolism and the vitality of GC cells is elucidated, underscoring the profound influence of metabolic reprogramming on tumor energy dynamics and the accrual of metabolic by-products, which further perpetuate malignant growth. A pivotal segment of the review entails an exploration of key metabolic-related genes implicated in GC pathogenesis. MYC and TP53 are spotlighted among others, delineating their pivotal roles in driving tumorigenesis through metabolic pathway modulation. These genetic pathways serve as critical nodes in the intricate network orchestrating GC development, providing valuable targets for therapeutic intervention. This review embarks on a forward-looking trajectory, delineating the potential therapeutic avenues stemming from insights into metabolic dysregulation in GC. It underscores the promise of targeted therapies directed towards specific metabolic pathways implicated in tumor progression, alongside the burgeoning potential of combination therapy strategies leveraging both metabolic and conventional anti-cancer modalities. In essence, this comprehensive review serves as a beacon, illuminating the intricate landscape of metabolic dysregulation in GC pathogenesis. Through its nuanced exploration of metabolic aberrations and their genetic underpinnings, it not only enriches our understanding of GC biology but also unveils novel therapeutic vistas poised to revolutionize its clinical management.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983299","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
SCG2 and CPE may be novel markers for the identification of pancreatic neuroendocrine tumors and solid pseudopapillary neoplasms. SCG2和CPE可能是鉴别胰腺神经内分泌肿瘤和实体假乳头状瘤的新型标记物。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-18 DOI: 10.21037/tcr-24-229
Wuhan Yang, Shubin Wang, Zhilei Zhang, Hao Guo, Chengyu Liu, Meng Zhao, Yueping Liu, Li Peng

Background: Distinguishing pancreatic neuroendocrine tumors (pNETs) from solid pseudopapillary neoplasms (SPNs) is challenging, primarily due to their overlapping pathological characteristics. To address this, our study aims to identify and validate novel biomarkers that effectively differentiate between these two conditions. We focus on the exploration of new immunohistochemical markers to enhance this distinction.

Methods: In this study, we analyzed genetic variations in pNETs and SPNs using the GSE43795 dataset from the Gene Expression Omnibus (GEO) database. Our approach was to identify genes with higher expression in pNETs compared to SPNs and normal pancreatic tissues. We conducted enrichment analyses to understand the functions of these genes. Furthermore, protein-protein interaction (PPI) network analysis was utilized to identify key genes associated with pNETs. Our sample consisted of 163 pancreatic tumor specimens, comprising 78 pNETs and 85 SPNs. We also collected clinicopathological data and used immunohistochemistry to measure the expression levels of these key genes.

Results: The enrichment analysis revealed that genes overexpressed in pNETs were mainly involved in signal release, vesicle transport, and ion pathway activation, playing significant roles in endocrine processes like insulin secretion, dopamine synapses, and circadian rhythm regulation. The PPI analysis identified secretogranin II (SCG2), carboxypeptidase E (CPE), and chromogranin A (CgA, CHGA) as key markers for differentiating pNETs from SPNs. Immunohistochemical validation of these markers demonstrated high sensitivity (SCG2: 98.7%, CPE: 97.4%) and specificity (100%), indicating their superior discriminative power compared to traditional markers like CgA, β-catenin, lymphoid enhancer-binding factor 1 (LEF1), and vimentin.

Conclusions: Our study indicates that SCG2 and CPE are effective, novel immunohistochemical biomarkers for differentiating pNETs from SPNs.

背景:将胰腺神经内分泌肿瘤(pNET)与实性假乳头状瘤(SPN)区分开来具有挑战性,这主要是由于它们的病理特征相互重叠。为了解决这个问题,我们的研究旨在鉴定和验证能有效区分这两种疾病的新型生物标记物。我们重点探索新的免疫组化标记物,以加强这种区分:在这项研究中,我们利用基因表达总库(GEO)数据库中的 GSE43795 数据集分析了 pNET 和 SPN 的基因变异。我们的方法是找出与 SPNs 和正常胰腺组织相比,pNETs 中表达量更高的基因。我们进行了富集分析,以了解这些基因的功能。此外,我们还利用蛋白-蛋白相互作用(PPI)网络分析来确定与 pNET 相关的关键基因。我们的样本包括 163 份胰腺肿瘤标本,其中 78 份为 pNET,85 份为 SPN。我们还收集了临床病理数据,并使用免疫组化方法测量了这些关键基因的表达水平:富集分析表明,pNET 中过表达的基因主要参与信号释放、囊泡转运和离子通道激活,在胰岛素分泌、多巴胺突触和昼夜节律调节等内分泌过程中发挥重要作用。PPI分析发现,泌泌素II(SCG2)、羧肽酶E(CPE)和嗜铬粒蛋白A(CgA,CHGA)是区分pNET和SPN的关键标志物。这些标记物的免疫组化验证显示了较高的灵敏度(SCG2:98.7%,CPE:97.4%)和特异性(100%),表明它们的鉴别力优于CgA、β-catenin、淋巴增强子结合因子1(LEF1)和波形蛋白等传统标记物:我们的研究表明,SCG2 和 CPE 是区分 pNET 和 SPN 的有效、新型免疫组化生物标记物。
{"title":"SCG2 and CPE may be novel markers for the identification of pancreatic neuroendocrine tumors and solid pseudopapillary neoplasms.","authors":"Wuhan Yang, Shubin Wang, Zhilei Zhang, Hao Guo, Chengyu Liu, Meng Zhao, Yueping Liu, Li Peng","doi":"10.21037/tcr-24-229","DOIUrl":"10.21037/tcr-24-229","url":null,"abstract":"<p><strong>Background: </strong>Distinguishing pancreatic neuroendocrine tumors (pNETs) from solid pseudopapillary neoplasms (SPNs) is challenging, primarily due to their overlapping pathological characteristics. To address this, our study aims to identify and validate novel biomarkers that effectively differentiate between these two conditions. We focus on the exploration of new immunohistochemical markers to enhance this distinction.</p><p><strong>Methods: </strong>In this study, we analyzed genetic variations in pNETs and SPNs using the GSE43795 dataset from the Gene Expression Omnibus (GEO) database. Our approach was to identify genes with higher expression in pNETs compared to SPNs and normal pancreatic tissues. We conducted enrichment analyses to understand the functions of these genes. Furthermore, protein-protein interaction (PPI) network analysis was utilized to identify key genes associated with pNETs. Our sample consisted of 163 pancreatic tumor specimens, comprising 78 pNETs and 85 SPNs. We also collected clinicopathological data and used immunohistochemistry to measure the expression levels of these key genes.</p><p><strong>Results: </strong>The enrichment analysis revealed that genes overexpressed in pNETs were mainly involved in signal release, vesicle transport, and ion pathway activation, playing significant roles in endocrine processes like insulin secretion, dopamine synapses, and circadian rhythm regulation. The PPI analysis identified secretogranin II (SCG2), carboxypeptidase E (CPE), and chromogranin A (CgA, CHGA) as key markers for differentiating pNETs from SPNs. Immunohistochemical validation of these markers demonstrated high sensitivity (SCG2: 98.7%, CPE: 97.4%) and specificity (100%), indicating their superior discriminative power compared to traditional markers like CgA, β-catenin, lymphoid enhancer-binding factor 1 (LEF1), and vimentin.</p><p><strong>Conclusions: </strong>Our study indicates that SCG2 and CPE are effective, novel immunohistochemical biomarkers for differentiating pNETs from SPNs.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319939/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983334","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
SLC12A8 upregulation promotes colorectal cancer progression and chemoresistance. SLC12A8 上调会促进结直肠癌的进展和化疗耐药性。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-24 DOI: 10.21037/tcr-24-87
Zhe Sun, Zhiyan Nie, Yao Xu, Yingshun Cui, Wenjian Ma, Tongcun Zhang

Background: Colorectal cancer (CRC), a prevalent gastrointestinal malignant disease, causes substantial morbidity and mortality. Identification of novel prognostic biomarkers and therapeutic targets is critically needed to improve patient outcomes. Although solute carrier family 12 member 8 (SLC12A8) has high expression in various tumors and affects tumor progression, its role in CRC remains unclear. The aim of this study was to investigate the functions of SLC12A8 in CRC.

Methods: SLC12A8 expression and its association with clinical significance in CRC patients were explored via multiple public databases, including The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), The Human Protein Atlas (HPA), The University of ALabama at Birmingham CANcer data analysis Portal (UALCAN), and Kaplan-Meier plotter. The effects of SLC12A8 on the CRC cell apoptosis, epithelial-mesenchymal transition (EMT), reactive oxygen species (ROS) production, and sensitivity to oxaliplatin were verified by in vitro experiments.

Results: SLC12A8 expression was upregulated in CRC tissues compared with normal colorectal tissues. Furthermore, high expression of SLC12A8 was associated with poorer prognosis in CRC patients. Pathway enrichment analyses revealed SLC12A8 involvement in oxidative stress and transforming growth factor-beta (TGF-β) signaling. Experiments in CRC cells showed that SLC12A8 upregulation promoted apoptosis resistance, EMT, and inhibited ROS production. Moreover, SLC12A8 knockdown enhanced the sensitivity of CRC cells to oxaliplatin chemotherapy.

Conclusions: Our integrative analyses identify SLC12A8 as a candidate biomarker for CRC progression. Targeting SLC12A8 may improve patient responses to oxaliplatin-based treatment regimens.

背景:结直肠癌(CRC)是一种常见的胃肠道恶性疾病,会导致严重的发病率和死亡率。为改善患者预后,亟需鉴定新的预后生物标志物和治疗靶点。虽然溶质运载家族 12 成员 8(SLC12A8)在多种肿瘤中高表达并影响肿瘤的进展,但它在 CRC 中的作用仍不清楚。本研究旨在探讨 SLC12A8 在 CRC 中的功能:方法:通过多个公共数据库,包括癌症基因组图谱(TCGA)、基因型-组织表达(GTEx)、人类蛋白质图谱(HPA)、阿拉巴马大学伯明翰分校CANcer数据分析门户网站(UALCAN)和Kaplan-Meier绘图仪,探讨SLC12A8在CRC患者中的表达及其与临床意义的关联。体外实验验证了SLC12A8对CRC细胞凋亡、上皮-间质转化(EMT)、活性氧(ROS)产生以及奥沙利铂敏感性的影响:结果:与正常结直肠组织相比,SLC12A8在CRC组织中表达上调。此外,SLC12A8的高表达与CRC患者较差的预后有关。通路富集分析显示,SLC12A8 参与了氧化应激和转化生长因子-β(TGF-β)信号转导。在 CRC 细胞中进行的实验表明,SLC12A8 上调可促进细胞凋亡抵抗、EMT 和抑制 ROS 的产生。此外,敲除 SLC12A8 还能增强 CRC 细胞对奥沙利铂化疗的敏感性:结论:我们的综合分析确定了 SLC12A8 是 CRC 进展的候选生物标记物。靶向 SLC12A8 可能会改善患者对基于奥沙利铂的治疗方案的反应。
{"title":"SLC12A8 upregulation promotes colorectal cancer progression and chemoresistance.","authors":"Zhe Sun, Zhiyan Nie, Yao Xu, Yingshun Cui, Wenjian Ma, Tongcun Zhang","doi":"10.21037/tcr-24-87","DOIUrl":"10.21037/tcr-24-87","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC), a prevalent gastrointestinal malignant disease, causes substantial morbidity and mortality. Identification of novel prognostic biomarkers and therapeutic targets is critically needed to improve patient outcomes. Although solute carrier family 12 member 8 (SLC12A8) has high expression in various tumors and affects tumor progression, its role in CRC remains unclear. The aim of this study was to investigate the functions of SLC12A8 in CRC.</p><p><strong>Methods: </strong>SLC12A8 expression and its association with clinical significance in CRC patients were explored via multiple public databases, including The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), The Human Protein Atlas (HPA), The University of ALabama at Birmingham CANcer data analysis Portal (UALCAN), and Kaplan-Meier plotter. The effects of SLC12A8 on the CRC cell apoptosis, epithelial-mesenchymal transition (EMT), reactive oxygen species (ROS) production, and sensitivity to oxaliplatin were verified by <i>in vitro</i> experiments.</p><p><strong>Results: </strong>SLC12A8 expression was upregulated in CRC tissues compared with normal colorectal tissues. Furthermore, high expression of SLC12A8 was associated with poorer prognosis in CRC patients. Pathway enrichment analyses revealed SLC12A8 involvement in oxidative stress and transforming growth factor-beta (TGF-β) signaling. Experiments in CRC cells showed that SLC12A8 upregulation promoted apoptosis resistance, EMT, and inhibited ROS production. Moreover, SLC12A8 knockdown enhanced the sensitivity of CRC cells to oxaliplatin chemotherapy.</p><p><strong>Conclusions: </strong>Our integrative analyses identify SLC12A8 as a candidate biomarker for CRC progression. Targeting SLC12A8 may improve patient responses to oxaliplatin-based treatment regimens.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319960/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983336","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
Exploring the effects of matrix metalloproteinase-13 on the malignant biological behavior of tongue squamous cell carcinoma via the TNF signaling pathway based on bioinformatics methods. 基于生物信息学方法探讨基质金属蛋白酶-13通过TNF信号通路对舌鳞癌恶性生物学行为的影响
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-26 DOI: 10.21037/tcr-24-1016
Junqin Lu, Yeqian Zhu, Jie Zhang, Ningning Cao

Background: Identification of the etiology, molecular mechanisms, and carcinogenic pathways of tongue squamous cell carcinoma (TSCC) is crucial for developing new diagnostic and therapeutic strategies. This study used bioinformatics methods to identify key genes in TSCC and explored the potential functions and pathway mechanisms related to the malignant biological behavior of TSCC.

Methods: Gene chip data sets (i.e., GSE13601 and GSE34106) containing the data of both TSCC patients and normal control subjects were selected from the Gene Expression Omnibus (GEO) database. Using a gene expression analysis tool (GEO2R) of the GEO database, the differentially expressed genes (DEGs) were identified using the following criteria: |log fold change| >1, and P<0.05. The GEO2R tool was also used to select the upregulated DEGs in the chip candidates based on a P value <0.05. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, Gene Ontology (GO) function analysis, and a protein-protein interaction (PPI) network analysis were then conducted. The results were displayed using R language packages, including volcano plots, Venn diagrams, heatmaps, and enriched pathway bubble charts. Genes from the MalaCards database were compared with the candidate genes, and a thorough review of the literature was conducted to determine the clinical significance of these genes. Finally, feature gene-directed chemical drugs or targeted drugs were predicted using the Comparative Toxicogenomics Database (CTD).

Results: In total, 767 upregulated DEGs were identified from GSE13601 and 695 from GSE34106. By intersecting the upregulated DEGs from both data sets using a Venn diagram, 100 DEGs related to TSCC were identified. The enrichment analysis of the KEGG signaling pathways identified the majority of the pathways associated with the upregulated DEGs, including the Toll-like receptor signaling pathway, the extracellular matrix-receptor interaction, the tumor necrosis factor (TNF) signaling pathway, cytokine-cytokine receptor interaction, the chemokine signaling pathway, the interlukin-17 signaling pathway, and natural killer cell-mediated cytotoxicity. The PPI network and module analyses of the shared DEGs ultimately resulted in five clusters and 55 candidate genes. A further intersection analysis of the TSCC-related genes in the MalaCards database via a Venn diagram identified three important shared DEGs; that is, matrix metalloproteinase-1 (MMP1), MMP9, and MMP13. In the CTD, seven drugs related to MMP13 were identified for treating tongue tumors.

Conclusions: This study identified key genes and signaling pathways involved in TSCC and thus extended understandings of the molecular mechanisms that underlie the development and progression of TSCC. Additionally, this study showed that MMP13 may influence the malignant biological behavior of TSCC thro

背景:鉴定舌鳞状细胞癌(TSCC)的病因、分子机制和致癌途径对于开发新的诊断和治疗策略至关重要。本研究采用生物信息学方法鉴定了TSCC中的关键基因,并探索了与TSCC恶性生物学行为相关的潜在功能和通路机制:方法:从基因表达总库(GEO)数据库中选取了包含TSCC患者和正常对照组数据的基因芯片数据集(即GSE13601和GSE34106)。利用 GEO 数据库的基因表达分析工具(GEO2R),按照以下标准鉴定差异表达基因(DEGs):|log fold change| >1,PResults:在 GSE13601 和 GSE34106 中分别发现了 767 个和 695 个上调 DEGs。利用维恩图将两个数据集中的上调 DEGs 相交,发现了 100 个与 TSCC 相关的 DEGs。KEGG 信号通路的富集分析确定了与上调 DEGs 相关的大部分通路,包括 Toll 样受体信号通路、细胞外基质-受体相互作用、肿瘤坏死因子(TNF)信号通路、细胞因子-细胞因子受体相互作用、趋化因子信号通路、interlukin-17 信号通路和自然杀伤细胞介导的细胞毒性。对共有的 DEGs 进行 PPI 网络和模块分析后,最终得出了 5 个集群和 55 个候选基因。通过维恩图对MalaCards数据库中的TSCC相关基因进行进一步交叉分析,发现了三个重要的共享DEG,即基质金属蛋白酶-1(MMP1)、MMP9和MMP13。在CTD中,发现了7种与MMP13相关的治疗舌肿瘤的药物:本研究确定了参与 TSCC 的关键基因和信号通路,从而扩展了对 TSCC 发生和发展的分子机制的认识。此外,本研究还发现 MMP13 可通过 TNF 信号通路影响 TSCC 的恶性生物学行为。这一发现可为早期鉴别诊断和靶向治疗研究提供理论依据。
{"title":"Exploring the effects of matrix metalloproteinase-13 on the malignant biological behavior of tongue squamous cell carcinoma via the TNF signaling pathway based on bioinformatics methods.","authors":"Junqin Lu, Yeqian Zhu, Jie Zhang, Ningning Cao","doi":"10.21037/tcr-24-1016","DOIUrl":"10.21037/tcr-24-1016","url":null,"abstract":"<p><strong>Background: </strong>Identification of the etiology, molecular mechanisms, and carcinogenic pathways of tongue squamous cell carcinoma (TSCC) is crucial for developing new diagnostic and therapeutic strategies. This study used bioinformatics methods to identify key genes in TSCC and explored the potential functions and pathway mechanisms related to the malignant biological behavior of TSCC.</p><p><strong>Methods: </strong>Gene chip data sets (i.e., GSE13601 and GSE34106) containing the data of both TSCC patients and normal control subjects were selected from the Gene Expression Omnibus (GEO) database. Using a gene expression analysis tool (GEO2R) of the GEO database, the differentially expressed genes (DEGs) were identified using the following criteria: |log fold change| >1, and P<0.05. The GEO2R tool was also used to select the upregulated DEGs in the chip candidates based on a P value <0.05. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, Gene Ontology (GO) function analysis, and a protein-protein interaction (PPI) network analysis were then conducted. The results were displayed using R language packages, including volcano plots, Venn diagrams, heatmaps, and enriched pathway bubble charts. Genes from the MalaCards database were compared with the candidate genes, and a thorough review of the literature was conducted to determine the clinical significance of these genes. Finally, feature gene-directed chemical drugs or targeted drugs were predicted using the Comparative Toxicogenomics Database (CTD).</p><p><strong>Results: </strong>In total, 767 upregulated DEGs were identified from GSE13601 and 695 from GSE34106. By intersecting the upregulated DEGs from both data sets using a Venn diagram, 100 DEGs related to TSCC were identified. The enrichment analysis of the KEGG signaling pathways identified the majority of the pathways associated with the upregulated DEGs, including the Toll-like receptor signaling pathway, the extracellular matrix-receptor interaction, the tumor necrosis factor (TNF) signaling pathway, cytokine-cytokine receptor interaction, the chemokine signaling pathway, the interlukin-17 signaling pathway, and natural killer cell-mediated cytotoxicity. The PPI network and module analyses of the shared DEGs ultimately resulted in five clusters and 55 candidate genes. A further intersection analysis of the TSCC-related genes in the MalaCards database via a Venn diagram identified three important shared DEGs; that is, matrix metalloproteinase-1 (<i>MMP1</i>), <i>MMP9</i>, and <i>MMP13</i>. In the CTD, seven drugs related to <i>MMP13</i> were identified for treating tongue tumors.</p><p><strong>Conclusions: </strong>This study identified key genes and signaling pathways involved in TSCC and thus extended understandings of the molecular mechanisms that underlie the development and progression of TSCC. Additionally, this study showed that <i>MMP13</i> may influence the malignant biological behavior of TSCC thro","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319986/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983359","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
Treatments and prognostic outcomes of combined hepatocellular-cholangiocarcinoma with distant metastasis: an analysis based on SEER data. 肝细胞胆管癌合并远处转移的治疗和预后结果:基于 SEER 数据的分析。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-17 DOI: 10.21037/tcr-24-447
Leilei Zhang, Yunxi Lu, Yuting Chen, Xiangling Lu, Xiaoli Lao

Background: Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a rare liver cancer with a poor prognosis, often diagnosed at an advanced stage. The management of cHCC-CCA with distant metastasis remains challenging, and prognostic factors are not well-defined. This study aimed to investigate prognostic factors and treatment outcomes for cHCC-CCA patients with distant metastasis.

Methods: Retrospective analysis was conducted using data from the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) database. Patients with distant metastasis [stage M1, according to the American Joint Committee on Cancer (AJCC) 7th edition] between January 2010 and December 2020 were included. Their characteristics, clinical profiles, and prognostic information were evaluated. Cox multifactorial survival analysis and Kaplan-Meier survival curves were used for statistical analysis.

Results: A total of 130 patients were included, with 78 (60%) receiving chemotherapy. Cox multivariate survival analysis revealed worse prognosis for Black individuals compared to White individuals (P<0.05). The median overall survival was 2 months for Black patients and 5 months for White patients. Chemotherapy significantly improved patient prognosis (P<0.05), while lung metastasis emerged as an independent risk factor (P<0.05). Kaplan-Meier survival curves confirmed the impact of lung metastasis and chemotherapy on overall survival. Patients with lung metastasis had lower survival rates (P<0.05), and those receiving chemotherapy had higher survival rates (P<0.05). Subgroup analysis based on age showed lower survival rates in patients aged 75 years or older compared to those below 75 years. Chemotherapy showed significant beneficial effects on the prognosis of patients below 75 years old, but no significant difference was observed in patients aged 75 years or above.

Conclusions: Chemotherapy improves the prognosis of cHCC-CCA patients with distant metastasis, especially for those under 75 years old. Black race and lung metastasis are poor prognostic factors.

背景:肝细胞胆管癌(cHCC-CCA)是一种罕见的肝癌,预后较差,通常在晚期确诊。对有远处转移的 cHCC-CCA 的治疗仍具有挑战性,而且预后因素尚未明确。本研究旨在调查有远处转移的cHCC-CCA患者的预后因素和治疗结果:方法:利用美国国家癌症研究所监测、流行病学和最终结果(SEER)数据库的数据进行回顾性分析。研究纳入了 2010 年 1 月至 2020 年 12 月期间的远处转移患者(根据美国癌症联合委员会(AJCC)第 7 版,分期为 M1)。对他们的特征、临床概况和预后信息进行了评估。统计分析采用 Cox 多因素生存分析和 Kaplan-Meier 生存曲线:共纳入130名患者,其中78人(60%)接受了化疗。Cox多因素生存分析显示,黑人的预后比白人差(结论:化疗可改善癌症患者的预后:化疗可改善有远处转移的 cHCC-CCA 患者的预后,尤其是 75 岁以下的患者。黑人和肺转移是不良预后因素。
{"title":"Treatments and prognostic outcomes of combined hepatocellular-cholangiocarcinoma with distant metastasis: an analysis based on SEER data.","authors":"Leilei Zhang, Yunxi Lu, Yuting Chen, Xiangling Lu, Xiaoli Lao","doi":"10.21037/tcr-24-447","DOIUrl":"10.21037/tcr-24-447","url":null,"abstract":"<p><strong>Background: </strong>Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a rare liver cancer with a poor prognosis, often diagnosed at an advanced stage. The management of cHCC-CCA with distant metastasis remains challenging, and prognostic factors are not well-defined. This study aimed to investigate prognostic factors and treatment outcomes for cHCC-CCA patients with distant metastasis.</p><p><strong>Methods: </strong>Retrospective analysis was conducted using data from the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) database. Patients with distant metastasis [stage M1, according to the American Joint Committee on Cancer (AJCC) 7<sup>th</sup> edition] between January 2010 and December 2020 were included. Their characteristics, clinical profiles, and prognostic information were evaluated. Cox multifactorial survival analysis and Kaplan-Meier survival curves were used for statistical analysis.</p><p><strong>Results: </strong>A total of 130 patients were included, with 78 (60%) receiving chemotherapy. Cox multivariate survival analysis revealed worse prognosis for Black individuals compared to White individuals (P<0.05). The median overall survival was 2 months for Black patients and 5 months for White patients. Chemotherapy significantly improved patient prognosis (P<0.05), while lung metastasis emerged as an independent risk factor (P<0.05). Kaplan-Meier survival curves confirmed the impact of lung metastasis and chemotherapy on overall survival. Patients with lung metastasis had lower survival rates (P<0.05), and those receiving chemotherapy had higher survival rates (P<0.05). Subgroup analysis based on age showed lower survival rates in patients aged 75 years or older compared to those below 75 years. Chemotherapy showed significant beneficial effects on the prognosis of patients below 75 years old, but no significant difference was observed in patients aged 75 years or above.</p><p><strong>Conclusions: </strong>Chemotherapy improves the prognosis of cHCC-CCA patients with distant metastasis, especially for those under 75 years old. Black race and lung metastasis are poor prognostic factors.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319941/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983366","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
Clinical research progress of telomerase targeted cancer immunotherapy: a literature review. 端粒酶靶向癌症免疫疗法的临床研究进展:文献综述。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-07-31 Epub Date: 2024-07-17 DOI: 10.21037/tcr-24-196
Yu Wang, Xiaoying Zhang, Guangming Chen, Mingzhe Shao

Background and objective: Telomerase is activated or overexpressed in 85-90% of tumors, which maintains the length of telomere and has become an important anti-cancer target. Increasing clinical and preclinical data suggest that telomerase-targeted cancer immunotherapy could achieve effective killing of tumor cells in vivo. This article reviews the research progress of telomerase targeted cancer immunotherapy in clinical and pre-clinical trials, aiming to provide a reference for further clinical research and treatment of cancers.

Methods: We investigated the research progress of telomerase immunotherapy in the last 20 years from four electronic databases.

Key content and findings: Telomerase-targeted immunotherapies have been developed with the arising of a new era in immuno-oncology, including peptide vaccines, DNA vaccines, dendritic cells (DCs), adoptive cell transfer (ACT) therapies, antibodies, etc. Some of them have been approved for undergoing clinical trials by the Food and Drug Administration (FDA) for the treatment of various cancers, such as pancreatic cancer, non-small cell lung cancer, melanoma, leukaemia. Of all the treatment modalities, vaccines are the primary treatment methods, some of which have been even entered into phase III clinical trials. The main clinical application direction of telomerase vaccine is the combination with other drugs and treatment modalities, including combination with other vaccines targeting human telomerase reverse transcriptase (hTERT), traditional chemotherapy drugs and immunosuppressors. We also summarized the recent findings of immunotherapy targeting hTERT, focusing on various vaccines and the current status of associated clinical trials. We further discussed the advantages, disadvantages and potential developmental directions of various telomerase-targeted immunotherapies.

Conclusions: Telomerase-targeted cancer immunotherapy has promising prospects in improving patient survival expectancy. This review may provide data support and design ideas for all researchers and pharmaceutical enterprises in this field.

背景和目的:端粒酶在85%-90%的肿瘤中被激活或过度表达,它能维持端粒的长度,已成为重要的抗癌靶点。越来越多的临床和临床前数据表明,以端粒酶为靶点的癌症免疫疗法可以在体内有效杀伤肿瘤细胞。本文回顾了端粒酶靶向癌症免疫疗法在临床和临床前试验中的研究进展,旨在为进一步的临床研究和癌症治疗提供参考:我们从四个电子数据库中调查了近20年来端粒酶免疫疗法的研究进展:随着免疫肿瘤学新时代的到来,端粒酶靶向免疫疗法应运而生,包括多肽疫苗、DNA疫苗、树突状细胞(DCs)、采纳性细胞转移(ACT)疗法、抗体等。其中一些疗法已获得美国食品药品管理局(FDA)批准,正在进行临床试验,用于治疗各种癌症,如胰腺癌、非小细胞肺癌、黑色素瘤、白血病等。在所有治疗方式中,疫苗是最主要的治疗方法,其中一些甚至已进入 III 期临床试验。端粒酶疫苗的主要临床应用方向是与其他药物和治疗方式的联合应用,包括与其他靶向人端粒酶逆转录酶(hTERT)的疫苗、传统化疗药物和免疫抑制剂的联合应用。我们还总结了针对 hTERT 的免疫疗法的最新研究成果,重点介绍了各种疫苗和相关临床试验的现状。我们进一步讨论了各种端粒酶靶向免疫疗法的优缺点和潜在发展方向:端粒酶靶向癌症免疫疗法在提高患者预期生存率方面前景广阔。本综述可为该领域的所有研究人员和制药企业提供数据支持和设计思路。
{"title":"Clinical research progress of telomerase targeted cancer immunotherapy: a literature review.","authors":"Yu Wang, Xiaoying Zhang, Guangming Chen, Mingzhe Shao","doi":"10.21037/tcr-24-196","DOIUrl":"10.21037/tcr-24-196","url":null,"abstract":"<p><strong>Background and objective: </strong>Telomerase is activated or overexpressed in 85-90% of tumors, which maintains the length of telomere and has become an important anti-cancer target. Increasing clinical and preclinical data suggest that telomerase-targeted cancer immunotherapy could achieve effective killing of tumor cells <i>in vivo</i>. This article reviews the research progress of telomerase targeted cancer immunotherapy in clinical and pre-clinical trials, aiming to provide a reference for further clinical research and treatment of cancers.</p><p><strong>Methods: </strong>We investigated the research progress of telomerase immunotherapy in the last 20 years from four electronic databases.</p><p><strong>Key content and findings: </strong>Telomerase-targeted immunotherapies have been developed with the arising of a new era in immuno-oncology, including peptide vaccines, DNA vaccines, dendritic cells (DCs), adoptive cell transfer (ACT) therapies, antibodies, etc. Some of them have been approved for undergoing clinical trials by the Food and Drug Administration (FDA) for the treatment of various cancers, such as pancreatic cancer, non-small cell lung cancer, melanoma, leukaemia. Of all the treatment modalities, vaccines are the primary treatment methods, some of which have been even entered into phase III clinical trials. The main clinical application direction of telomerase vaccine is the combination with other drugs and treatment modalities, including combination with other vaccines targeting human telomerase reverse transcriptase (hTERT), traditional chemotherapy drugs and immunosuppressors. We also summarized the recent findings of immunotherapy targeting hTERT, focusing on various vaccines and the current status of associated clinical trials. We further discussed the advantages, disadvantages and potential developmental directions of various telomerase-targeted immunotherapies.</p><p><strong>Conclusions: </strong>Telomerase-targeted cancer immunotherapy has promising prospects in improving patient survival expectancy. This review may provide data support and design ideas for all researchers and pharmaceutical enterprises in this field.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":null,"pages":null},"PeriodicalIF":1.5,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319969/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983284","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
Translational 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