首页 > 最新文献

Anticancer research最新文献

英文 中文
Bladder Volume <200 ml During a Course of Moderate Hypofractionated Irradiation in Patients With Localized Prostate Cancer.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17458
Dirk Rades, Charlotte Kristiansen, Christine Vestergaard Madsen, Jan-Dirk Küter, Laura Splettstösser, Cathrin Thieme, Jon Cacicedo, Stefan Janssen

Background/aim: Hypo-fractionated radiotherapy (HF-RT) is gaining popularity in prostate cancer treatment. HF-RT can lead to cystitis, particularly in cases with small bladder volumes. This study evaluated the bladder volume during a course of moderate HF-RT. This knowledge is required for the protocol of a prospective trial.

Patients and methods: Seventy-six patients receiving HF-RT (20×3.0 Gy) for prostate cancer were retrospectively evaluated. The number of HF-RT sessions with a bladder volume <200 ml and corresponding risk factors were investigated.

Results: Mean and median numbers of sessions with a bladder volume <200 ml were 13.4 (±6.7) and 16.0 (interquartile range=8.0-19.0), respectively. Higher numbers of radiotherapy sessions with a bladder volume <200 ml were associated with a pre-radiotherapy volume <200 ml (p<0.001). Mean numbers of sessions with a bladder volume <200 ml were 16.0 (±5.5) in patients with a pre-radiotherapy bladder volume <200 ml and 7.9 (±5.9) in patients with a bladder volume ≥200 ml, respectively.

Conclusion: Bladder volume was <200 ml during many HF-RT sessions. Patients with a pre-radiotherapy bladder volume <200 ml may benefit from an app reminding them to drink water before their HF-RT sessions.

{"title":"Bladder Volume <200 ml During a Course of Moderate Hypofractionated Irradiation in Patients With Localized Prostate Cancer.","authors":"Dirk Rades, Charlotte Kristiansen, Christine Vestergaard Madsen, Jan-Dirk Küter, Laura Splettstösser, Cathrin Thieme, Jon Cacicedo, Stefan Janssen","doi":"10.21873/anticanres.17458","DOIUrl":"10.21873/anticanres.17458","url":null,"abstract":"<p><strong>Background/aim: </strong>Hypo-fractionated radiotherapy (HF-RT) is gaining popularity in prostate cancer treatment. HF-RT can lead to cystitis, particularly in cases with small bladder volumes. This study evaluated the bladder volume during a course of moderate HF-RT. This knowledge is required for the protocol of a prospective trial.</p><p><strong>Patients and methods: </strong>Seventy-six patients receiving HF-RT (20×3.0 Gy) for prostate cancer were retrospectively evaluated. The number of HF-RT sessions with a bladder volume <200 ml and corresponding risk factors were investigated.</p><p><strong>Results: </strong>Mean and median numbers of sessions with a bladder volume <200 ml were 13.4 (±6.7) and 16.0 (interquartile range=8.0-19.0), respectively. Higher numbers of radiotherapy sessions with a bladder volume <200 ml were associated with a pre-radiotherapy volume <200 ml (p<0.001). Mean numbers of sessions with a bladder volume <200 ml were 16.0 (±5.5) in patients with a pre-radiotherapy bladder volume <200 ml and 7.9 (±5.9) in patients with a bladder volume ≥200 ml, respectively.</p><p><strong>Conclusion: </strong>Bladder volume was <200 ml during many HF-RT sessions. Patients with a pre-radiotherapy bladder volume <200 ml may benefit from an app reminding them to drink water before their HF-RT sessions.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"701-708"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073489","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 of Matrix Metalloproteinase-1 Promoter Genotypes With Endometriosis Risk.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17436
Po-Chuen Shieh, Hou-Yu Shih, Chin-Liang Chuang, Chia-Wen Tsai, Wen-Shin Chang, Meng-Gi Bau, Yun-Chi Wang, Te-Chun Hsia, DA-Tian Bau, Jai-Sing Yang

Background/aim: Over-expression of matrix metalloproteinase-1 (MMP-1) has been suggested as a biomarker for endometriosis. However, the genetic influence of MMP-1 in the pathogenesis of endometriosis remains unclear, with its role yet to be fully elucidated. This study aimed to investigate the association between MMP-1 rs1799750 promoter polymorphisms and the risk of developing endometriosis.

Patients and methods: This hospital-based case-control study included 203 women diagnosed with endometriosis and 636 age-matched controls. Genotyping of the MMP-1 rs1799750 polymorphism was conducted using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis.

Results: Among the patients with endometriosis, the distribution of genotypes 2G/2G, 2G/1G, and 1G/1G at MMP-1 rs1799750 was 52.7%, 41.4%, and 5.9%, respectively. This distribution significantly differed from that of the control group, which exhibited frequencies of 41.3%, 48.3%, and 10.4%, respectively (p for trend=0.0092). In the dominant model, carriers of the 2G/1G and 1G/1G genotypes had a reduced prevalence in the endometriosis group compared to 2G/2G carriers [odds ratio (OR)=0.63, 95% confidence interval (95%CI)=0.46-0.87, p=0.0058]. Additionally, the 1G allele frequency in the endometriosis group was 26.6%, significantly lower than the 34.5% observed in controls (OR=0.69, 95%CI=0.54-0.88, p=0.0037).

Conclusion: The 1G allele of MMP-1 rs1799750 is associated with reduced susceptibility to endometriosis in the Taiwanese population. These results highlight the potential of MMP-1 rs1799750 polymorphism as a protective genetic marker, warranting further investigations to explore its genotype-phenotype correlation and underlying biological mechanisms.

{"title":"Association of <i>Matrix Metalloproteinase-1</i> Promoter Genotypes With Endometriosis Risk.","authors":"Po-Chuen Shieh, Hou-Yu Shih, Chin-Liang Chuang, Chia-Wen Tsai, Wen-Shin Chang, Meng-Gi Bau, Yun-Chi Wang, Te-Chun Hsia, DA-Tian Bau, Jai-Sing Yang","doi":"10.21873/anticanres.17436","DOIUrl":"https://doi.org/10.21873/anticanres.17436","url":null,"abstract":"<p><strong>Background/aim: </strong>Over-expression of matrix metalloproteinase-1 (MMP-1) has been suggested as a biomarker for endometriosis. However, the genetic influence of MMP-1 in the pathogenesis of endometriosis remains unclear, with its role yet to be fully elucidated. This study aimed to investigate the association between MMP-1 rs1799750 promoter polymorphisms and the risk of developing endometriosis.</p><p><strong>Patients and methods: </strong>This hospital-based case-control study included 203 women diagnosed with endometriosis and 636 age-matched controls. Genotyping of the MMP-1 rs1799750 polymorphism was conducted using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis.</p><p><strong>Results: </strong>Among the patients with endometriosis, the distribution of genotypes 2G/2G, 2G/1G, and 1G/1G at MMP-1 rs1799750 was 52.7%, 41.4%, and 5.9%, respectively. This distribution significantly differed from that of the control group, which exhibited frequencies of 41.3%, 48.3%, and 10.4%, respectively (p for trend=0.0092). In the dominant model, carriers of the 2G/1G and 1G/1G genotypes had a reduced prevalence in the endometriosis group compared to 2G/2G carriers [odds ratio (OR)=0.63, 95% confidence interval (95%CI)=0.46-0.87, p=0.0058]. Additionally, the 1G allele frequency in the endometriosis group was 26.6%, significantly lower than the 34.5% observed in controls (OR=0.69, 95%CI=0.54-0.88, p=0.0037).</p><p><strong>Conclusion: </strong>The 1G allele of MMP-1 rs1799750 is associated with reduced susceptibility to endometriosis in the Taiwanese population. These results highlight the potential of MMP-1 rs1799750 polymorphism as a protective genetic marker, warranting further investigations to explore its genotype-phenotype correlation and underlying biological mechanisms.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"465-471"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073483","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
Recombinant Methioninase (rMETase) Synergistically Sensitizes Ivermectin-resistant MCF-7 Breast Cancer Cells 9.9 Fold to Low-dose Ivermectin.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17434
Sei Morinaga, Qinghong Han, Kohei Mizuta, Byung Mo Kang, Chihiro Hozumi, Michael Bouvet, Norio Yamamoto, Katsuhiro Hayashi, Hiroaki Kimura, Shinji Miwa, Kentaro Igarashi, Takashi Higuchi, Hiroyuki Tsuchiya, Satoru Demura, Robert M Hoffman

Background/aim: Ivermectin is a widely-used anti-parasitic agent and has shown early promise as an anticancer agent. Recombinant methioninase (rMETase) is a methionine-depleting enzyme targeting the methionine addiction of cancer and has broad efficacy against all tested cancer types. However, the combination efficacy of ivermectin and rMETase on breast cancer cells remains unexplored. The present study aimed to determine the synergistic efficacy of ivermectin and rMETase on MCF-7 human breast cancer cells in vitro.

Materials and methods: The IC10 of ivermectin and IC50 of rMETase were determined on MCF-7 cells using the WST-8 reagent to measure cell viability in vitro. MCF-7 cells were treated with four groups: untreated control; ivermectin alone (4.89 μM, IC10); rMETase alone (2.75 U/ml, IC50); and a combination of ivermectin (4.89 μM) and rMETase (2.75 U/ml). Cell viability was assessed 72 hours after treatment with the WST-8 reagent.

Results: Treatment with ivermectin (4.89 μM) did not significantly reduce the viability of MCF-7 cells. rMETase (2.75 U/ml) alone significantly reduced MCF-7 cell viability compared to the control group. The combination of ivermectin and rMETase resulted in a significantly greater reduction in cell viability than either agent alone, including a 9.9-fold greater efficacy than ivermectin alone, demonstrating synergistic efficacy (p<0.05).

Conclusion: The combination of ivermectin and rMETase had synergistic efficacy against MCF-7 breast cancer cells in vitro. The present findings suggest that the combination of ivermectin and rMETase is a promising strategy for breast cancer requiring further preclinical and clinical evaluation.

{"title":"Recombinant Methioninase (rMETase) Synergistically Sensitizes Ivermectin-resistant MCF-7 Breast Cancer Cells 9.9 Fold to Low-dose Ivermectin.","authors":"Sei Morinaga, Qinghong Han, Kohei Mizuta, Byung Mo Kang, Chihiro Hozumi, Michael Bouvet, Norio Yamamoto, Katsuhiro Hayashi, Hiroaki Kimura, Shinji Miwa, Kentaro Igarashi, Takashi Higuchi, Hiroyuki Tsuchiya, Satoru Demura, Robert M Hoffman","doi":"10.21873/anticanres.17434","DOIUrl":"https://doi.org/10.21873/anticanres.17434","url":null,"abstract":"<p><strong>Background/aim: </strong>Ivermectin is a widely-used anti-parasitic agent and has shown early promise as an anticancer agent. Recombinant methioninase (rMETase) is a methionine-depleting enzyme targeting the methionine addiction of cancer and has broad efficacy against all tested cancer types. However, the combination efficacy of ivermectin and rMETase on breast cancer cells remains unexplored. The present study aimed to determine the synergistic efficacy of ivermectin and rMETase on MCF-7 human breast cancer cells in vitro.</p><p><strong>Materials and methods: </strong>The IC<sub>10</sub> of ivermectin and IC<sub>50</sub> of rMETase were determined on MCF-7 cells using the WST-8 reagent to measure cell viability in vitro. MCF-7 cells were treated with four groups: untreated control; ivermectin alone (4.89 μM, IC<sub>10</sub>); rMETase alone (2.75 U/ml, IC<sub>50</sub>); and a combination of ivermectin (4.89 μM) and rMETase (2.75 U/ml). Cell viability was assessed 72 hours after treatment with the WST-8 reagent.</p><p><strong>Results: </strong>Treatment with ivermectin (4.89 μM) did not significantly reduce the viability of MCF-7 cells. rMETase (2.75 U/ml) alone significantly reduced MCF-7 cell viability compared to the control group. The combination of ivermectin and rMETase resulted in a significantly greater reduction in cell viability than either agent alone, including a 9.9-fold greater efficacy than ivermectin alone, demonstrating synergistic efficacy (p<0.05).</p><p><strong>Conclusion: </strong>The combination of ivermectin and rMETase had synergistic efficacy against MCF-7 breast cancer cells in vitro. The present findings suggest that the combination of ivermectin and rMETase is a promising strategy for breast cancer requiring further preclinical and clinical evaluation.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"451-455"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073505","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
A New PD-L1 Nanobody Enhances Cell Death in Lung Cancer In Vitro and In Vivo.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17442
Xiao-Tong Tan, Shi-Wei Huang, Yu-Chuan Lin, Fang-Yu Lin, DER-Yang Cho, Shao-Chih Chiu

Background/aim: The development of immune checkpoint blockade (ICB) agents targeting programmed death protein 1(PD-1), PD-1 ligand (PD-L1), cytotoxic T-lymphocyte- associated protein 4 (CTLA-4), and CD40 has gained increasing interest in clinical cancer treatment. Among these targets, blocking of PD-1 binding to its ligand PD-L1 managed to induce immune responses and inhibit tumor growth. In this work, we aimed to screen a specific PD-L1 nanobody against human PD-L1, derived through phage display assay, and further study its biological characteristics and antitumor ability.

Materials and methods: Specific PD-L1 nanobody was screened through phage display and its biological characteristics were explored by surface plasmon resonance (SPR) analysis. In addition, its cytotoxicity and antitumor ability was confirmed in vitro and in vivo.

Results: After all, an anti-PD-L1 nanobody with high specificity and affinity was generated. This PD-L1 nanobody was highly specific for human PD-L1 and had strong penetration ability due to its size. PD-L1 nanobody enhanced immune cell-killing ability by inhibiting the immune checkpoint and further activating innate response. Furthermore, this new PD-L1 nanobody also had high binding affinity, as shown by its use in western blotting, flow cytometry staining and immunofluorescence staining methods.

Conclusion: The new PD-L1 nanobody substantially improved upon the FDA-approved PD-L1 monoclonal antibody by surpassing the disadvantage of having large molecular weight (MW) and low tissue penetration. The cytotoxicity and antitumor ability of PD-L1 nanobody, in vitro and in vivo, also support its potential as a therapeutic agent for lung cancer immunotherapy.

{"title":"A New PD-L1 Nanobody Enhances Cell Death in Lung Cancer <i>In Vitro</i> and <i>In Vivo</i>.","authors":"Xiao-Tong Tan, Shi-Wei Huang, Yu-Chuan Lin, Fang-Yu Lin, DER-Yang Cho, Shao-Chih Chiu","doi":"10.21873/anticanres.17442","DOIUrl":"https://doi.org/10.21873/anticanres.17442","url":null,"abstract":"<p><strong>Background/aim: </strong>The development of immune checkpoint blockade (ICB) agents targeting programmed death protein 1(PD-1), PD-1 ligand (PD-L1), cytotoxic T-lymphocyte- associated protein 4 (CTLA-4), and CD40 has gained increasing interest in clinical cancer treatment. Among these targets, blocking of PD-1 binding to its ligand PD-L1 managed to induce immune responses and inhibit tumor growth. In this work, we aimed to screen a specific PD-L1 nanobody against human PD-L1, derived through phage display assay, and further study its biological characteristics and antitumor ability.</p><p><strong>Materials and methods: </strong>Specific PD-L1 nanobody was screened through phage display and its biological characteristics were explored by surface plasmon resonance (SPR) analysis. In addition, its cytotoxicity and antitumor ability was confirmed in vitro and in vivo.</p><p><strong>Results: </strong>After all, an anti-PD-L1 nanobody with high specificity and affinity was generated. This PD-L1 nanobody was highly specific for human PD-L1 and had strong penetration ability due to its size. PD-L1 nanobody enhanced immune cell-killing ability by inhibiting the immune checkpoint and further activating innate response. Furthermore, this new PD-L1 nanobody also had high binding affinity, as shown by its use in western blotting, flow cytometry staining and immunofluorescence staining methods.</p><p><strong>Conclusion: </strong>The new PD-L1 nanobody substantially improved upon the FDA-approved PD-L1 monoclonal antibody by surpassing the disadvantage of having large molecular weight (MW) and low tissue penetration. The cytotoxicity and antitumor ability of PD-L1 nanobody, in vitro and in vivo, also support its potential as a therapeutic agent for lung cancer immunotherapy.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"535-547"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073507","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
Noncancerous Exosomes Establish a Growth Promoting Paracrine Effect on Triple Negative Breast Cancer Cells.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17431
Letitia A Yearby, Manasa Kotina, Afia Ohemeng, Bipika Banjara, Mounika Pamukuntla, Paige Roberts, Jasmine Johnson, Kaitlyn Perkins, Josiah Ochieng, Jillian L Pope, Syreeta L Tilghman, Kitani Parker Lemieux

Background/aim: Previous studies have demonstrated that breast cancer cells secrete exosomes into the tumor microenvironment, promoting tumor progression. However, the paracrine influence of noncancerous breast epithelial cells on the growth of triple-negative breast cancer (TNBC) cells has largely been overlooked. We hypothesize that exosomes from noncancerous breast epithelial cells are secreted into the tumor microenvironment, stimulating TNBC growth.

Materials and methods: Exosome-containing media were prepared using exosomes isolated from triple-negative patient-derived xenografts (PDX) or noncancerous MCF-10A breast epithelial cells and used to treat MCF-7 or TNBC cells. Exosome-containing media from MCF-10A cells were characterized using ELISA. Subsequently, MDA-MB-231 and MDA-MB-468 cells treated with the MCF-10A exosome-containing media, and their impact on proliferation, migration, protein expression and gene expression were analyzed using Alamar blue assays, wound healing assays, western blotting, immunofluorescence, and gene expression arrays, respectively.

Results: Exosomes extracted from the PDX and MCF-10A cells stimulated the growth of all examined cell lines. The MCF-10A exosome-containing media expressed CD9 and CD63; however, the MDA-MB-231 and MDA-MB-468 cells treated with these media exhibited differential expression of these proteins. Exposure of MDA-MB-231 and MDA-MB-468 cells to the MCF-10A exosome-containing media stimulated their growth and migration. Exposure of MDA-MB-231 cells to MCF-10A exosome-containing media caused down-regulation of genes involved in cell-cell adhesion, DNA damage response, epithelial-mesenchymal transition drivers, tumor suppression, and up-regulation of the MYC oncogene.

Conclusion: Secreted factors from noncancerous cells, identified as exosomes, induce cancer cell proliferation and prime the tumor microenvironment by enhancing disease progression-associated pathways.

{"title":"Noncancerous Exosomes Establish a Growth Promoting Paracrine Effect on Triple Negative Breast Cancer Cells.","authors":"Letitia A Yearby, Manasa Kotina, Afia Ohemeng, Bipika Banjara, Mounika Pamukuntla, Paige Roberts, Jasmine Johnson, Kaitlyn Perkins, Josiah Ochieng, Jillian L Pope, Syreeta L Tilghman, Kitani Parker Lemieux","doi":"10.21873/anticanres.17431","DOIUrl":"https://doi.org/10.21873/anticanres.17431","url":null,"abstract":"<p><strong>Background/aim: </strong>Previous studies have demonstrated that breast cancer cells secrete exosomes into the tumor microenvironment, promoting tumor progression. However, the paracrine influence of noncancerous breast epithelial cells on the growth of triple-negative breast cancer (TNBC) cells has largely been overlooked. We hypothesize that exosomes from noncancerous breast epithelial cells are secreted into the tumor microenvironment, stimulating TNBC growth.</p><p><strong>Materials and methods: </strong>Exosome-containing media were prepared using exosomes isolated from triple-negative patient-derived xenografts (PDX) or noncancerous MCF-10A breast epithelial cells and used to treat MCF-7 or TNBC cells. Exosome-containing media from MCF-10A cells were characterized using ELISA. Subsequently, MDA-MB-231 and MDA-MB-468 cells treated with the MCF-10A exosome-containing media, and their impact on proliferation, migration, protein expression and gene expression were analyzed using Alamar blue assays, wound healing assays, western blotting, immunofluorescence, and gene expression arrays, respectively.</p><p><strong>Results: </strong>Exosomes extracted from the PDX and MCF-10A cells stimulated the growth of all examined cell lines. The MCF-10A exosome-containing media expressed CD9 and CD63; however, the MDA-MB-231 and MDA-MB-468 cells treated with these media exhibited differential expression of these proteins. Exposure of MDA-MB-231 and MDA-MB-468 cells to the MCF-10A exosome-containing media stimulated their growth and migration. Exposure of MDA-MB-231 cells to MCF-10A exosome-containing media caused down-regulation of genes involved in cell-cell adhesion, DNA damage response, epithelial-mesenchymal transition drivers, tumor suppression, and up-regulation of the MYC oncogene.</p><p><strong>Conclusion: </strong>Secreted factors from noncancerous cells, identified as exosomes, induce cancer cell proliferation and prime the tumor microenvironment by enhancing disease progression-associated pathways.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"419-431"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073352","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
Unusual Presentation of Advanced Urothelial Cancer in a Young Patient.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17448
Daniela Guevara, Nara Shin, Alexandra Boiko, Ivan Valiev, Ahmed G Elsaeed, Juan Miguel Mosquera, Majd Al Assaad, Jyothi Manohar, Michael Sigouros, Alisa Zaichikova, Victoria Fomchenkova, Leysan Yunusova, Sofia Smirnova, Olivier Elemento, David Nanus, Cora N Sternberg

Background/aim: Urothelial carcinoma, common in older adults, is rare in younger populations and even less common in the prostatic urethra. Advanced disease is typically managed with platinum-based chemotherapy, immune checkpoint inhibitors, and targeted therapies. However, rare presentations in young patients with aggressive disease highlight the need for innovative and personalized treatment strategies.

Case report: This case report presents a rare instance of metastatic urothelial carcinoma originating in the prostatic urethra of a 37-year-old male. Initial symptoms led to diagnosis through imaging, biopsy, and genetic profiling, revealing mutations in TP53 and RB1. The patient underwent multiple treatments, including dose-dense chemotherapy, pembrolizumab immunotherapy, and targeted antibody-drug conjugates (Enfortumab Vedotin and Sacituzumab Govitecan). Despite aggressive therapies, disease management remained challenging, leading to experimental treatments, including a personalized vaccine.

Conclusion: This case underscores the importance of precision medicine and the need for innovative treatment options for rare and aggressive cancers.

{"title":"Unusual Presentation of Advanced Urothelial Cancer in a Young Patient.","authors":"Daniela Guevara, Nara Shin, Alexandra Boiko, Ivan Valiev, Ahmed G Elsaeed, Juan Miguel Mosquera, Majd Al Assaad, Jyothi Manohar, Michael Sigouros, Alisa Zaichikova, Victoria Fomchenkova, Leysan Yunusova, Sofia Smirnova, Olivier Elemento, David Nanus, Cora N Sternberg","doi":"10.21873/anticanres.17448","DOIUrl":"https://doi.org/10.21873/anticanres.17448","url":null,"abstract":"<p><strong>Background/aim: </strong>Urothelial carcinoma, common in older adults, is rare in younger populations and even less common in the prostatic urethra. Advanced disease is typically managed with platinum-based chemotherapy, immune checkpoint inhibitors, and targeted therapies. However, rare presentations in young patients with aggressive disease highlight the need for innovative and personalized treatment strategies.</p><p><strong>Case report: </strong>This case report presents a rare instance of metastatic urothelial carcinoma originating in the prostatic urethra of a 37-year-old male. Initial symptoms led to diagnosis through imaging, biopsy, and genetic profiling, revealing mutations in TP53 and RB1. The patient underwent multiple treatments, including dose-dense chemotherapy, pembrolizumab immunotherapy, and targeted antibody-drug conjugates (Enfortumab Vedotin and Sacituzumab Govitecan). Despite aggressive therapies, disease management remained challenging, leading to experimental treatments, including a personalized vaccine.</p><p><strong>Conclusion: </strong>This case underscores the importance of precision medicine and the need for innovative treatment options for rare and aggressive cancers.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"613-618"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073487","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
Prognostic Stratification Using Early Prostate-specific Antigen Kinetics in Men With Metastatic Hormone-sensitive Prostate Cancer.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17463
Tasuku Hiroshige, Hiroki Suekane, Takaho Tokunaga, Mami Uegaki, Masahito Iwashita, Hiroki Taura, Taishi Hirano, Tomotaro Mitani, Mitsunori Matsuo, Tsukasa Igawa

Background/aim: The prognostic significance of prostate-specific antigen (PSA) kinetics in metastatic castration-sensitive prostate cancer (mCSPC) patients treated with upfront therapy remains unclear. This study investigated the correlation between early PSA response and clinical outcomes in patients with mCSPC who received upfront therapy.

Patients and methods: We analyzed 106 patients with mCSPC who received upfront therapies [abiraterone acetate (ABI), enzalutamide (ENZ), apalutamide (APA), and docetaxel (DOC)] at Kurume University Hospital and its affiliated hospitals.

Results: Thirty-nine, 15, 38, and 14 patients were treated with ABI, DOC, ENZ, and APA, respectively. Among the total number of patients, 67 met the criteria for high-volume disease. Additionally, 83 patients were categorized as high risk. Patients with a PSA decline rate of ≥90% at 4 and 12 weeks post-upfront therapies had a significantly longer time to develop CRPC than those with a PSA decline of <90%. PSA cutoff values >26 ng/ml at 4 weeks post-upfront therapies and a PSA decline rate of ≥90% at 12 weeks post-upfront therapies were independent predictors of poor prognosis. Furthermore, patients were stratified into three groups based on PSA levels at 4 weeks and PSA decline rate at 12 weeks.

Conclusion: A larger PSA decline within three months of initiating upfront therapy is significantly associated with a longer time to CRPC in patients with mCSPC treated with upfront therapy. A combination of early PSA kinetics can be used to stratify the risk of CRPC progression in patients with mCSPC treated with upfront therapies.

{"title":"Prognostic Stratification Using Early Prostate-specific Antigen Kinetics in Men With Metastatic Hormone-sensitive Prostate Cancer.","authors":"Tasuku Hiroshige, Hiroki Suekane, Takaho Tokunaga, Mami Uegaki, Masahito Iwashita, Hiroki Taura, Taishi Hirano, Tomotaro Mitani, Mitsunori Matsuo, Tsukasa Igawa","doi":"10.21873/anticanres.17463","DOIUrl":"https://doi.org/10.21873/anticanres.17463","url":null,"abstract":"<p><strong>Background/aim: </strong>The prognostic significance of prostate-specific antigen (PSA) kinetics in metastatic castration-sensitive prostate cancer (mCSPC) patients treated with upfront therapy remains unclear. This study investigated the correlation between early PSA response and clinical outcomes in patients with mCSPC who received upfront therapy.</p><p><strong>Patients and methods: </strong>We analyzed 106 patients with mCSPC who received upfront therapies [abiraterone acetate (ABI), enzalutamide (ENZ), apalutamide (APA), and docetaxel (DOC)] at Kurume University Hospital and its affiliated hospitals.</p><p><strong>Results: </strong>Thirty-nine, 15, 38, and 14 patients were treated with ABI, DOC, ENZ, and APA, respectively. Among the total number of patients, 67 met the criteria for high-volume disease. Additionally, 83 patients were categorized as high risk. Patients with a PSA decline rate of ≥90% at 4 and 12 weeks post-upfront therapies had a significantly longer time to develop CRPC than those with a PSA decline of <90%. PSA cutoff values >26 ng/ml at 4 weeks post-upfront therapies and a PSA decline rate of ≥90% at 12 weeks post-upfront therapies were independent predictors of poor prognosis. Furthermore, patients were stratified into three groups based on PSA levels at 4 weeks and PSA decline rate at 12 weeks.</p><p><strong>Conclusion: </strong>A larger PSA decline within three months of initiating upfront therapy is significantly associated with a longer time to CRPC in patients with mCSPC treated with upfront therapy. A combination of early PSA kinetics can be used to stratify the risk of CRPC progression in patients with mCSPC treated with upfront therapies.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"751-759"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073494","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
Safety and Survival Benefit of Adjuvant Chemotherapy for Elderly Patients With Stage II/III Colon Cancer.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17468
In Jun Yang, Dong Ha Kim, Kyung-Ha Lee, Ji Yeon Kim

Background/aim: To evaluate the safety and survival benefits of adjuvant chemotherapy (AC) in elderly patients who underwent radical resection for stage II/III colon cancer.

Patients and methods: This retrospective study included patients aged >70 years treated at a tertiary hospital between January 2012 and December 2017. We evaluated the clinical and pathological characteristics and adverse events of chemotherapy. The 5-year overall survival (OS) and disease-free survival (DFS) of the surgery-only (SO) and AC groups were compared by stage using the Kaplan-Meier method and Cox-regression analysis.

Results: Of the 163 patients included in the study, 75 were diagnosed with stage II cancer, with 43 patients in the SO group and 32 in the AC group. A total of 88 patients were diagnosed with stage III cancer, including 20 in the SO group and 68 in the AC group. Patients with stage II disease in the SO group were older, with less frequent venous invasion than the AC group. Comorbidities, tumor location, and surgical methods did not differ. In stage III, age, comorbidities, tumor location, surgical methods, and pathological outcomes did not differ. The 5-year OS and DFS did not differ significantly in those with stage II disease but were significantly better in the AC than the SO group in stage III cases (48.2% vs. 71.6%, p=0.012; and 42.6% vs. 60.0%, p=0.029).

Conclusion: AC may provide a survival advantage in elderly patients with stage III colon cancer.

{"title":"Safety and Survival Benefit of Adjuvant Chemotherapy for Elderly Patients With Stage II/III Colon Cancer.","authors":"In Jun Yang, Dong Ha Kim, Kyung-Ha Lee, Ji Yeon Kim","doi":"10.21873/anticanres.17468","DOIUrl":"https://doi.org/10.21873/anticanres.17468","url":null,"abstract":"<p><strong>Background/aim: </strong>To evaluate the safety and survival benefits of adjuvant chemotherapy (AC) in elderly patients who underwent radical resection for stage II/III colon cancer.</p><p><strong>Patients and methods: </strong>This retrospective study included patients aged >70 years treated at a tertiary hospital between January 2012 and December 2017. We evaluated the clinical and pathological characteristics and adverse events of chemotherapy. The 5-year overall survival (OS) and disease-free survival (DFS) of the surgery-only (SO) and AC groups were compared by stage using the Kaplan-Meier method and Cox-regression analysis.</p><p><strong>Results: </strong>Of the 163 patients included in the study, 75 were diagnosed with stage II cancer, with 43 patients in the SO group and 32 in the AC group. A total of 88 patients were diagnosed with stage III cancer, including 20 in the SO group and 68 in the AC group. Patients with stage II disease in the SO group were older, with less frequent venous invasion than the AC group. Comorbidities, tumor location, and surgical methods did not differ. In stage III, age, comorbidities, tumor location, surgical methods, and pathological outcomes did not differ. The 5-year OS and DFS did not differ significantly in those with stage II disease but were significantly better in the AC than the SO group in stage III cases (48.2% vs. 71.6%, p=0.012; and 42.6% vs. 60.0%, p=0.029).</p><p><strong>Conclusion: </strong>AC may provide a survival advantage in elderly patients with stage III colon cancer.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"797-809"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073509","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
Enhanced Potential of Durvalumab in the Initial Treatment of Advanced Biliary Tract Cancer.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17456
Jungo Yasuda, Hironori Shiozaki, Taro Sakamoto, Yasuro Futagawa, Tomoyoshi Okamoto, Toru Ikegami

Background/aim: The prognosis of biliary tract cancer is extremely poor, with a 5-year survival rate of 20%. Surgery is the only treatment that can be expected to cure biliary tract cancer, but because many cases are unresectable or recurrent, chemotherapy has become the standard treatment. The effects of first-line administration of durvalumab have not been explored. This study examined whether durvalumab has an additive effect in the first line.

Patients and methods: Twenty-three patients who were diagnosed with recurrent or non-resected biliary tract cancer requiring anticancer chemotherapy were recruited. Three of these cases were excluded because they had only received one course of durvalumab. We retrospectively collected clinical and laboratory data. Progression-free survival (PFS) and overall survival (OS) were compared between patients who received durvalumab as first-line therapy (first group, FG) and those who received it as second-line or later therapy (second group, SG). PFS and OS were also compared in durvalumab-treated patients aged 75 years and older (older group) and in younger patients. Immune-related adverse events (irAEs) were graded using the Common Terminology Criteria for Adverse Events (CTCAE v5) based on the clinical notation available in the patient charts.

Results: Kaplan-Meier curves showed that SG was significantly associated with worse PFS (p=0.018), and the FG group also showed significantly prolonged OS (p=0.030). In addition, PFS from the start of durvalumab treatment was significantly longer in the older group compared to the younger group. However, no significant difference in OS was observed between the two groups.

Conclusion: Durvalumab appears to contribute to prolonged PFS and OS when administered as an initial treatment. It may also contribute to improved outcomes in older patients with biliary tract cancer.

{"title":"Enhanced Potential of Durvalumab in the Initial Treatment of Advanced Biliary Tract Cancer.","authors":"Jungo Yasuda, Hironori Shiozaki, Taro Sakamoto, Yasuro Futagawa, Tomoyoshi Okamoto, Toru Ikegami","doi":"10.21873/anticanres.17456","DOIUrl":"https://doi.org/10.21873/anticanres.17456","url":null,"abstract":"<p><strong>Background/aim: </strong>The prognosis of biliary tract cancer is extremely poor, with a 5-year survival rate of 20%. Surgery is the only treatment that can be expected to cure biliary tract cancer, but because many cases are unresectable or recurrent, chemotherapy has become the standard treatment. The effects of first-line administration of durvalumab have not been explored. This study examined whether durvalumab has an additive effect in the first line.</p><p><strong>Patients and methods: </strong>Twenty-three patients who were diagnosed with recurrent or non-resected biliary tract cancer requiring anticancer chemotherapy were recruited. Three of these cases were excluded because they had only received one course of durvalumab. We retrospectively collected clinical and laboratory data. Progression-free survival (PFS) and overall survival (OS) were compared between patients who received durvalumab as first-line therapy (first group, FG) and those who received it as second-line or later therapy (second group, SG). PFS and OS were also compared in durvalumab-treated patients aged 75 years and older (older group) and in younger patients. Immune-related adverse events (irAEs) were graded using the Common Terminology Criteria for Adverse Events (CTCAE v5) based on the clinical notation available in the patient charts.</p><p><strong>Results: </strong>Kaplan-Meier curves showed that SG was significantly associated with worse PFS (p=0.018), and the FG group also showed significantly prolonged OS (p=0.030). In addition, PFS from the start of durvalumab treatment was significantly longer in the older group compared to the younger group. However, no significant difference in OS was observed between the two groups.</p><p><strong>Conclusion: </strong>Durvalumab appears to contribute to prolonged PFS and OS when administered as an initial treatment. It may also contribute to improved outcomes in older patients with biliary tract cancer.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"685-690"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073214","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
Camptothecin Triggers Apoptosis in Human and Mouse Drug-resistant Glioblastoma Cells via ROS-mediated Activation of the p53-p21-CD1/CDK2-E2F1-Bcl-xL Signaling Axis.
IF 1.6 4区 医学 Q4 ONCOLOGY Pub Date : 2025-02-01 DOI: 10.21873/anticanres.17444
Gong-Jhe Wu, Jui-Tai Chen, Yih-Giun Cherng, Chien-Ju Lin, Shing-Hwa Liu, Ruei-Ming Chen

Background/aim: Glioblastoma multiforme (GBM) is the most aggressive brain tumor. Temozolomide (TMZ) is the first-line treatment for GBM. However, most patients with GBM develop drug resistance. Our previous study showed the effects of camptothecin (CPT) and CRLX101, a nanoparticle of CPT, in suppressing GBM growth by targeting drug-sensitive glioblastoma cells. This study evaluated the effects of CPT on drug-resistant glioblastoma cells and explored the underlying molecular mechanisms.

Materials and methods: Expression of type I topoisomerase (Topo-1) gene in GBM was analyzed using the UALCAN database. Human U87MG-R and mouse GL261-R TMZ-resistant glioblastoma cells were developed. After CPT treatment, apoptotic events were successively determined. The role of the p53-p21-cyclin D1 (CD1)/cyclin-dependent kinase 6 (CDK6)-E2F1-Bcl-xL signaling axis was subsequently investigated.

Results: The expression of Topo-1 gene was up-regulated in human GBM compared to normal human brains. Treatment of human U87MG-R cells with CPT decreased cell viability. Sequentially, exposure to CPT led to activation of caspase-3, fragmentation of chromosomal DNA, and cell apoptosis. Furthermore, intracellular reactive oxygen species (iROS) were augmented following CPT treatment. Suppression of iROS production concurrently alleviated CPT-triggered apoptotic insults. CPT enhanced the levels of p53, phosphorylated p53, and p21. In contrast, levels of CDK6, CD1, E2F1, and Bcl-xL were decreased by CPT. Attenuating p53 transactivation activity using pifithrin-α also mitigated the CPT-induced apoptosis. The effects of CPT on killing drug-resistant glioblastoma cells were further confirmed in mouse GL261-R cells.

Conclusion: CPT could effectively induce apoptosis in drug-resistant glioblastoma cells via iROS-mediated activation of the p53-p21-CD1/CDK6-E2F1-Bcl-xL axis.

{"title":"Camptothecin Triggers Apoptosis in Human and Mouse Drug-resistant Glioblastoma Cells <i>via</i> ROS-mediated Activation of the p53-p21-CD1/CDK2-E2F1-Bcl-xL Signaling Axis.","authors":"Gong-Jhe Wu, Jui-Tai Chen, Yih-Giun Cherng, Chien-Ju Lin, Shing-Hwa Liu, Ruei-Ming Chen","doi":"10.21873/anticanres.17444","DOIUrl":"https://doi.org/10.21873/anticanres.17444","url":null,"abstract":"<p><strong>Background/aim: </strong>Glioblastoma multiforme (GBM) is the most aggressive brain tumor. Temozolomide (TMZ) is the first-line treatment for GBM. However, most patients with GBM develop drug resistance. Our previous study showed the effects of camptothecin (CPT) and CRLX101, a nanoparticle of CPT, in suppressing GBM growth by targeting drug-sensitive glioblastoma cells. This study evaluated the effects of CPT on drug-resistant glioblastoma cells and explored the underlying molecular mechanisms.</p><p><strong>Materials and methods: </strong>Expression of type I topoisomerase (Topo-1) gene in GBM was analyzed using the UALCAN database. Human U87MG-R and mouse GL261-R TMZ-resistant glioblastoma cells were developed. After CPT treatment, apoptotic events were successively determined. The role of the p53-p21-cyclin D1 (CD1)/cyclin-dependent kinase 6 (CDK6)-E2F1-Bcl-xL signaling axis was subsequently investigated.</p><p><strong>Results: </strong>The expression of Topo-1 gene was up-regulated in human GBM compared to normal human brains. Treatment of human U87MG-R cells with CPT decreased cell viability. Sequentially, exposure to CPT led to activation of caspase-3, fragmentation of chromosomal DNA, and cell apoptosis. Furthermore, intracellular reactive oxygen species (iROS) were augmented following CPT treatment. Suppression of iROS production concurrently alleviated CPT-triggered apoptotic insults. CPT enhanced the levels of p53, phosphorylated p53, and p21. In contrast, levels of CDK6, CD1, E2F1, and Bcl-xL were decreased by CPT. Attenuating p53 transactivation activity using pifithrin-α also mitigated the CPT-induced apoptosis. The effects of CPT on killing drug-resistant glioblastoma cells were further confirmed in mouse GL261-R cells.</p><p><strong>Conclusion: </strong>CPT could effectively induce apoptosis in drug-resistant glioblastoma cells via iROS-mediated activation of the p53-p21-CD1/CDK6-E2F1-Bcl-xL axis.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"45 2","pages":"565-577"},"PeriodicalIF":1.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143073492","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
期刊
Anticancer 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