Pub Date : 2024-09-28DOI: 10.1007/s00432-024-05963-4
Thomas Zeyen, Lea L Friker, Daniel Paech, Niklas Schaefer, Johannes Weller, Valentina Zschernack, Julian P Layer, Matthias Schneider, Anna-Laura Potthoff, Marit Bernhardt, Christine Sanders, Glen Kristiansen, Michael Hoelzel, Eleni Gkika, Alexander Radbruch, Torsten Pietsch, Ulrich Herrlinger, Christina Schaub
Purpose: Little is known about the effect of SARS-CoV-2 infection on glioblastoma (GBM) growth, metabolism, and prognosis. Immunological changes within GBM tissue are potentially symptomatic, underlining the urgent need for a better understanding of this phenomenon. To date, the complex underlying biology has not been fully elucidated. A decisive role of the tumor microenvironment (TME) and the components of the immune system acting within it is assumed.
Methods: Immunohistochemical staining of SARS-CoV-2 spike protein and immune cell infiltration of TME was performed on the tumor tissue of one patient. This patient developed hemiparesis 14 days after symptomatic SARS-CoV-2 infection, leading to tumor diagnosis. Subsequently and after biopsy, there was an unexpectedly good response to chemotherapy only. In looking for further evidence of the potential of SARS-CoV-2 to influence the course of GBM, two additional adult patients that had transient MRI changes and neurological deterioration following SARS-CoV-2 infection were evaluated.
Results: In the patient for whom neurological deterioration in the course of SARS-CoV-2 led to GBM diagnosis, immunohistochemistry revealed virus-specific protein accumulation in the tumor cells, microglial activation, and the formation of T-cell nodules. In the other two patients, the findings were compatible with symptomatic pseudoprogression that occurred in a temporal relationship with SARS-CoV-2 infection.
Conclusion: The results indicate a possible association between clinically relevant changes in GBM biology and SARS-CoV-2 infection, with histological confirmation of SARS-CoV-2-associated changes within the tumor tissue. The exact pathomechanism and underlying inflammatory pathways require further investigation.
{"title":"Transient MRI changes and neurological deterioration in glioblastoma upon SARS-CoV-2 infection.","authors":"Thomas Zeyen, Lea L Friker, Daniel Paech, Niklas Schaefer, Johannes Weller, Valentina Zschernack, Julian P Layer, Matthias Schneider, Anna-Laura Potthoff, Marit Bernhardt, Christine Sanders, Glen Kristiansen, Michael Hoelzel, Eleni Gkika, Alexander Radbruch, Torsten Pietsch, Ulrich Herrlinger, Christina Schaub","doi":"10.1007/s00432-024-05963-4","DOIUrl":"10.1007/s00432-024-05963-4","url":null,"abstract":"<p><strong>Purpose: </strong>Little is known about the effect of SARS-CoV-2 infection on glioblastoma (GBM) growth, metabolism, and prognosis. Immunological changes within GBM tissue are potentially symptomatic, underlining the urgent need for a better understanding of this phenomenon. To date, the complex underlying biology has not been fully elucidated. A decisive role of the tumor microenvironment (TME) and the components of the immune system acting within it is assumed.</p><p><strong>Methods: </strong>Immunohistochemical staining of SARS-CoV-2 spike protein and immune cell infiltration of TME was performed on the tumor tissue of one patient. This patient developed hemiparesis 14 days after symptomatic SARS-CoV-2 infection, leading to tumor diagnosis. Subsequently and after biopsy, there was an unexpectedly good response to chemotherapy only. In looking for further evidence of the potential of SARS-CoV-2 to influence the course of GBM, two additional adult patients that had transient MRI changes and neurological deterioration following SARS-CoV-2 infection were evaluated.</p><p><strong>Results: </strong>In the patient for whom neurological deterioration in the course of SARS-CoV-2 led to GBM diagnosis, immunohistochemistry revealed virus-specific protein accumulation in the tumor cells, microglial activation, and the formation of T-cell nodules. In the other two patients, the findings were compatible with symptomatic pseudoprogression that occurred in a temporal relationship with SARS-CoV-2 infection.</p><p><strong>Conclusion: </strong>The results indicate a possible association between clinically relevant changes in GBM biology and SARS-CoV-2 infection, with histological confirmation of SARS-CoV-2-associated changes within the tumor tissue. The exact pathomechanism and underlying inflammatory pathways require further investigation.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"437"},"PeriodicalIF":2.7,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438616/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142347237","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-28DOI: 10.1007/s00432-024-05959-0
Yauhen Lizunou, Anna-Laura Potthoff, Niklas Schäfer, Andreas Waha, Valeri Borger, Ulrich Herrlinger, Hartmut Vatter, Patrick Schuss, Matthias Schneider
Purpose: The rarity of cerebellar glioblastoma presents a significant challenge in clinical practice due to the lack of extensive prognostic data on long-term survival rates, rendering it an underrepresented entity compared to its supratentorial counterpart. This study aims to analyze potential differences in survival outcome between patients with cerebellar and supratentorial glioblastomas.
Methods: From 2009 to 2020, 8 patients underwent surgical treatment for cerebellar glioblastoma at the authors' institution. These patients were individually matched with a cohort of 205 consecutive patients from our institutional database with supratentorial glioblastoma, taking into account key prognostic parameters. Progression-free survival (PFS) and overall survival (OS) rates were compared. Additionally, we performed a systematic literature review to compile further survival data on cerebellar glioblastoma patients.
Results: The median OS for cerebellar glioblastoma patients was 18 months (95% CI 11-25). The balanced matched-pair analysis showed no significant difference in survival when compared to patients with supratentorial glioblastoma, exhibiting a median OS of 23 months (95% CI 0-62) (p = 0.63). Respective values for PFS were 8 months (95% CI 4-12) for cerebellar and 7 months (95% CI 0-16) for supratentorial glioblastoma (p = 0.2). The systematic review revealed that median OS for cerebellar glioblastoma in current literature ranges from 7 to 21 months.
Conclusions: The present findings indicate that patients with supra- and infratentorial glioblastoma do not significantly differ in regard to survival outcome parameters. This similarity in prognosis might encourage clinicians to consider surgical interventions for both supra- and infratentorial glioblastoma in a similar manner.
目的:小脑胶质母细胞瘤的罕见性为临床实践带来了巨大挑战,因为缺乏有关长期存活率的广泛预后数据,使其与幕上胶质母细胞瘤相比代表性不足。本研究旨在分析小脑胶质母细胞瘤和幕上胶质母细胞瘤患者生存结果的潜在差异:2009年至2020年,作者所在机构共对8名小脑胶质母细胞瘤患者进行了手术治疗。考虑到主要预后参数,我们将这些患者与本机构数据库中的205名连续的幕上胶质母细胞瘤患者进行了单独配对。比较了无进展生存率(PFS)和总生存率(OS)。此外,我们还进行了系统性文献回顾,以进一步收集小脑胶质母细胞瘤患者的生存数据:小脑胶质母细胞瘤患者的中位生存期为18个月(95% CI 11-25)。平衡配对分析显示,小脑胶质母细胞瘤患者的中位生存期为23个月(95% CI 0-62)(P = 0.63),与幕上胶质母细胞瘤患者相比,小脑胶质母细胞瘤患者的生存期无明显差异。小脑和幕上胶质母细胞瘤的PFS分别为8个月(95% CI 4-12)和7个月(95% CI 0-16)(P = 0.2)。系统综述显示,目前文献中小脑胶质母细胞瘤的中位OS为7至21个月:本研究结果表明,幕上和幕下胶质母细胞瘤患者的生存结果参数没有明显差异。这种相似的预后可能会鼓励临床医生以类似的方式考虑对脑室内上部和脑室内下部胶质母细胞瘤进行手术干预。
{"title":"Cerebellar glioblastoma in adults: a comparative single-center matched pair analysis and systematic review of the literature.","authors":"Yauhen Lizunou, Anna-Laura Potthoff, Niklas Schäfer, Andreas Waha, Valeri Borger, Ulrich Herrlinger, Hartmut Vatter, Patrick Schuss, Matthias Schneider","doi":"10.1007/s00432-024-05959-0","DOIUrl":"10.1007/s00432-024-05959-0","url":null,"abstract":"<p><strong>Purpose: </strong>The rarity of cerebellar glioblastoma presents a significant challenge in clinical practice due to the lack of extensive prognostic data on long-term survival rates, rendering it an underrepresented entity compared to its supratentorial counterpart. This study aims to analyze potential differences in survival outcome between patients with cerebellar and supratentorial glioblastomas.</p><p><strong>Methods: </strong>From 2009 to 2020, 8 patients underwent surgical treatment for cerebellar glioblastoma at the authors' institution. These patients were individually matched with a cohort of 205 consecutive patients from our institutional database with supratentorial glioblastoma, taking into account key prognostic parameters. Progression-free survival (PFS) and overall survival (OS) rates were compared. Additionally, we performed a systematic literature review to compile further survival data on cerebellar glioblastoma patients.</p><p><strong>Results: </strong>The median OS for cerebellar glioblastoma patients was 18 months (95% CI 11-25). The balanced matched-pair analysis showed no significant difference in survival when compared to patients with supratentorial glioblastoma, exhibiting a median OS of 23 months (95% CI 0-62) (p = 0.63). Respective values for PFS were 8 months (95% CI 4-12) for cerebellar and 7 months (95% CI 0-16) for supratentorial glioblastoma (p = 0.2). The systematic review revealed that median OS for cerebellar glioblastoma in current literature ranges from 7 to 21 months.</p><p><strong>Conclusions: </strong>The present findings indicate that patients with supra- and infratentorial glioblastoma do not significantly differ in regard to survival outcome parameters. This similarity in prognosis might encourage clinicians to consider surgical interventions for both supra- and infratentorial glioblastoma in a similar manner.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"432"},"PeriodicalIF":2.7,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438707/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142347231","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-28DOI: 10.1007/s00432-024-05955-4
Julia Maurer, Anna Saibold, Katharina Gerl, Michael Koller, Oliver Koelbl, Tobias Pukrop, Sandra Windschuettl, Sabine Einhell, Anne Herrmann-Johns, Georgios Raptis, Karolina Mueller
Purpose: The evaluation of treatment success and progression in oncology patient-reported outcomes (PROs) is playing an increasingly important role. Meanwhile, PROs are a component of the certification requirements of the German Cancer Society for oncology centers. PROs are used to provide supportive therapy. There is currently no instrument that fully covers the requirements. At the University Hospital Regensburg (UKR), a digital ONCOlogical-ROUTinE-Screening (ONCO-ROUTES) procedure was developed in order to assess the need for supportive therapy in a standardized way and to provide patients with supportive interventions tailored to their needs.
Methods: On the basis of current requirements and guidelines, the development of ONCO-ROUTES was supported by experts in focus groups and interviews, and digitalization was carried out in connection with the IT infrastructure.
Results: A Needs-based, Quality-of-life (QoL) and Symptoms Screening (NQS2) tool already established in the routine at the UKR was further developed into ONCO-ROUTES, which is made up of the domains therapy phase, nutrition, tobacco use, alcohol use, quality of life, general condition/functional status, physical activity, psychooncology, social services, and further support needs. By linking the digitized questionnaire to the hospital information system, the results are available for immediate use in routine operations and thus for the referral of patients for further supportive therapy.
Conclusion: The digital PRO application ONCO-ROUTES is designed to involve patients in monitoring additional supportive needs and thus, improves supportive interdisciplinary treatment.
目的:在肿瘤患者报告结果(PROs)中,对治疗成功率和进展情况的评估正发挥着越来越重要的作用。同时,PROs 也是德国癌症协会对肿瘤中心认证要求的一部分。患者报告结果可用于提供支持性治疗。目前还没有一种工具能完全满足要求。雷根斯堡大学医院(UKR)开发了一种数字ONCOlogical-ROUTinE-Screening(ONCO-ROUTES)程序,旨在以标准化的方式评估支持性治疗的需求,并为患者提供符合其需求的支持性干预措施:方法:在现有要求和指南的基础上,ONCO-ROUTES的开发得到了焦点小组和访谈专家的支持,并结合IT基础设施实现了数字化:结果:在英国皇家研究理事会的例行工作中已经建立的基于需求的生活质量(QoL)和症状筛查(NQS2)工具被进一步开发成 ONCO-ROUTES,它由治疗阶段、营养、吸烟、饮酒、生活质量、一般状况/功能状态、体育活动、肿瘤心理、社会服务和进一步的支持需求等领域组成。通过将数字化问卷与医院信息系统相连接,可在日常工作中立即使用问卷结果,从而转介病人接受进一步的支持性治疗:数字 PRO 应用程序 ONCO-ROUTES 的设计目的是让病人参与监测额外的支持需求,从而改善跨学科支持治疗。
{"title":"Systematic development of a patient-reported ONCOlogical-ROUTinE-Screening (ONCO-ROUTES) procedure at the University Cancer Center Regensburg.","authors":"Julia Maurer, Anna Saibold, Katharina Gerl, Michael Koller, Oliver Koelbl, Tobias Pukrop, Sandra Windschuettl, Sabine Einhell, Anne Herrmann-Johns, Georgios Raptis, Karolina Mueller","doi":"10.1007/s00432-024-05955-4","DOIUrl":"10.1007/s00432-024-05955-4","url":null,"abstract":"<p><strong>Purpose: </strong>The evaluation of treatment success and progression in oncology patient-reported outcomes (PROs) is playing an increasingly important role. Meanwhile, PROs are a component of the certification requirements of the German Cancer Society for oncology centers. PROs are used to provide supportive therapy. There is currently no instrument that fully covers the requirements. At the University Hospital Regensburg (UKR), a digital ONCOlogical-ROUTinE-Screening (ONCO-ROUTES) procedure was developed in order to assess the need for supportive therapy in a standardized way and to provide patients with supportive interventions tailored to their needs.</p><p><strong>Methods: </strong>On the basis of current requirements and guidelines, the development of ONCO-ROUTES was supported by experts in focus groups and interviews, and digitalization was carried out in connection with the IT infrastructure.</p><p><strong>Results: </strong>A Needs-based, Quality-of-life (QoL) and Symptoms Screening (NQS<sup>2</sup>) tool already established in the routine at the UKR was further developed into ONCO-ROUTES, which is made up of the domains therapy phase, nutrition, tobacco use, alcohol use, quality of life, general condition/functional status, physical activity, psychooncology, social services, and further support needs. By linking the digitized questionnaire to the hospital information system, the results are available for immediate use in routine operations and thus for the referral of patients for further supportive therapy.</p><p><strong>Conclusion: </strong>The digital PRO application ONCO-ROUTES is designed to involve patients in monitoring additional supportive needs and thus, improves supportive interdisciplinary treatment.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"435"},"PeriodicalIF":2.7,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438834/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142347236","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-28DOI: 10.1007/s00432-024-05962-5
Leszek Kraj, Paulina Chmiel, Andrzej Śliwczyński, Łukasz Szymański, Krzysztof Woźniak, Maciej Słodkowski, Tomasz Stokłosa, Lucjan Wyrwicz
Purpose: Pancreatic cancer remains a significant public health challenge, with poor long-term outcomes due to the lack of effective treatment options. Repurposing commonly used clinical drugs, such as ACE inhibitors, ARBs, CCBs, and metformin, may enhance the efficacy of chemotherapy and offer a promising therapeutic strategy for improving patient outcomes.
Methods: A retrospective analysis of concomitant treatment with ACE-Is, ARBs, CCBs, and metformin alongside gemcitabine chemotherapy in patients with pancreatic cancer was conducted. Treatment responses were evaluated, with overall survival (OS) estimated using the Kaplan-Meier method. Additionally, the Cox proportional hazards model was employed to assess the impact of these specific agents on patient survival.
Results: 4628 patients with various stages of pancreatic cancer were identified in the database between 2007 and 2016. The estimated overall survival (OS) in the analyzed group was 6.9 months (95% CI 6.4-7). The use of any of the analyzed drugs was associated with a significant improvement in mOS of 7.5 months (95% CI 6.8-7.8) vs. 6.7 months (95% CI 6.4-7.0) for patients who did not have additional treatment (p < 0.0001). ARBs, ACE-Is, CCBs, and metformin varied in their effectiveness in prolonging mOS among patients. The longest mOS of 8.9 months (95% CI 7.7-11.6) was observed in patients receiving additional therapy with ARBs, while the shortest mOS of 7.7 months (95% CI 6.5-8.9) was achieved by patients receiving metformin. In the adjusted Cox analysis, metformin was associated with a significantly weaker effect on mOS (p = 0.029). A particularly interesting trend in prolonging 5-year survival was demonstrated by ARBs and CCBs with 14.1% (95% CI 9-22%) and 14.8% (95% CI 11.1-19.6%), respectively, compared to patients not taking these drugs, who achieved a 5-year OS of 3.8% (95% CI 3.2-4.4%).
Conclusion: Our results demonstrate a significant positive impact of ARBs, ACE inhibitors, and CCBs on survival in patients with pancreatic cancer treated with gemcitabine. The addition of these inexpensive and relatively safe drugs in patients with additional comorbidities may represent a potential therapeutic option in this indication. However, prospective clinical trials to evaluate the optimal patient population and further studies to determine the potential impact of these agents on chemotherapy are necessary.
目的:胰腺癌仍然是一项重大的公共卫生挑战,由于缺乏有效的治疗方案,长期疗效不佳。临床常用药物(如 ACE 抑制剂、ARBs、CCBs 和二甲双胍)的再利用可能会增强化疗的疗效,并为改善患者预后提供一种有前景的治疗策略:方法: 对胰腺癌患者在接受吉西他滨化疗的同时接受 ACE-Is、ARBs、CCBs 和二甲双胍治疗的情况进行了回顾性分析。对治疗反应进行了评估,并采用卡普兰-梅耶法估算了总生存期(OS)。此外,还采用了 Cox 比例危险模型来评估这些特定药物对患者生存期的影响:2007年至2016年期间,数据库中确定了4628名不同分期的胰腺癌患者。分析组的估计总生存期(OS)为6.9个月(95% CI 6.4-7)。与未接受额外治疗的患者的 6.7 个月(95% CI 6.4-7.0)总生存期相比,使用任何一种分析药物都能显著改善患者的总生存期,达到 7.5 个月(95% CI 6.8-7.8)(P 结论:我们的研究结果表明,使用任何一种分析药物都能显著改善患者的总生存期,达到 7.5 个月(95% CI 6.8-7.8):我们的研究结果表明,ARBs、ACE 抑制剂和 CCBs 对接受吉西他滨治疗的胰腺癌患者的生存期有明显的积极影响。在有其他合并症的患者中添加这些廉价且相对安全的药物可能是该适应症的一种潜在治疗选择。然而,有必要进行前瞻性临床试验以评估最佳患者人群,并进一步研究确定这些药物对化疗的潜在影响。
{"title":"Synergistic effects of calcium channel blockers and renin-angiotensin inhibitors with gemcitabine-based chemotherapy on the survival of patients with pancreatic cancer.","authors":"Leszek Kraj, Paulina Chmiel, Andrzej Śliwczyński, Łukasz Szymański, Krzysztof Woźniak, Maciej Słodkowski, Tomasz Stokłosa, Lucjan Wyrwicz","doi":"10.1007/s00432-024-05962-5","DOIUrl":"10.1007/s00432-024-05962-5","url":null,"abstract":"<p><strong>Purpose: </strong>Pancreatic cancer remains a significant public health challenge, with poor long-term outcomes due to the lack of effective treatment options. Repurposing commonly used clinical drugs, such as ACE inhibitors, ARBs, CCBs, and metformin, may enhance the efficacy of chemotherapy and offer a promising therapeutic strategy for improving patient outcomes.</p><p><strong>Methods: </strong>A retrospective analysis of concomitant treatment with ACE-Is, ARBs, CCBs, and metformin alongside gemcitabine chemotherapy in patients with pancreatic cancer was conducted. Treatment responses were evaluated, with overall survival (OS) estimated using the Kaplan-Meier method. Additionally, the Cox proportional hazards model was employed to assess the impact of these specific agents on patient survival.</p><p><strong>Results: </strong>4628 patients with various stages of pancreatic cancer were identified in the database between 2007 and 2016. The estimated overall survival (OS) in the analyzed group was 6.9 months (95% CI 6.4-7). The use of any of the analyzed drugs was associated with a significant improvement in mOS of 7.5 months (95% CI 6.8-7.8) vs. 6.7 months (95% CI 6.4-7.0) for patients who did not have additional treatment (p < 0.0001). ARBs, ACE-Is, CCBs, and metformin varied in their effectiveness in prolonging mOS among patients. The longest mOS of 8.9 months (95% CI 7.7-11.6) was observed in patients receiving additional therapy with ARBs, while the shortest mOS of 7.7 months (95% CI 6.5-8.9) was achieved by patients receiving metformin. In the adjusted Cox analysis, metformin was associated with a significantly weaker effect on mOS (p = 0.029). A particularly interesting trend in prolonging 5-year survival was demonstrated by ARBs and CCBs with 14.1% (95% CI 9-22%) and 14.8% (95% CI 11.1-19.6%), respectively, compared to patients not taking these drugs, who achieved a 5-year OS of 3.8% (95% CI 3.2-4.4%).</p><p><strong>Conclusion: </strong>Our results demonstrate a significant positive impact of ARBs, ACE inhibitors, and CCBs on survival in patients with pancreatic cancer treated with gemcitabine. The addition of these inexpensive and relatively safe drugs in patients with additional comorbidities may represent a potential therapeutic option in this indication. However, prospective clinical trials to evaluate the optimal patient population and further studies to determine the potential impact of these agents on chemotherapy are necessary.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"434"},"PeriodicalIF":2.7,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438632/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142347235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: To explore the value of 99mTc-isonitrile deoxyglucosamine (CNDG) SPECT/CT in the staging and resectability diagnosis of non-small cell lung cancer (NSCLC) compared with contrast-enhanced CT (CECT).
Methods: This research was approved by the hospital ethics review committee. Sixty-three patients with NSCLC received 99mTc-CNDG SPECT/CT, CECT and initial TNM staging before treatment. Thirty-three patients who underwent radical surgery underwent postoperative pathological TNM staging as the reference standard. Another thirty patients underwent radiochemotherapy; among them, the reference standard of 7 patients of N staging and 5 patients of M staging was based on biopsy pathology, and the diagnosis of the remaining lesions was confirmed by at least one different image or clinical imaging follow-up for more than 3 months. The McNemar test and receiver operating characteristic (ROC) curve analysis were used to compare the diagnostic accuracy of staging and resectability of 99mTc-CNDG SPECT/CT and CECT in NSCLC, respectively.
Results: For all patients and surgical patients, the accuracies of 99mTc-CNDG SPECT/CT in diagnosing the T stage and N stage were higher than those of CECT (all patients: 90.5%, 88.9% vs. 79.4%, 60.3%; surgical patients: 81.8%, 78.8% vs. 60.6%, 51.5%), and the differences were statistically significant (all patients: T stage, P = 0.016; N stage, P = 0.000; surgical patients: T stage, P = 0.016; N stage, P = 0.004). For all patients, the accuracy of 99mTc-CNDG SPECT/CT in diagnosing the M stage was higher than that of CECT (96.8% vs. 90.5%), but the difference was not statistically significant (P = 0.289). ROC curve analysis showed that the accuracy of 99mTc-CNDG SPECT/CT in diagnosing the potential resectability of NSCLC was significantly better than that of CECT (P = 0.046).
Conclusion: This preliminary clinical study shows that 99mTc-CNDG SPECT/CT is of great value for accurate clinical staging of NSCLC compared with CECT and can significantly improve the accuracy of resectability diagnosis.
{"title":"A prospective study to compare the diagnostic accuracy of <sup>99m</sup>Tc-CNDG SPECT/CT and contrast-enhanced CT in staging of non-small cell lung cancer.","authors":"Qinfen Wang, Zhensheng Deng, Chuangang Lu, Lijun Chen, Jiangjun Qin, Ping Wang","doi":"10.1007/s00432-024-05953-6","DOIUrl":"10.1007/s00432-024-05953-6","url":null,"abstract":"<p><strong>Objective: </strong>To explore the value of <sup>99m</sup>Tc-isonitrile deoxyglucosamine (CNDG) SPECT/CT in the staging and resectability diagnosis of non-small cell lung cancer (NSCLC) compared with contrast-enhanced CT (CECT).</p><p><strong>Methods: </strong>This research was approved by the hospital ethics review committee. Sixty-three patients with NSCLC received <sup>99m</sup>Tc-CNDG SPECT/CT, CECT and initial TNM staging before treatment. Thirty-three patients who underwent radical surgery underwent postoperative pathological TNM staging as the reference standard. Another thirty patients underwent radiochemotherapy; among them, the reference standard of 7 patients of N staging and 5 patients of M staging was based on biopsy pathology, and the diagnosis of the remaining lesions was confirmed by at least one different image or clinical imaging follow-up for more than 3 months. The McNemar test and receiver operating characteristic (ROC) curve analysis were used to compare the diagnostic accuracy of staging and resectability of <sup>99m</sup>Tc-CNDG SPECT/CT and CECT in NSCLC, respectively.</p><p><strong>Results: </strong>For all patients and surgical patients, the accuracies of <sup>99m</sup>Tc-CNDG SPECT/CT in diagnosing the T stage and N stage were higher than those of CECT (all patients: 90.5%, 88.9% vs. 79.4%, 60.3%; surgical patients: 81.8%, 78.8% vs. 60.6%, 51.5%), and the differences were statistically significant (all patients: T stage, P = 0.016; N stage, P = 0.000; surgical patients: T stage, P = 0.016; N stage, P = 0.004). For all patients, the accuracy of <sup>99m</sup>Tc-CNDG SPECT/CT in diagnosing the M stage was higher than that of CECT (96.8% vs. 90.5%), but the difference was not statistically significant (P = 0.289). ROC curve analysis showed that the accuracy of <sup>99m</sup>Tc-CNDG SPECT/CT in diagnosing the potential resectability of NSCLC was significantly better than that of CECT (P = 0.046).</p><p><strong>Conclusion: </strong>This preliminary clinical study shows that <sup>99m</sup>Tc-CNDG SPECT/CT is of great value for accurate clinical staging of NSCLC compared with CECT and can significantly improve the accuracy of resectability diagnosis.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"430"},"PeriodicalIF":2.7,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11427526/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142347230","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-23DOI: 10.1007/s00432-024-05878-0
Yulang Jiang, Dengcheng Hui, Ziyang Pan, Yongxin Yu, Lu Liu, Xiaofan Yu, Chao Wu, Mingyu Sun
Background: Ferroptosis, a novel iron-ion-dependent metabolic cell death mode with lipid peroxides as the main driving substrate, plays an irreplaceable role in the development and preventive treatment of hepatocellular carcinoma. Curcumin has potent pharmacological anti-tumor effects.
Aim of the study: We aimed to evaluate the ex vivo and in vivo cancer inhibitory activity of curcumin and its specific mechanism of action.
Materials and methods: We used the hepatocellular carcinoma cell lines HepG2 and SMMC7721 to assess the direct inhibition of hepatocellular carcinoma proliferation by curcumin in vitro and a tumor xenograft model to evaluate the in vivo cancer inhibitory effect of curcumin.
Results: In this study, we found that ferroptosis's inhibitors specifically reversed the curcumin-induced cell death pattern in HCC. After curcumin intervention, there was a substantial increase in MDA levels and iron ion levels, and a decrease in intracellular GSH levels. Meanwhile, the expression of GPX4 and SLC7A11 was significantly reduced at the protein levels, while ACSL4 and PTGS2 expression was significantly increased.
Conclusions: This study showed that curcumin significantly decreased the proliferation of HCC cells and significantly increased the sensitivity of ferroptosis. These results suggest that ACSL4 is a viable target for curcumin-induced ferroptosis in treating HCC.
{"title":"Curcumin promotes ferroptosis in hepatocellular carcinoma via upregulation of ACSL4.","authors":"Yulang Jiang, Dengcheng Hui, Ziyang Pan, Yongxin Yu, Lu Liu, Xiaofan Yu, Chao Wu, Mingyu Sun","doi":"10.1007/s00432-024-05878-0","DOIUrl":"10.1007/s00432-024-05878-0","url":null,"abstract":"<p><strong>Background: </strong>Ferroptosis, a novel iron-ion-dependent metabolic cell death mode with lipid peroxides as the main driving substrate, plays an irreplaceable role in the development and preventive treatment of hepatocellular carcinoma. Curcumin has potent pharmacological anti-tumor effects.</p><p><strong>Aim of the study: </strong>We aimed to evaluate the ex vivo and in vivo cancer inhibitory activity of curcumin and its specific mechanism of action.</p><p><strong>Materials and methods: </strong>We used the hepatocellular carcinoma cell lines HepG2 and SMMC7721 to assess the direct inhibition of hepatocellular carcinoma proliferation by curcumin in vitro and a tumor xenograft model to evaluate the in vivo cancer inhibitory effect of curcumin.</p><p><strong>Results: </strong>In this study, we found that ferroptosis's inhibitors specifically reversed the curcumin-induced cell death pattern in HCC. After curcumin intervention, there was a substantial increase in MDA levels and iron ion levels, and a decrease in intracellular GSH levels. Meanwhile, the expression of GPX4 and SLC7A11 was significantly reduced at the protein levels, while ACSL4 and PTGS2 expression was significantly increased.</p><p><strong>Conclusions: </strong>This study showed that curcumin significantly decreased the proliferation of HCC cells and significantly increased the sensitivity of ferroptosis. These results suggest that ACSL4 is a viable target for curcumin-induced ferroptosis in treating HCC.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"429"},"PeriodicalIF":2.7,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11420324/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142287955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-22DOI: 10.1007/s00432-024-05944-7
Florian Bürtin, Liema Elias, Sebastian Hinz, Michael Forster, Guido Hildebrandt, Bernd Frerker, Felix Bock
Background: Liquid biopsy is a minimally invasive procedure investigating tumor mutations.
Methods: In our retrospective study, we investigated whether molecular therapy monitoring of patients receiving neoadjuvant radio(chemo)therapy on a daily routine is possible in 17 patients with locally advanced rectal cancer. Six patients received short-course radiotherapy (5 × 5 Gy) with subsequent surgery, six patients were treated according RAPIDO protocol with short-course radiotherapy followed by chemotherapy (FOLFOX4) and subsequent surgery and five patients received conventional neoadjuvant radiochemotherapy with 5-FU followed by surgery. Response was assessed by Dworak. Liquid biopsies were taken before and immediately after neoadjuvant radiotherapy to isolate and ultradeeply sequence cell free DNA with a panel of 127 genes. Somatic mutations were determined bioinformatically by comparison with normal DNA from leukocytes to distinguish them from germline variants or aging mutations.
Results: In 12 patients (71%) at least one somatic mutation was detected. In 8/12 patients a decrease and in 4/12 an increase or mixed response in ctDNA was seen. Statistical correlation between ctDNA analysis and clinical response could not be seen.
Conclusion: ctDNA is responding to neoadjuvant therapy and liquid biopsy is easily integrated into a daily routine. As part of translational research this protocol leaves room for further investigations.
{"title":"ctDNA responds to neoadjuvant treatment in locally advanced rectal cancer.","authors":"Florian Bürtin, Liema Elias, Sebastian Hinz, Michael Forster, Guido Hildebrandt, Bernd Frerker, Felix Bock","doi":"10.1007/s00432-024-05944-7","DOIUrl":"10.1007/s00432-024-05944-7","url":null,"abstract":"<p><strong>Background: </strong>Liquid biopsy is a minimally invasive procedure investigating tumor mutations.</p><p><strong>Methods: </strong>In our retrospective study, we investigated whether molecular therapy monitoring of patients receiving neoadjuvant radio(chemo)therapy on a daily routine is possible in 17 patients with locally advanced rectal cancer. Six patients received short-course radiotherapy (5 × 5 Gy) with subsequent surgery, six patients were treated according RAPIDO protocol with short-course radiotherapy followed by chemotherapy (FOLFOX4) and subsequent surgery and five patients received conventional neoadjuvant radiochemotherapy with 5-FU followed by surgery. Response was assessed by Dworak. Liquid biopsies were taken before and immediately after neoadjuvant radiotherapy to isolate and ultradeeply sequence cell free DNA with a panel of 127 genes. Somatic mutations were determined bioinformatically by comparison with normal DNA from leukocytes to distinguish them from germline variants or aging mutations.</p><p><strong>Results: </strong>In 12 patients (71%) at least one somatic mutation was detected. In 8/12 patients a decrease and in 4/12 an increase or mixed response in ctDNA was seen. Statistical correlation between ctDNA analysis and clinical response could not be seen.</p><p><strong>Conclusion: </strong>ctDNA is responding to neoadjuvant therapy and liquid biopsy is easily integrated into a daily routine. As part of translational research this protocol leaves room for further investigations.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"428"},"PeriodicalIF":2.7,"publicationDate":"2024-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11417078/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142287954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-20DOI: 10.1007/s00432-024-05939-4
JingCheng Zhang, Sheng Zhou, SiSi Jiang, Fang He, Yan Tu, HuiXian Hu
Background: Double-hit lymphoma (DHL) with c-MYC gene translocation is highly aggressive and has a poor prognosis. In DHL cells, activation-induced cytidine deaminase (AID) promotes antibody class switch recombination (CSR), ultimately leading to c-MYC gene translocation caused by Myc/IgH DNA double-strand breaks. However, currently there is still no method to suppress the expression of AID.
Methods: In this study, we compared the clinical significance of AID expression in DHL, Additionally, two human double-hit lymphoma cell lines were used to analyze the effect of imatinib mesylate on c-MYC in vitro, and the therapeutic effect was also evaluated in xenograft mouse models.
Results: Imatinib mesylate downregulated the AID and c-MYC proteins in patients with chronic myelogenous leukemia associated with DHL. In addition, imatinib mesylate reduced AID and c-MYC expression in SU-DHL-4 and OCI-Ly18 DHL cells. Imatinib mesylate exerted significant inhibitory effects on the proliferation and metastasis of SU-DHL-4 and OCI-Ly18 cells. Finally, imatinib mesylate reduced not only tumor burden in DHL mouse models, but also AID and c-MYC expression in vivo.
Conclusion: These findings reveal that imatinib mesylate effectively reduces the carcinogenic function of c-MYC in DHL, providing novel strategies for developing therapies targeting c-MYC-driven DHL.
背景:伴有c-MYC基因易位的双击淋巴瘤(DHL)具有高度侵袭性,预后较差。在DHL细胞中,活化诱导的胞苷脱氨酶(AID)促进抗体类开关重组(CSR),最终导致Myc/IgH DNA双链断裂引起的c-MYC基因易位。然而,目前仍没有抑制 AID 表达的方法:本研究比较了AID在DHL中表达的临床意义,并使用两种人类双打淋巴瘤细胞系分析了甲磺酸伊马替尼对体外c-MYC的影响,还在异种移植小鼠模型中评估了其治疗效果:结果:甲磺酸伊马替尼能下调与DHL相关的慢性髓性白血病患者的AID和c-MYC蛋白。此外,甲磺酸伊马替尼还能降低AID和c-MYC在SU-DHL-4和OCI-Ly18 DHL细胞中的表达。甲磺酸伊马替尼对SU-DHL-4和OCI-Ly18细胞的增殖和转移具有显著的抑制作用。最后,甲磺酸伊马替尼不仅降低了DHL小鼠模型的肿瘤负荷,还降低了体内AID和c-MYC的表达:这些研究结果表明,甲磺酸伊马替尼能有效降低c-MYC在DHL中的致癌功能,为开发针对c-MYC驱动的DHL的疗法提供了新策略。
{"title":"Imatinib mesylate reduces c-MYC expression in double-hit lymphoma cells by suppressing inducible cytidine deaminase.","authors":"JingCheng Zhang, Sheng Zhou, SiSi Jiang, Fang He, Yan Tu, HuiXian Hu","doi":"10.1007/s00432-024-05939-4","DOIUrl":"10.1007/s00432-024-05939-4","url":null,"abstract":"<p><strong>Background: </strong>Double-hit lymphoma (DHL) with c-MYC gene translocation is highly aggressive and has a poor prognosis. In DHL cells, activation-induced cytidine deaminase (AID) promotes antibody class switch recombination (CSR), ultimately leading to c-MYC gene translocation caused by Myc/IgH DNA double-strand breaks. However, currently there is still no method to suppress the expression of AID.</p><p><strong>Methods: </strong>In this study, we compared the clinical significance of AID expression in DHL, Additionally, two human double-hit lymphoma cell lines were used to analyze the effect of imatinib mesylate on c-MYC in vitro, and the therapeutic effect was also evaluated in xenograft mouse models.</p><p><strong>Results: </strong>Imatinib mesylate downregulated the AID and c-MYC proteins in patients with chronic myelogenous leukemia associated with DHL. In addition, imatinib mesylate reduced AID and c-MYC expression in SU-DHL-4 and OCI-Ly18 DHL cells. Imatinib mesylate exerted significant inhibitory effects on the proliferation and metastasis of SU-DHL-4 and OCI-Ly18 cells. Finally, imatinib mesylate reduced not only tumor burden in DHL mouse models, but also AID and c-MYC expression in vivo.</p><p><strong>Conclusion: </strong>These findings reveal that imatinib mesylate effectively reduces the carcinogenic function of c-MYC in DHL, providing novel strategies for developing therapies targeting c-MYC-driven DHL.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"426"},"PeriodicalIF":2.7,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11413099/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142287956","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-20DOI: 10.1007/s00432-024-05949-2
Chenyu Shen, Wenxi Jiang, Ruiqing Chen, Lingbing Li, Yunbo Wu, Long Tan, Yadong Chen, Weiqiang Zhang, Zhijun Wang
Background: The treatment of unresectable hepatocellular carcinoma (uHCC) challenging due to unfulfilled clinical requirements.
Objective: To evaluate the safety and efficacy of combining transarterial chemoembolization (TACE) with sintilimab and lenvatinib in the treatment of uHCC.
Methods: We retrospectively analyzed the data of patients with uHCC who were treated with a combination of TACE, sintilimab, and lenvatinib between May 2019 and December 2021 at the Chinese PLA General Hospital. Systemic treatment was started 1 week after TACE was performed. Sintilimab was administered intravenously at a dosage of 200 mg every three weeks, and lenvatinib was given orally at dosages of 8 mg or 12 mg daily, contingent upon the weight of the patients. The primary endpoint was the objective response rate (ORR) as per the mRECIST. Secondary endpoints were disease control rate (DCR), progression-free survival (PFS), overall survival (OS) and treatment-related adverse events (tr-AEs).
Results: A total of 32 patients were enrolled in the study. Among them, 9 patients were classified as Barcelona Clinic Liver Cancer-B (BCLC-B), 23 patients were classified as BCLC-C, 14 patients diagnosed with portal vein tumors, and 12 patients were diagnosed with extra hepatic metastases. The ORR and DCR were 75% and 90.6% respectively, with 4 patients exhibiting (12.5%) complete response, 20 patients exhibiting (62.5%) partial response, 5 patients exhibiting (15.6%) stable disease, and 3 patients exhibiting (9.4%) progressive disease. With a median follow-up time of 19.6 months, the median PFS was 9.9 months, and the median OS was 33.3 months. A total of 31 patients experienced different degrees of tr-AEs, of which 2 were grade 3 tr-AEs.
Conclusion: The combination therapy of TACE, sintilimab, and lenvatinib demonstrates satisfactory efficacy in the treatment of uHCC with manageable tr-AEs.
{"title":"Transarterial chemoembolization combined with sintilimab and lenvatinib for the treatment of unresectable hepatocellular carcinoma: a retrospective study.","authors":"Chenyu Shen, Wenxi Jiang, Ruiqing Chen, Lingbing Li, Yunbo Wu, Long Tan, Yadong Chen, Weiqiang Zhang, Zhijun Wang","doi":"10.1007/s00432-024-05949-2","DOIUrl":"10.1007/s00432-024-05949-2","url":null,"abstract":"<p><strong>Background: </strong>The treatment of unresectable hepatocellular carcinoma (uHCC) challenging due to unfulfilled clinical requirements.</p><p><strong>Objective: </strong>To evaluate the safety and efficacy of combining transarterial chemoembolization (TACE) with sintilimab and lenvatinib in the treatment of uHCC.</p><p><strong>Methods: </strong>We retrospectively analyzed the data of patients with uHCC who were treated with a combination of TACE, sintilimab, and lenvatinib between May 2019 and December 2021 at the Chinese PLA General Hospital. Systemic treatment was started 1 week after TACE was performed. Sintilimab was administered intravenously at a dosage of 200 mg every three weeks, and lenvatinib was given orally at dosages of 8 mg or 12 mg daily, contingent upon the weight of the patients. The primary endpoint was the objective response rate (ORR) as per the mRECIST. Secondary endpoints were disease control rate (DCR), progression-free survival (PFS), overall survival (OS) and treatment-related adverse events (tr-AEs).</p><p><strong>Results: </strong>A total of 32 patients were enrolled in the study. Among them, 9 patients were classified as Barcelona Clinic Liver Cancer-B (BCLC-B), 23 patients were classified as BCLC-C, 14 patients diagnosed with portal vein tumors, and 12 patients were diagnosed with extra hepatic metastases. The ORR and DCR were 75% and 90.6% respectively, with 4 patients exhibiting (12.5%) complete response, 20 patients exhibiting (62.5%) partial response, 5 patients exhibiting (15.6%) stable disease, and 3 patients exhibiting (9.4%) progressive disease. With a median follow-up time of 19.6 months, the median PFS was 9.9 months, and the median OS was 33.3 months. A total of 31 patients experienced different degrees of tr-AEs, of which 2 were grade 3 tr-AEs.</p><p><strong>Conclusion: </strong>The combination therapy of TACE, sintilimab, and lenvatinib demonstrates satisfactory efficacy in the treatment of uHCC with manageable tr-AEs.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"150 9","pages":"427"},"PeriodicalIF":2.7,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11415473/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142287964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
This study aimed to investigate the effect of MYO3B on endometrial cancer (EC) proliferation and invasion.
Methods
The expression of MYO3B in EC tissues and cells was analyzed using TCGA database, immunohistochemical staining, real-time PCR, and western blot (WB). Cell proliferation was detected by CCK8, Annexin V-APC/PI flow cytometry was used to detect apoptosis, intracellular calcium ion (Ca2+) was detected by flow cytometry with Fluo-4 AM fluorescent probe, cell migration by scratch assay, and cell invasion by Transwell assay, and the expression of proteins related to Ca2+ homeostasis and RhoA/ROCK1 signaling pathway was detected by WB and immunofluorescence staining.
Results
The expression of MYO3B was an influential factor in EC recurrence, and the expression of MYO3B was significantly up-regulated in EC tissues and cells, but down-regulated in KLE cells, and MYO3B knockdown inhibited the proliferation, migration, and invasion ability of EC cells and promoted apoptosis, suggesting that MYO3B plays a tumor-promoting role in EC. Furthermore, MYO3B knockdown decreased Ca2+ concentration in EC cells and the RhoA/ROCK1 signaling pathway was inhibited, and the effect of MYO3B knockdown on RhoA/ROCK1 signaling was reversed by treatment with the Calmodulin agonist CALP-2, and the effects of MYO3B knockdown on cell proliferation, migration, and invasion were reversed after treatment with the RhoA agonist U-46,619.
Conclusion
MYO3B promotes the proliferation and migration of endometrial cancer cells via Ca2+-RhoA/ROCK1 signaling pathway. High expression of MYO3B may be a biomarker for EC metastasis.
{"title":"MYO3B promotes cancer progression in endometrial cancer by mediating the calcium ion-RhoA/ROCK1 signaling pathway","authors":"Chunmei Zhang, Huifeng Zhang, Xiaofeng Yang, Sufen Li, Liang Wang, Huancheng Su, Jiaolin Yang, Yuanyuan Ding, Xinglin Zhang, Bao Qiang, Sanyuan Zhang","doi":"10.1007/s00432-024-05940-x","DOIUrl":"https://doi.org/10.1007/s00432-024-05940-x","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>This study aimed to investigate the effect of MYO3B on endometrial cancer (EC) proliferation and invasion.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>The expression of MYO3B in EC tissues and cells was analyzed using TCGA database, immunohistochemical staining, real-time PCR, and western blot (WB). Cell proliferation was detected by CCK8, Annexin V-APC/PI flow cytometry was used to detect apoptosis, intracellular calcium ion (Ca<sup>2+</sup>) was detected by flow cytometry with Fluo-4 AM fluorescent probe, cell migration by scratch assay, and cell invasion by Transwell assay, and the expression of proteins related to Ca<sup>2+</sup> homeostasis and RhoA/ROCK1 signaling pathway was detected by WB and immunofluorescence staining.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>The expression of MYO3B was an influential factor in EC recurrence, and the expression of MYO3B was significantly up-regulated in EC tissues and cells, but down-regulated in KLE cells, and MYO3B knockdown inhibited the proliferation, migration, and invasion ability of EC cells and promoted apoptosis, suggesting that MYO3B plays a tumor-promoting role in EC. Furthermore, MYO3B knockdown decreased Ca<sup>2+</sup> concentration in EC cells and the RhoA/ROCK1 signaling pathway was inhibited, and the effect of MYO3B knockdown on RhoA/ROCK1 signaling was reversed by treatment with the Calmodulin agonist CALP-2, and the effects of MYO3B knockdown on cell proliferation, migration, and invasion were reversed after treatment with the RhoA agonist U-46,619.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>MYO3B promotes the proliferation and migration of endometrial cancer cells via Ca<sup>2+</sup>-RhoA/ROCK1 signaling pathway. High expression of MYO3B may be a biomarker for EC metastasis.</p>","PeriodicalId":15118,"journal":{"name":"Journal of Cancer Research and Clinical Oncology","volume":"85 1","pages":""},"PeriodicalIF":3.6,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142260980","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}