Background: Primary testicular lymphoma (PTL) is a rare, aggressive malignancy, representing a small fraction of testicular tumors and non-Hodgkin lymphomas, yet it is the most common testicular malignancy in older men. Diffuse large B-cell lymphoma (DLBCL), which is typically the aggressive subtype, dominates PTL and shows diffuse B-cell infiltration. Venous tumor thrombus, uncommon in lymphomas, is uniquely reported in this case of testicular DLBCL with gonadal vein involvement.
Case summary: A 62-year-old man presented with a two-month history of painless left testicular swelling and stiffness. Diagnostic imaging [ultrasonography, computed tomography (CT), and 18F-fluorodeoxyglucose positron emission tomography/CT (18F-FDG-PET/CT)] revealed bilateral testicular masses and a gonadal vein tumor thrombus (SUVmax 16.5). Left orchiectomy confirmed DLBCL with CD20, Bcl-2, and MUM1 positivity (Ki-67: approximately 80%). The disease was staged as Ann Arbor stage IVA (International Prognostic Index score 3, high-intermediate risk). The patient received Rituximab, Polatuzumab Vedotin, Cyclophosphamide, Epirubicin, and Prednisolone chemotherapy, completing the first cycle with good tolerability. No adverse events were reported, and follow-up is ongoing to assess long-term outcomes. This case highlights the diagnostic utility of 18F-FDG-PET/CT and the importance of multidisciplinary management in rare PTL presentations with tumor thrombus.
Conclusion: This case demonstrates the diagnostic complexities of PTL with gonadal vein tumor thrombus, underscoring the importance of considering lymphoma in elderly patients with testicular masses and venous involvement. A multi-disciplinary team including urologists, hematologists, and radiation oncologists is needed to ensure appropriate therapy.
{"title":"Primary testicular diffuse large B-cell lymphoma with gonadal vein tumor thrombus: A case report and review of the literature.","authors":"Yu-Zhi Zuo, Zhen Liang, Bo-Ju Pan, Wei-Gang Yan, Zhi-En Zhou","doi":"10.5306/wjco.v16.i11.111527","DOIUrl":"10.5306/wjco.v16.i11.111527","url":null,"abstract":"<p><strong>Background: </strong>Primary testicular lymphoma (PTL) is a rare, aggressive malignancy, representing a small fraction of testicular tumors and non-Hodgkin lymphomas, yet it is the most common testicular malignancy in older men. Diffuse large B-cell lymphoma (DLBCL), which is typically the aggressive subtype, dominates PTL and shows diffuse B-cell infiltration. Venous tumor thrombus, uncommon in lymphomas, is uniquely reported in this case of testicular DLBCL with gonadal vein involvement.</p><p><strong>Case summary: </strong>A 62-year-old man presented with a two-month history of painless left testicular swelling and stiffness. Diagnostic imaging [ultrasonography, computed tomography (CT), and <sup>18</sup>F-fluorodeoxyglucose positron emission tomography/CT (<sup>18</sup>F-FDG-PET/CT)] revealed bilateral testicular masses and a gonadal vein tumor thrombus (SUVmax 16.5). Left orchiectomy confirmed DLBCL with CD20, Bcl-2, and MUM1 positivity (Ki-67: approximately 80%). The disease was staged as Ann Arbor stage IVA (International Prognostic Index score 3, high-intermediate risk). The patient received Rituximab, Polatuzumab Vedotin, Cyclophosphamide, Epirubicin, and Prednisolone chemotherapy, completing the first cycle with good tolerability. No adverse events were reported, and follow-up is ongoing to assess long-term outcomes. This case highlights the diagnostic utility of <sup>18</sup>F-FDG-PET/CT and the importance of multidisciplinary management in rare PTL presentations with tumor thrombus.</p><p><strong>Conclusion: </strong>This case demonstrates the diagnostic complexities of PTL with gonadal vein tumor thrombus, underscoring the importance of considering lymphoma in elderly patients with testicular masses and venous involvement. A multi-disciplinary team including urologists, hematologists, and radiation oncologists is needed to ensure appropriate therapy.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"111527"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678962/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701656","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-24DOI: 10.5306/wjco.v16.i11.111397
Fatmah H Alsharif
Background: Cancer survivorship is a growing concern globally, yet few studies have explored the quality of life (QoL) outcomes among survivors in the Middle East, particularly in Saudi Arabia.
Aim: To assess QoL using the World Health Organization Quality of Life-BREF (WHOQOL-BREF) and to evaluate the impact of demographic and clinical factors among Saudi cancer survivors.
Methods: We conducted a cross-sectional study of 102 adult cancer survivors recruited from a tertiary hospital in Jeddah, Saudi Arabia. Participants completed the WHOQOL-BREF, which assesses four QoL domains, including physical health, psychological health, social relationships, and environment. Univariate and multivariable robust linear regression models (Huber estimator) were used to identify QoL score predictors, adjusted for key sociodemographic and clinical variables.
Results: The mean participant age was 44.5 years; 72.5% of the participants were female. The mean domain scores were as follows: physical health was 3.05 ± 0.53, psychological health was 3.56 ± 0.79, social relationships was 3.39 ± 0.84, and environment was 3.29 ± 0.74. Socioeconomic and social vulnerability factors, including low income, rental housing, widowed/divorced marital status, and lower education, were independently associated with poorer QoL scores. Residents in rural settings had significantly lower scores in all domains.
Conclusion: Our findings reveal noticeable disparities in QoL among Saudi cancer survivors driven by socioeconomic and demographic factors. These insights underscore the need for context-sensitive survivorship programs in Saudi Arabia, with special attention to social support, mental health, and economic stability.
{"title":"Quality of life among Saudi cancer survivors: The role of social and demographic factors.","authors":"Fatmah H Alsharif","doi":"10.5306/wjco.v16.i11.111397","DOIUrl":"10.5306/wjco.v16.i11.111397","url":null,"abstract":"<p><strong>Background: </strong>Cancer survivorship is a growing concern globally, yet few studies have explored the quality of life (QoL) outcomes among survivors in the Middle East, particularly in Saudi Arabia.</p><p><strong>Aim: </strong>To assess QoL using the World Health Organization Quality of Life-BREF (WHOQOL-BREF) and to evaluate the impact of demographic and clinical factors among Saudi cancer survivors.</p><p><strong>Methods: </strong>We conducted a cross-sectional study of 102 adult cancer survivors recruited from a tertiary hospital in Jeddah, Saudi Arabia. Participants completed the WHOQOL-BREF, which assesses four QoL domains, including physical health, psychological health, social relationships, and environment. Univariate and multivariable robust linear regression models (Huber estimator) were used to identify QoL score predictors, adjusted for key sociodemographic and clinical variables.</p><p><strong>Results: </strong>The mean participant age was 44.5 years; 72.5% of the participants were female. The mean domain scores were as follows: physical health was 3.05 ± 0.53, psychological health was 3.56 ± 0.79, social relationships was 3.39 ± 0.84, and environment was 3.29 ± 0.74. Socioeconomic and social vulnerability factors, including low income, rental housing, widowed/divorced marital status, and lower education, were independently associated with poorer QoL scores. Residents in rural settings had significantly lower scores in all domains.</p><p><strong>Conclusion: </strong>Our findings reveal noticeable disparities in QoL among Saudi cancer survivors driven by socioeconomic and demographic factors. These insights underscore the need for context-sensitive survivorship programs in Saudi Arabia, with special attention to social support, mental health, and economic stability.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"111397"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-24DOI: 10.5306/wjco.v16.i11.111419
Jelena Roganovic, Mia Radosevic, Ana Dordevic
The gut microbiome plays a pivotal role in immune homeostasis and systemic inflammatory regulation, both of which are critically involved in the pathogenesis and progression of pediatric leukemias. Recent evidence reveals that children with leukemia often exhibit distinct gut microbiome profiles at diagnosis, marked by reduced microbial diversity and the enrichment of pro-inflammatory taxa such as Enterococcus and Streptococcus. This microbial dysbiosis may promote leukemogenesis by disrupting immune regulation and driving chronic inflammation. Chemotherapy significantly alters the gut microbiome, inducing dysbiosis characterized by a loss of beneficial commensals and the dominance of pathobionts. Specific microbial signatures, such as the enrichment of Bacteroides, correlate with reduced inflammation and improved prognosis, underscoring the gut microbiome's prognostic value. Emerging therapies, including dietary adjustments, probiotics, and fecal gut microbiome transplantation, aim to restore microbial balance and reduce treatment-related complications. Moreover, gut microbiome profiling shows potential for identifying biomarkers linked to leukemia predisposition, paving the way for early diagnosis and tailored preventive strategies. This mini-review explores recent advancements in understanding the influence of the gut microbiome on pediatric leukemias, emphasizing its role as both a therapeutic target and a prognostic biomarker. Integrating gut microbiome research into clinical practice may help optimize treatment outcomes and improve quality of life for children with leukemia.
{"title":"Role of the gut microbiome in the development and prognosis of pediatric leukemia.","authors":"Jelena Roganovic, Mia Radosevic, Ana Dordevic","doi":"10.5306/wjco.v16.i11.111419","DOIUrl":"10.5306/wjco.v16.i11.111419","url":null,"abstract":"<p><p>The gut microbiome plays a pivotal role in immune homeostasis and systemic inflammatory regulation, both of which are critically involved in the pathogenesis and progression of pediatric leukemias. Recent evidence reveals that children with leukemia often exhibit distinct gut microbiome profiles at diagnosis, marked by reduced microbial diversity and the enrichment of pro-inflammatory taxa such as <i>Enterococcus</i> and <i>Streptococcus</i>. This microbial dysbiosis may promote leukemogenesis by disrupting immune regulation and driving chronic inflammation. Chemotherapy significantly alters the gut microbiome, inducing dysbiosis characterized by a loss of beneficial commensals and the dominance of pathobionts. Specific microbial signatures, such as the enrichment of <i>Bacteroides</i>, correlate with reduced inflammation and improved prognosis, underscoring the gut microbiome's prognostic value. Emerging therapies, including dietary adjustments, probiotics, and fecal gut microbiome transplantation, aim to restore microbial balance and reduce treatment-related complications. Moreover, gut microbiome profiling shows potential for identifying biomarkers linked to leukemia predisposition, paving the way for early diagnosis and tailored preventive strategies. This mini-review explores recent advancements in understanding the influence of the gut microbiome on pediatric leukemias, emphasizing its role as both a therapeutic target and a prognostic biomarker. Integrating gut microbiome research into clinical practice may help optimize treatment outcomes and improve quality of life for children with leukemia.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"111419"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678948/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-24DOI: 10.5306/wjco.v16.i11.110257
Noura A A Ebrahim, Tamer S Eissa, Mustafa A Hussein, Omnia Mohamed Korany, Nancy H Amin
Background: Claudin-6 (CLDN6), a tight junction protein typically restricted to embryonic tissues, is re-expressed in various cancers. However, its prognostic significance in high-grade endometrial carcinoma (HGEC) remains unclear.
Aim: To investigate the expression pattern of CLDN6 in HGEC and assess its correlation with clinicopathological parameters and patient survival.
Methods: Immunohistochemical analysis of CLDN6 expression was performed on formalin-fixed, paraffin-embedded tissues from 80 patients diagnosed with HGEC. Associations between CLDN6 expression and histological subtype, the International Federation of Gynecology and Obstetrics (FIGO) stage, depth of myometrial invasion, lymphovascular space invasion, recurrence, and survival outcomes were statistically analysed. Univariate and multivariate Cox regression models were used to identify independent prognostic factors.
Results: High CLDN6 expression was detected in a subset of HGEC patients and was significantly associated with nonendometrioid histology (P = 0.026), advanced FIGO stage (P = 0.015), deep myometrial invasion (P = 0.038), and recurrence (P = 0.002). While Kaplan-Meier analysis did not reveal a statistically significant difference in disease-free survival or overall survival between the high CLDN6 expression group and the low CLDN6 expression group, multivariate Cox regression revealed that CLDN6 overexpression was an independent predictor of shorter disease-free survival [hazard ratio (HR) = 68.98, P = 0.022] and overall survival (HR = 24.023, P = 0.038).
Conclusion: CLDN6 overexpression is associated with aggressive tumor features and poor clinical outcomes in HGEC, suggesting its utility as a prognostic biomarker and potential therapeutic target.
背景:Claudin-6 (CLDN6)是一种通常局限于胚胎组织的紧密连接蛋白,在多种癌症中重新表达。然而,其在高级别子宫内膜癌(HGEC)中的预后意义尚不清楚。目的:探讨CLDN6在HGEC中的表达规律,并探讨其与临床病理参数及患者生存期的关系。方法:对80例HGEC患者进行福尔马林固定、石蜡包埋组织CLDN6表达的免疫组化分析。统计学分析CLDN6表达与组织学亚型、FIGO分期、肌层浸润深度、淋巴血管间隙浸润、复发及生存结局的关系。采用单因素和多因素Cox回归模型确定独立预后因素。结果:在HGEC患者中检测到高表达的CLDN6,并与非子宫内膜样组织学(P = 0.026)、晚期FIGO分期(P = 0.015)、深部肌层浸润(P = 0.038)和复发(P = 0.002)显著相关。Kaplan-Meier分析未显示CLDN6高表达组与低表达组无病生存期或总生存期差异有统计学意义,但多因素Cox回归分析显示,CLDN6过表达是较短无病生存期[风险比(HR) = 68.98, P = 0.022]和总生存期(HR = 24.023, P = 0.038)的独立预测因子。结论:CLDN6过表达与HGEC侵袭性肿瘤特征和不良临床结果相关,提示其作为预后生物标志物和潜在治疗靶点的应用价值。
{"title":"Tight junction disruption <i>via</i> claudin-6 overexpression promotes invasion and recurrence in high-grade endometrial tumors.","authors":"Noura A A Ebrahim, Tamer S Eissa, Mustafa A Hussein, Omnia Mohamed Korany, Nancy H Amin","doi":"10.5306/wjco.v16.i11.110257","DOIUrl":"10.5306/wjco.v16.i11.110257","url":null,"abstract":"<p><strong>Background: </strong>Claudin-6 (CLDN6), a tight junction protein typically restricted to embryonic tissues, is re-expressed in various cancers. However, its prognostic significance in high-grade endometrial carcinoma (HGEC) remains unclear.</p><p><strong>Aim: </strong>To investigate the expression pattern of CLDN6 in HGEC and assess its correlation with clinicopathological parameters and patient survival.</p><p><strong>Methods: </strong>Immunohistochemical analysis of CLDN6 expression was performed on formalin-fixed, paraffin-embedded tissues from 80 patients diagnosed with HGEC. Associations between CLDN6 expression and histological subtype, the International Federation of Gynecology and Obstetrics (FIGO) stage, depth of myometrial invasion, lymphovascular space invasion, recurrence, and survival outcomes were statistically analysed. Univariate and multivariate Cox regression models were used to identify independent prognostic factors.</p><p><strong>Results: </strong>High CLDN6 expression was detected in a subset of HGEC patients and was significantly associated with nonendometrioid histology (<i>P</i> = 0.026), advanced FIGO stage (<i>P</i> = 0.015), deep myometrial invasion (<i>P</i> = 0.038), and recurrence (<i>P</i> = 0.002). While Kaplan-Meier analysis did not reveal a statistically significant difference in disease-free survival or overall survival between the high CLDN6 expression group and the low CLDN6 expression group, multivariate Cox regression revealed that CLDN6 overexpression was an independent predictor of shorter disease-free survival [hazard ratio (HR) = 68.98, <i>P</i> = 0.022] and overall survival (HR = 24.023, <i>P</i> = 0.038).</p><p><strong>Conclusion: </strong>CLDN6 overexpression is associated with aggressive tumor features and poor clinical outcomes in HGEC, suggesting its utility as a prognostic biomarker and potential therapeutic target.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"110257"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678961/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-24DOI: 10.5306/wjco.v16.i11.112313
Pratishtha Gupta, Shama Parveen, Saurabh Kumar, Ana Ahtsham, Monisha Banerjee
Organelle integrity and maintenance of protein homeostasis and purpose is essential for fundamental equilibrium and survivability. Autophagy is the primary process which regulates the distribution of different cell loads to lysosomes for destruction and reuse. Extensive research illustrates the protective functions of autophagy against various diseases. Though in cancer, noticeably contrasting functions of autophagy have been evaluated in the prohibition of preliminary tumor evolution vs the continuance and, anabolic and catabolic variations of well-established and invasive tumors. Autophagy possesses numerous roles in tumor microenvironment (TME) establishment and associated immune cells function which is addressed in recent studies. Autophagic machinery which is employed in different autophagy-related pathways contributes to metastatic diseases and are distinct from classical autophagy. Therapeutic strategies based on the inhibition or induction of autophagy and related processes has helped in the designing of efficient anticancer drugs. According to the review, we evaluate and decipher the various purposes of autophagy and its association with autophagy mechanisms in course of tumor development, invasion and progression. We summarize the latest findings involving the role of these activities including tumor cells and TME and define further breakthrough in therapy aiming at autophagic activities in cancer.
{"title":"Attack cancer: Through autophagic modulations as suppressor or promoter.","authors":"Pratishtha Gupta, Shama Parveen, Saurabh Kumar, Ana Ahtsham, Monisha Banerjee","doi":"10.5306/wjco.v16.i11.112313","DOIUrl":"10.5306/wjco.v16.i11.112313","url":null,"abstract":"<p><p>Organelle integrity and maintenance of protein homeostasis and purpose is essential for fundamental equilibrium and survivability. Autophagy is the primary process which regulates the distribution of different cell loads to lysosomes for destruction and reuse. Extensive research illustrates the protective functions of autophagy against various diseases. Though in cancer, noticeably contrasting functions of autophagy have been evaluated in the prohibition of preliminary tumor evolution <i>vs</i> the continuance and, anabolic and catabolic variations of well-established and invasive tumors. Autophagy possesses numerous roles in tumor microenvironment (TME) establishment and associated immune cells function which is addressed in recent studies. Autophagic machinery which is employed in different autophagy-related pathways contributes to metastatic diseases and are distinct from classical autophagy. Therapeutic strategies based on the inhibition or induction of autophagy and related processes has helped in the designing of efficient anticancer drugs. According to the review, we evaluate and decipher the various purposes of autophagy and its association with autophagy mechanisms in course of tumor development, invasion and progression. We summarize the latest findings involving the role of these activities including tumor cells and TME and define further breakthrough in therapy aiming at autophagic activities in cancer.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"112313"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145702039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-24DOI: 10.5306/wjco.v16.i11.112029
Xiao-Ting Ma, Guang-Yu Yao, Jian-Li Li, Xi-Cheng Wang, Yi Ba
Background: Human epidermal growth factor receptor 2 (HER2)-positive gastric cancer (GC) represents a distinct molecular cancer subtype that is often associated with a poor prognosis. While perioperative chemotherapy regimens are currently the primary recommendation for locally advanced HER2-positive GC, combination therapies incorporating immune checkpoint inhibitors are under active investigation.
Case summary: The present case describes a patient with locally advanced HER2-positive GC who underwent perioperative treatment with chemotherapy combined with trastuzumab. Although significant tumor shrinkage was observed, surgical pathology results did not confirm the achievement of a pathological complete response. The current treatment strategies for advanced GC were also reviewed. Relevant case reports, retrospective studies, and prospective clinical trials were retrieved for analysis after searching the PubMed/MEDLINE, EMBASE, Cochrane Library, Web of Science, and American Society of Clinical Oncology/European Society for Medical Oncology conference abstracts between 2014 and 2024.
Conclusion: Large-scale phase III clinical trials are needed to verify the efficacy of combined neoadjuvant treatment application for GC.
背景:人表皮生长因子受体2 (HER2)阳性胃癌(GC)是一种独特的分子癌症亚型,通常与预后不良相关。虽然围手术期化疗方案目前是局部晚期her2阳性胃癌的主要推荐方案,但结合免疫检查点抑制剂的联合治疗正在积极研究中。病例总结:本病例描述了一位局部晚期her2阳性胃癌患者,接受化疗联合曲妥珠单抗围手术期治疗。虽然观察到明显的肿瘤缩小,但手术病理结果并没有证实病理完全缓解的实现。综述了目前晚期气相色谱的治疗策略。检索PubMed/MEDLINE、EMBASE、Cochrane图书馆、Web of Science和美国临床肿瘤学会/欧洲肿瘤医学学会2014 - 2024年会议摘要,检索相关病例报告、回顾性研究和前瞻性临床试验进行分析。结论:联合新辅助治疗应用于胃癌的疗效尚需大规模III期临床试验验证。
{"title":"Perioperative immunotherapy combined with standard therapy for human epidermal growth factor receptor 2-positive locally advanced gastric cancer: A case report.","authors":"Xiao-Ting Ma, Guang-Yu Yao, Jian-Li Li, Xi-Cheng Wang, Yi Ba","doi":"10.5306/wjco.v16.i11.112029","DOIUrl":"10.5306/wjco.v16.i11.112029","url":null,"abstract":"<p><strong>Background: </strong>Human epidermal growth factor receptor 2 (HER2)-positive gastric cancer (GC) represents a distinct molecular cancer subtype that is often associated with a poor prognosis. While perioperative chemotherapy regimens are currently the primary recommendation for locally advanced HER2-positive GC, combination therapies incorporating immune checkpoint inhibitors are under active investigation.</p><p><strong>Case summary: </strong>The present case describes a patient with locally advanced HER2-positive GC who underwent perioperative treatment with chemotherapy combined with trastuzumab. Although significant tumor shrinkage was observed, surgical pathology results did not confirm the achievement of a pathological complete response. The current treatment strategies for advanced GC were also reviewed. Relevant case reports, retrospective studies, and prospective clinical trials were retrieved for analysis after searching the PubMed/MEDLINE, EMBASE, Cochrane Library, Web of Science, and American Society of Clinical Oncology/European Society for Medical Oncology conference abstracts between 2014 and 2024.</p><p><strong>Conclusion: </strong>Large-scale phase III clinical trials are needed to verify the efficacy of combined neoadjuvant treatment application for GC.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"112029"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678912/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145702177","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Magnetic resonance imaging (MRI) and endoscopic ultrasonography (EUS) are recommended in combination for screening pancreatic cancer in high-risk individuals. However, in clinical practice, MRI and EUS are increasingly utilized for pancreatic surveillance during routine health examinations.
Aim: To investigate the feasibility of these imaging modalities for screening in low-risk individuals.
Methods: This retrospective study included patients at low risk for pancreatic cancer who underwent MRI or EUS at two health evaluation centers between March 2019 and December 2024. Basic characteristics, laboratory data, and imaging results were collected.
Results: A total of 3364 low-risk individuals underwent pancreatic screening: 1553 (46.1%) received MRI, and 1811 underwent EUS. No significant differences were observed in age or sex distribution between the groups. In imaging screening, EUS demonstrated a higher detection rate of abnormal pancreatic lesions (12.8% vs 2.6%; P < 0.001). MRI detected more cystic lesions than did EUS (P < 0.001). EUS identified smaller nodular lesions compared to MRI (9.2 mm vs 18.0 mm; P = 0.044). The MRI group had a higher number of confirmed intraductal papillary mucinous neoplasms (P = 0.031), whereas the EUS group identified more suspected branch-duct intraductal papillary mucinous neoplasms (P < 0.001). Pancreatic adenocarcinoma was found in three patients (0.08%), with no significant difference in detection rates between EUS and MRI (0.11% vs 0.06%; P = 0.656).
Conclusion: In low-risk individuals, MRI and EUS offer comparable effectiveness for pancreatic cancer surveillance. The choice of imaging strategy for health evaluation depends on cost considerations and degree of invasiveness.
背景:磁共振成像(MRI)和超声内镜检查(EUS)被推荐用于筛查高危人群的胰腺癌。然而,在临床实践中,MRI和EUS在常规健康检查中越来越多地用于胰腺监测。目的:探讨这些影像学方法在低危人群中筛查的可行性。方法:本回顾性研究纳入了2019年3月至2024年12月在两个健康评估中心接受MRI或EUS检查的低风险胰腺癌患者。收集基本特征、实验室数据和影像学结果。结果:共有3364名低危个体接受了胰腺筛查:1553名(46.1%)接受了MRI检查,1811名接受了EUS检查。各组之间的年龄和性别分布没有显著差异。影像学筛查中,EUS对胰腺异常病变的检出率更高(12.8% vs 2.6%; P < 0.001)。MRI比EUS检出更多囊性病变(P < 0.001)。与MRI相比,EUS发现较小的结节病变(9.2 mm vs 18.0 mm; P = 0.044)。MRI组确诊的导管内乳头状黏液性肿瘤较多(P = 0.031),而EUS组确诊的支管导管内乳头状黏液性肿瘤较多(P < 0.001)。3例患者发现胰腺腺癌(0.08%),EUS与MRI检出率差异无统计学意义(0.11% vs 0.06%; P = 0.656)。结论:在低危人群中,MRI和EUS对胰腺癌监测的有效性相当。健康评估的成像策略的选择取决于成本考虑和侵入程度。
{"title":"Role of endoscopic ultrasonography or magnetic resonance imaging for screening of pancreatic cancer in low-risk individuals.","authors":"Wei-Chen Lin, Lo-Yip Yu, Yang-Che Kuo, Chen-Wang Chang, Horng-Yuan Wang, Shou-Chuan Shih, Ching-Wei Chang, Hsiang-Hung Lin, Yi-Hsueh Chan, Ying-Chun Lin, Kuang-Chun Hu","doi":"10.5306/wjco.v16.i11.112030","DOIUrl":"10.5306/wjco.v16.i11.112030","url":null,"abstract":"<p><strong>Background: </strong>Magnetic resonance imaging (MRI) and endoscopic ultrasonography (EUS) are recommended in combination for screening pancreatic cancer in high-risk individuals. However, in clinical practice, MRI and EUS are increasingly utilized for pancreatic surveillance during routine health examinations.</p><p><strong>Aim: </strong>To investigate the feasibility of these imaging modalities for screening in low-risk individuals.</p><p><strong>Methods: </strong>This retrospective study included patients at low risk for pancreatic cancer who underwent MRI or EUS at two health evaluation centers between March 2019 and December 2024. Basic characteristics, laboratory data, and imaging results were collected.</p><p><strong>Results: </strong>A total of 3364 low-risk individuals underwent pancreatic screening: 1553 (46.1%) received MRI, and 1811 underwent EUS. No significant differences were observed in age or sex distribution between the groups. In imaging screening, EUS demonstrated a higher detection rate of abnormal pancreatic lesions (12.8% <i>vs</i> 2.6%; <i>P</i> < 0.001). MRI detected more cystic lesions than did EUS (<i>P</i> < 0.001). EUS identified smaller nodular lesions compared to MRI (9.2 mm <i>vs</i> 18.0 mm; <i>P</i> = 0.044). The MRI group had a higher number of confirmed intraductal papillary mucinous neoplasms (<i>P</i> = 0.031), whereas the EUS group identified more suspected branch-duct intraductal papillary mucinous neoplasms (<i>P</i> < 0.001). Pancreatic adenocarcinoma was found in three patients (0.08%), with no significant difference in detection rates between EUS and MRI (0.11% <i>vs</i> 0.06%; <i>P</i> = 0.656).</p><p><strong>Conclusion: </strong>In low-risk individuals, MRI and EUS offer comparable effectiveness for pancreatic cancer surveillance. The choice of imaging strategy for health evaluation depends on cost considerations and degree of invasiveness.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"112030"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678969/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701678","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Pseudoachalasia mimics primary achalasia in symptoms and diagnostic findings, as observed in gastroscopy and barium swallow studies. However, pseudoachalasia, often associated with malignancies like metastatic breast cancer, requires prompt differentiation to avoid misdiagnosis and inappropriate treatment. This report highlights a rare case of pseudoachalasia secondary to metastatic breast cancer and highlights the diagnostic value of esophageal motility changes.
Case summary: A 52-year-old woman presented with a one-year history of intermittent dysphagia following breast cancer surgery. Initial examinations suggested achalasia, but the patient's high-resolution manometry (HRM) results showed a rapid shift from ineffective esophageal motility to type II achalasia within four months. Further investigations revealed metastatic adenocarcinoma of the cardia, originating from the breast.
Conclusion: In patients with a history of malignancy, rapidly evolving esophageal motility abnormalities should raise suspicion of pseudoachalasia. HRM plays a crucial role in differentiating between primary and secondary achalasia. Early diagnosis through advanced imaging and pathology is essential for proper management.
{"title":"Dynamic esophageal manometry reveals pseudoachalasia secondary to metastatic breast cancer: A case report.","authors":"Hong-Yan Pan, Wei Liu, Wei Ding, Zhi-Mo Wang, Yan-Yan Feng, Ai-Hua Yu, Chun-Sheng Cheng","doi":"10.5306/wjco.v16.i11.111764","DOIUrl":"10.5306/wjco.v16.i11.111764","url":null,"abstract":"<p><strong>Background: </strong>Pseudoachalasia mimics primary achalasia in symptoms and diagnostic findings, as observed in gastroscopy and barium swallow studies. However, pseudoachalasia, often associated with malignancies like metastatic breast cancer, requires prompt differentiation to avoid misdiagnosis and inappropriate treatment. This report highlights a rare case of pseudoachalasia secondary to metastatic breast cancer and highlights the diagnostic value of esophageal motility changes.</p><p><strong>Case summary: </strong>A 52-year-old woman presented with a one-year history of intermittent dysphagia following breast cancer surgery. Initial examinations suggested achalasia, but the patient's high-resolution manometry (HRM) results showed a rapid shift from ineffective esophageal motility to type II achalasia within four months. Further investigations revealed metastatic adenocarcinoma of the cardia, originating from the breast.</p><p><strong>Conclusion: </strong>In patients with a history of malignancy, rapidly evolving esophageal motility abnormalities should raise suspicion of pseudoachalasia. HRM plays a crucial role in differentiating between primary and secondary achalasia. Early diagnosis through advanced imaging and pathology is essential for proper management.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"111764"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678947/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145702037","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-24DOI: 10.5306/wjco.v16.i11.110792
Mirza Muhammad Hadeed Khawar, Muhammad Hanan Abid, Muhammad Bilal A Cheema, Muneeb Khawar, Muneeba Shaukat, Muhammad Huzaifa A Khan, Muneeb Saifullah, Rimsha Noureen, Hamza Aka Khail, Ali Akram Qureshi, Muhammad Abbas Khokhar
Background: Dissection of the right paraesophageal lymph node (RPELN) in managing papillary thyroid carcinoma remains a contentious issue. This meta-analysis assesses previously established and novel risk factors associated with RPELN metastasis.
Aim: To evaluate previously established and novel risk factors associated with RPELN metastasis in patients with papillary thyroid carcinoma papillary thyroid carcinoma through a comprehensive meta-analysis.
Methods: We searched MEDLINE (via PubMed), ScienceDirect, Scopus and EMBASE up to December 2024. Studies were assessed using the Newcastle-Ottawa Scale. Statistical analysis was conducted with RevMan version 5.4, using the Q-test and I2-test for heterogeneity. Sensitivity was evaluated with the leave-one-out method, and publication bias with the Egger regression test and funnel plot.
Results: Of 2444 articles retrieved, 26 were included in our meta-analysis with 16427 patients. The RPELN metastasis rate was 12.98% [95% confidence interval (CI): 12.46%-13.50%]. The pooled results suggested that age < 55 years [odds ratio (OR) = 1.71, 95%CI: 1.35-2.16, P < 0.00001], sex (OR = 0.60, 95%CI: 0.54-0.67, P < 0.00001), tumor size 1 cm (OR = 3.37, 95%CI: 2.69-4.21, P < 0.00001), multifocality (OR = 1.81, 95%CI: 1.49-2.20, P < 0.00001), capsular invasion (OR = 2.94, 95%CI: 2.05-4.20, P < 0.00001), vascular invasion (OR = 2.16, 95%CI: 1.56-2.99, P < 0.00001), extra-thyroid extension (OR = 3.30, 95%CI: 1.82-5.98, P < 0.0001), central lymph node metastasis (OR = 7.77, 95%CI: 4.73-12.76, P < 0.00001), lateral lymph node metastasis (OR = 6.94, 95%CI: 6.11-7.89, P < 0.00001), Hashimoto thyroiditis (OR = 0.79, 95%CI: 0.69-0.92, P = 0.002), micro-calcifications (OR = 2.29, 95%CI: 1.20-4.37, P = 0.01), and echogenicity (OR = 0.62, 95%CI: 0.40-0.98, P = 0.04) should be considered with RPELN metastasis.
Conclusion: The male < 55, tumor size > 1 cm, multifocality, capsular and vascular invasion, extrathyroidal extension, lymph node metastasis, and Hashimoto thyroiditis were significantly associated with RPELN metastasis and should be carefully assessed during dissection.
背景:在甲状腺乳头状癌的治疗中,右侧食道旁淋巴结(RPELN)的切除仍然是一个有争议的问题。这项荟萃分析评估了与RPELN转移相关的先前建立的和新的危险因素。目的:通过一项综合荟萃分析,评估甲状腺乳头状癌患者RPELN转移的相关危险因素。方法:检索截止到2024年12月的MEDLINE(通过PubMed)、ScienceDirect、Scopus和EMBASE。研究采用纽卡斯尔-渥太华量表进行评估。采用RevMan version 5.4进行统计分析,采用q检验和I - 2检验异质性。用留一法评价敏感性,用Egger回归检验和漏斗图评价发表偏倚。结果:在检索到的2444篇文章中,有26篇纳入我们的荟萃分析,涉及16427名患者。RPELN转移率为12.98%[95%可信区间(CI): 12.46% ~ 13.50%]。合并结果显示,年龄< 55岁[比值比(OR) = 1.71, 95%CI: 1.35-2.16, P < 0.00001],性别(OR = 0.60, 95%CI: 0.54-0.67, P < 0.00001),肿瘤大小1 cm (OR = 3.37, 95%CI: 2.69-4.21, P < 0.00001),多灶性(OR = 1.81, 95%CI: 1.49-2.20, P < 0.00001),包膜侵犯(OR = 2.94, 95%CI: 2.05-4.20, P < 0.00001),血管侵犯(OR = 2.16, 95%CI: 1.56-2.99, P < 0.00001),甲状腺外扩张(OR = 3.30, 95%CI: P < 0.00001):1.82 ~ 5.98, P < 0.0001)、中央淋巴结转移(OR = 7.77, 95%CI: 4.73 ~ 12.76, P < 0.00001)、外侧淋巴结转移(OR = 6.94, 95%CI: 6.11 ~ 7.89, P < 0.00001)、桥本甲状腺炎(OR = 0.79, 95%CI: 0.69 ~ 0.92, P = 0.002)、微钙化(OR = 2.29, 95%CI: 1.20 ~ 4.37, P = 0.01)、回声增强(OR = 0.62, 95%CI: 0.40 ~ 0.98, P = 0.04)应作为RPELN转移的考虑因素。结论:男性< 55岁、肿瘤大小bbb1cm、多灶性、包膜及血管侵犯、甲状腺外展、淋巴结转移、桥本甲状腺炎与RPELN转移有显著相关性,应在解剖时仔细评估。
{"title":"Clinicopathological predictors of right para esophageal lymph node metastasis in papillary thyroid carcinoma: A systematic review and meta-analysis.","authors":"Mirza Muhammad Hadeed Khawar, Muhammad Hanan Abid, Muhammad Bilal A Cheema, Muneeb Khawar, Muneeba Shaukat, Muhammad Huzaifa A Khan, Muneeb Saifullah, Rimsha Noureen, Hamza Aka Khail, Ali Akram Qureshi, Muhammad Abbas Khokhar","doi":"10.5306/wjco.v16.i11.110792","DOIUrl":"10.5306/wjco.v16.i11.110792","url":null,"abstract":"<p><strong>Background: </strong>Dissection of the right paraesophageal lymph node (RPELN) in managing papillary thyroid carcinoma remains a contentious issue. This meta-analysis assesses previously established and novel risk factors associated with RPELN metastasis.</p><p><strong>Aim: </strong>To evaluate previously established and novel risk factors associated with RPELN metastasis in patients with papillary thyroid carcinoma papillary thyroid carcinoma through a comprehensive meta-analysis.</p><p><strong>Methods: </strong>We searched MEDLINE (<i>via</i> PubMed), ScienceDirect, Scopus and EMBASE up to December 2024. Studies were assessed using the Newcastle-Ottawa Scale. Statistical analysis was conducted with RevMan version 5.4, using the <i>Q</i>-test and <i>I</i> <sup>2</sup>-test for heterogeneity. Sensitivity was evaluated with the leave-one-out method, and publication bias with the Egger regression test and funnel plot.</p><p><strong>Results: </strong>Of 2444 articles retrieved, 26 were included in our meta-analysis with 16427 patients. The RPELN metastasis rate was 12.98% [95% confidence interval (CI): 12.46%-13.50%]. The pooled results suggested that age < 55 years [odds ratio (OR) = 1.71, 95%CI: 1.35-2.16, <i>P</i> < 0.00001], sex (OR = 0.60, 95%CI: 0.54-0.67, <i>P</i> < 0.00001), tumor size 1 cm (OR = 3.37, 95%CI: 2.69-4.21, <i>P</i> < 0.00001), multifocality (OR = 1.81, 95%CI: 1.49-2.20, <i>P</i> < 0.00001), capsular invasion (OR = 2.94, 95%CI: 2.05-4.20, <i>P</i> < 0.00001), vascular invasion (OR = 2.16, 95%CI: 1.56-2.99, <i>P</i> < 0.00001), extra-thyroid extension (OR = 3.30, 95%CI: 1.82-5.98, <i>P</i> < 0.0001), central lymph node metastasis (OR = 7.77, 95%CI: 4.73-12.76, <i>P</i> < 0.00001), lateral lymph node metastasis (OR = 6.94, 95%CI: 6.11-7.89, <i>P</i> < 0.00001), Hashimoto thyroiditis (OR = 0.79, 95%CI: 0.69-0.92, <i>P</i> = 0.002), micro-calcifications (OR = 2.29, 95%CI: 1.20-4.37, <i>P</i> = 0.01), and echogenicity (OR = 0.62, 95%CI: 0.40-0.98, <i>P</i> = 0.04) should be considered with RPELN metastasis.</p><p><strong>Conclusion: </strong>The male < 55, tumor size > 1 cm, multifocality, capsular and vascular invasion, extrathyroidal extension, lymph node metastasis, and Hashimoto thyroiditis were significantly associated with RPELN metastasis and should be carefully assessed during dissection.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"110792"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678945/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145702019","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-24DOI: 10.5306/wjco.v16.i11.108667
Samarah Arjumand, Asef Raj, Kazi Milenur Rahman Prattay, Humair Bin Md Omer, Faruque Azam
Chimeric antigen receptor T (CAR-T) cell therapy represents a major advance in cancer immunotherapy, offering targeted treatment options, particularly for hematologic malignancies. This review comprehensively explores the structural evolution, production processes, and cytotoxic mechanisms underlying CAR-T function. Therapy involves engineering autologous T cells with synthetic receptors that allow major histocompatibility complex-independent recognition of tumor-associated antigens. Key structural components such as antigen recognition domains, spacers, transmembrane, and intracellular domains are optimized to enhance specificity, persistence, and cytotoxicity. CAR-T therapy exerts antitumor effects via granzyme-perforin degranulation, Fas/Fas ligand signaling, and cytokine secretion. Over time, the development of second- to fifth-generation CARs has incorporated costimulatory molecules, transcriptional regulation, and logic-gated control to improve efficacy and safety. Additionally, novel engineering strategies such as dual CARs, tandem CARs, SynNotch systems, and universal or inhibitory CARs have expanded antigen targeting and reduced off-tumor toxicity. Emerging gene delivery technologies, including viral vectors, transposons, CRISPR/Cas9, and RNA-based electroporation, are improving CAR-T production. Despite notable clinical success, particularly in CD19- and B-cell maturation antigen-targeted therapies, CAR-T applications face challenges, including cell exhaustion, antigen escape, and therapy-induced toxicities, such as cytokine release syndrome and neurotoxicity. Ongoing efforts in engineering innovation, clinical trials, and regulatory support continue to shape CAR-T therapy into a safer, more precise tool for cancer treatment. This review highlights current advances while outlining the barriers and future prospects of CAR-T immunotherapy.
{"title":"Chimeric antigen receptor T cell therapy: Revolutionizing cancer treatment.","authors":"Samarah Arjumand, Asef Raj, Kazi Milenur Rahman Prattay, Humair Bin Md Omer, Faruque Azam","doi":"10.5306/wjco.v16.i11.108667","DOIUrl":"10.5306/wjco.v16.i11.108667","url":null,"abstract":"<p><p>Chimeric antigen receptor T (CAR-T) cell therapy represents a major advance in cancer immunotherapy, offering targeted treatment options, particularly for hematologic malignancies. This review comprehensively explores the structural evolution, production processes, and cytotoxic mechanisms underlying CAR-T function. Therapy involves engineering autologous T cells with synthetic receptors that allow major histocompatibility complex-independent recognition of tumor-associated antigens. Key structural components such as antigen recognition domains, spacers, transmembrane, and intracellular domains are optimized to enhance specificity, persistence, and cytotoxicity. CAR-T therapy exerts antitumor effects <i>via</i> granzyme-perforin degranulation, Fas/Fas ligand signaling, and cytokine secretion. Over time, the development of second- to fifth-generation CARs has incorporated costimulatory molecules, transcriptional regulation, and logic-gated control to improve efficacy and safety. Additionally, novel engineering strategies such as dual CARs, tandem CARs, SynNotch systems, and universal or inhibitory CARs have expanded antigen targeting and reduced off-tumor toxicity. Emerging gene delivery technologies, including viral vectors, transposons, CRISPR/Cas9, and RNA-based electroporation, are improving CAR-T production. Despite notable clinical success, particularly in CD19- and B-cell maturation antigen-targeted therapies, CAR-T applications face challenges, including cell exhaustion, antigen escape, and therapy-induced toxicities, such as cytokine release syndrome and neurotoxicity. Ongoing efforts in engineering innovation, clinical trials, and regulatory support continue to shape CAR-T therapy into a safer, more precise tool for cancer treatment. This review highlights current advances while outlining the barriers and future prospects of CAR-T immunotherapy.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 11","pages":"108667"},"PeriodicalIF":3.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12678908/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145702073","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}