{"title":"Enhanced recovery after surgery protocols in gastrectomy with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for gastric cancer.","authors":"Chawisa Nampoolsuksan, Thammawat Parakonthun","doi":"10.5306/wjco.v16.i8.107533","DOIUrl":"10.5306/wjco.v16.i8.107533","url":null,"abstract":"<p><p>Gastric cancer with peritoneal carcinomatosis (PC) remains a formidable challenge in oncological care, especially regarding surgical intervention. Integrating enhanced recovery after surgery (ERAS) protocols into gastrectomy with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy has emerged as a promising approach. This minireview explores the influence of ERAS on surgical and oncological outcomes in this multifaceted procedure. Recent evidence suggests that ERAS, comprising multimodal strategies, improves postoperative recovery, reduces complications, and enhances quality of life. It may also contribute to better survival outcomes by minimizing perioperative morbidity and thereby facilitating the timely initiation of adjuvant therapy. Mechanistically, ERAS promotes early mobilization, attenuates postoperative immunosuppression, and supports timely adjuvant therapies, which are crucial in managing carcinomatosis. This minireview underscores the importance of multidisciplinary collaboration and individualized patient care to maximize ERAS benefits. Large-scale, prospective investigations are warranted to validate these findings and refine ERAS protocols for this specialized patient cohort. Further research will facilitate ongoing advancements in oncological surgery and perioperative care, ultimately improving outcomes for patients with gastric cancer and PC.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"107533"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400245/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993756","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}
{"title":"Potential risks of histone deacetylase inhibitors in cancer therapeutics and feasible combination therapeutic strategies.","authors":"Shuai Xiao, Xiao-Zhen Xu, Meng Liao, Dan-Dan Song, Jing-Feng Tang, Ce-Fan Zhou","doi":"10.5306/wjco.v16.i8.108768","DOIUrl":"10.5306/wjco.v16.i8.108768","url":null,"abstract":"<p><p>Histone deacetylase inhibitors (HDACis), such as trichostatin A (TSA), have been recognized as promising anti-cancer agents due to their capacity to restore epigenetic regulation and reactivate tumor suppressor genes. However, emerging evidence indicates that unintended pro-metastatic effects may offset the therapeutic benefits of HDACis. Chen <i>et al</i> elucidate this paradox, demonstrating that TSA-induced hyperacetylation activates the BRD4/c-Myc/ER-stress axis, thereby promoting epithelial-mesenchymal transition and metastasis in esophageal squamous cell carcinoma (ESCC). Furthermore, they clarify the clinical significance of histone acetylation in the prognostic evaluation of ESCC. Their findings underscore the complexity of epigenetic therapies and highlight the necessity of reevaluating the associated risks and combinatorial therapeutic strategies with HDACi-based treatments. Here, we summarize the potential risks of HDACis therapy and discuss feasible combination therapeutic strategies.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"108768"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400233/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993747","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}
{"title":"Adding programmed death 1/programmed death ligand 1 inhibitors to first-line standard-of-care therapy for metastatic colorectal cancer: A meta-analysis.","authors":"Ting Zheng, Xing-Xing Li, Li Zhou, Jian-Jiang Jin","doi":"10.5306/wjco.v16.i8.106873","DOIUrl":"10.5306/wjco.v16.i8.106873","url":null,"abstract":"<p><strong>Background: </strong>In recent years, emerging clinical research has prioritized assessment of combined therapeutic efficacy and safety parameters when programmed death 1 or its ligand (PD-1/L1) inhibitors are incorporated into first-line standard-of-care (SOC) therapy for metastatic colorectal cancer (mCRC). However, data obtained from these trials demonstrated conflicting evidence concerning survival benefits and clinical outcomes.</p><p><strong>Aim: </strong>To evaluate the therapeutic impact and safety parameters of combining PD-1/L1 inhibitors with SOC protocols as first-line treatment for mCRC.</p><p><strong>Methods: </strong>Four biomedical databases (PubMed, Embase, Cochrane Library, Web of Science) were systematically interrogated to identify eligible studies published up to October 12, 2024. The analysis focused on evaluating the primary outcome of overall survival (OS) in the mCRC population with secondary outcomes of progression-free survival (PFS), overall response rate (ORR), and incidence rate of grade ≥ 3 adverse events. Additionally, we performed exploratory analyses in the microsatellite stable/mismatch repair-proficient (MSS/pMMR) subpopulation, based on a subset of the included studies. Subgroup analyses according to PD-1/L1 inhibitor use were conducted in both the overall population and the MSS/pMMR subgroup.</p><p><strong>Results: </strong>This pooled analysis incorporated six randomized controlled trials involving 675 patients with mCRC receiving first-line therapy. The combination of PD-1/L1 inhibitors with SOC regimens demonstrated a significant PFS advantage over SOC monotherapy in intention-to-treat populations [hazard ratio (HR) = 0.8, 95% confidence interval (CI): 0.65-0.98, <i>P</i> = 0.033]. Nevertheless, the MSS/pMMR subgroup showed no PFS benefit (HR = 0.83, 95%CI: 0.67-1.03, <i>P</i> = 0.091), and no cohort exhibited OS improvement (intention-to-treat: HR = 0.84, 95%CI: 0.66-1.05, <i>P</i> = 0.124; MSS/pMMR: HR = 0.79, 95%CI: 0.60-1.03, <i>P</i> = 0.083). Comparable outcomes were observed for ORR (risk ratio = 1.03, 95%CI: 0.90-1.17, <i>P</i> = 0.711) and incidence rate of grade ≥ 3 adverse events (risk ratio = 1.12, 95%CI: 0.93-1.36, <i>P</i> = 0.245) between treatment arms.</p><p><strong>Conclusion: </strong>The findings indicated that integrating PD-1/L1 blocking agents with SOC regimens for mCRC as first-line treatment failed to demonstrate significant improvements in ORR. Existing clinical data remain inadequate to establish OS advantages, particularly in patients with MSS/pMMR, despite exhibiting manageable toxicity profiles. Subsequent confirmation through rigorously designed phase III clinical trials remains essential to verify these therapeutic outcomes.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"106873"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400206/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993597","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}
{"title":"Cell reprogramming in cancer: Interplay of genetic, epigenetic mechanisms, and the tumor microenvironment in carcinogenesis and metastasis.","authors":"Santosh Shenoy","doi":"10.5306/wjco.v16.i8.106838","DOIUrl":"10.5306/wjco.v16.i8.106838","url":null,"abstract":"<p><p>Cell plasticity, also known as lineage plasticity, refers to the ability of a cell to reprogram and change its phenotypic identity in response to various cues. This phenomenon is context-dependent, playing a crucial role in embryonic development, tissue regeneration, and wound healing. However, when dysregulated, cell plasticity contributes to cancer initiation, progression, metastasis, and therapeutic resistance. Throughout different stages of tumor development, cancer cells exploit various forms of plasticity to evade normal regulatory mechanisms that govern cell division and homeostasis. Recent evidence highlights the complex interplay between genetic and epigenetic factors, the tumor microenvironment, and epithelial-to-mesenchymal transition in driving cancer cell plasticity. This dynamic reprogramming suggests that \"deregulated cell plasticity\" could be considered an additional hallmark of cancer. Advancements in next-generation sequencing and single-cell RNA analysis, combined with artificial intelligence technologies such as deep learning, along with Google's AlphaFold may help predict the trajectories of cancer cells. By predicting protein three-dimensional structures and identifying both active and potential allosteric binding sites, AlphaFold 2 can accelerate the development of new cancer drugs and therapies. For example, allosteric drugs, bind to the allosteric rather than the active sites, can induce conformational changes in proteins, affecting their activities. This can then alter the conformation of an active site that a drug-resistant mutation has created, permitting a blocked orthosteric drug to bind and this enables the design of more effective drugs that can synergize with traditional orthosteric drugs to bind and regain its efficacy. These innovations could provide deeper insights into the intricate mechanisms of cancer progression and resistance, ultimately paving the way for more precise, durable, and personalized oncologic treatments.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"106838"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400225/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993612","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}
{"title":"Exploration of preventive treatment for high risk patients with metachronous multiple esophageal squamous cell carcinoma: A case report.","authors":"Dan Chen, Ding-Fu Zhong, Dong Liu","doi":"10.5306/wjco.v16.i8.108371","DOIUrl":"10.5306/wjco.v16.i8.108371","url":null,"abstract":"<p><strong>Background: </strong>Metachronous multiple esophageal squamous cell carcinomas (ESCCs) may occur in some patients after endoscopic resection. Multiple dysplastic lesions in the esophagus increase risk of multiple squamous cell carcinomas (SCCs). Endoscopic imaging technology such as narrow band imaging (NBI), can detect early SCC. Lugol chromoendoscopy is also the conventional standard technique for detecting superficial ESCC. However, little is known about the interval from the first SCC to the metachronous SCC. Effective methods to prevent multiple metachronous SCCs are needed in survivors of esophageal SCC.</p><p><strong>Case summary: </strong>A 56-year-old man showed a slightly elevated reddish area in the middle thoracic esophagus at 30 cm from the incisors on gastroscopy for routine examination. Esophageal mucosa lesion was about 2.5 cm. NBI and magnifying gastroscopy confirmed intra-epithelial papillary loop type B-1 according to the Japan Esophageal Society Classification. Lugol chromoendoscopy was used to evaluate the dysplastic squamous epithelium in the esophagus. Biopsy pathology revealed severe dysplastic squamous epithelium. Computed tomography showed no lymph node metastasis. His complete blood test and tumor markers were within reference values. He had no history of alcohol consumption and smoking. Mucosal lesion was dissected by endoscopic submucosal dissection (ESD). Postoperative pathological results showed moderately differentiated squamous carcinoma. No cancer thrombus was seen in the vasculature, and the surrounding cut edge was not involved. The patient underwent radiotherapy within 2 months after ESD. The multiple Lugol-voiding lesions disappeared, and enhanced chest computed tomography revealed no lymph node metastasis.</p><p><strong>Conclusion: </strong>This is the first case of multiple dysplastic lesions of esophagus cured by radiotherapy. Radiotherapy after minimally invasive endoscopic treatment might be a safe and effective optional therapeutic strategy to prevent metachronous multiple esophageal SCCs.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"108371"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400183/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993742","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}
{"title":"Predictive model for sphincter preservation in lower rectal cancer.","authors":"Yajnadatta Sarangi, Ashok Kumar","doi":"10.5306/wjco.v16.i8.107596","DOIUrl":"10.5306/wjco.v16.i8.107596","url":null,"abstract":"<p><strong>Background: </strong>Low rectal cancer poses a significant surgical challenge because of its close proximity to the anal sphincter, often requiring radical resection with permanent colostomy to achieve oncological safety. Revisited rectal anatomy, advances in surgical techniques and neoadjuvant therapies have enabled the possibility of sphincter-preserving procedures, however, it is uniformly not applicable. Selecting appropriate candidates for sphincter preservation is crucial, as an ill-advised approach may compromise oncological outcome or lead to poor functional outcomes. Currently there is no consensus - which clinical, anatomical, or molecular factors most accurately predict the feasibility of sphincter-preserving surgery (SPS) in this subset of patients. By identifying these predictors, the study seeks to support improved patient selection, enhance surgical planning, and ultimately contribute to better functional and oncological outcomes in patients with low rectal cancer.</p><p><strong>Aim: </strong>To identify predictive factors that determine the feasibility of SPS in patients with low rectal cancer.</p><p><strong>Methods: </strong>A comprehensive literature search was conducted using PubMed/MEDLINE databases. The search focused on various factors influencing the feasibility of SPS in low rectal cancer. These included patient-related factors, anatomical considerations, findings from different imaging modalities, advancements in diagnostic tools and techniques, and the role of neoadjuvant chemoradiotherapy. The relevance of each factor in predicting the potential for sphincter preservation was critically analyzed and presented based on the current evidence.</p><p><strong>Results: </strong>Multiple studies have identified a range of predictive factors influencing the feasibility of SPS in low rectal cancer. Patient-related factors include age, sex, preoperative continence status, comorbidities, and body mass index. Anatomical considerations, such as tumor distance from the anal verge, involvement of the external anal sphincter, and levator ani muscles, also play a critical role. Additionally, a favourable response to neoadjuvant chemoradiotherapy has been associated with improved suitability for sphincter preservation. Several biomarkers, such as inflammatory markers like interleukins and C-reactive protein, as well as tumor markers like carcinoembryonic antigen, are important. Molecular markers, including BRAF and KRAS mutations and microsatellite instability status, have been linked to prognosis and may further guide decision-making regarding sphincter-preserving approaches. Artificial intelligence (AI) can further add in to select an ideal patient for sphincter preservation.</p><p><strong>Conclusion: </strong>SPS is feasible in low rectal cancer and depends on patient factors, tumor anatomy and biology, preoperative treatment response, and biomarkers. In addition, tools and technology including AI can further help in selecti","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"107596"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400217/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993770","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}
{"title":"Successful cure of a patient with tracheoesophageal fistula in cervical esophageal cancer: A case report and review of literature.","authors":"Haider Zuhair Waheed, Can-Qiang Huang, Yang-Yang Bao, Zhe Chen, Heng-Chao Chen, Zai-Zai Cao, Jiang-Tao Zhong, Peng Ye, Shui-Qiao Fu, Shui-Hong Zhou","doi":"10.5306/wjco.v16.i8.109217","DOIUrl":"10.5306/wjco.v16.i8.109217","url":null,"abstract":"<p><strong>Background: </strong>Tracheoesophageal fistula (TEF) is a life-threatening complication of advanced esophageal squamous cell carcinoma (ESCC). Cervical ESCC is rare and frequently diagnosed at an advanced stage. Managing cervical esophageal cancer (CEC) is challenging, requiring intervention by a multidisciplinary team (MDT) and innovative surgical management.</p><p><strong>Case summary: </strong>Here, we present a 59-year-old male patient with a 5-month history of CEC and difficulty eating for over 20 days, who developed TEF secondary to recurrent ESCC after chemoradiotherapy. He underwent total pharyngolaryngoesophagectomy, left thyroidectomy, and lymphadenectomy. Gastric pull-up was performed to restore gastrointestinal continuity, and a 7 cm × 5 cm supraclavicular artery island flap (SCAIF) was used to reconstruct the lower tracheal defect. Despite severe postoperative complications, he recovered by successful management by a MDT. A 7 cm × 6 cm pectoralis major myocutaneous flap was successfully used to repair the necrotic gastric conduit defect. The patient recovered, regaining the ability to eat and breathe effectively. At the 27-month follow-up, he was alive without recurrence or metastasis.</p><p><strong>Conclusion: </strong>This study highlights the efficacy of gastric pull-up and SCAIF reconstruction in managing TEF secondary to recurrent ESCC.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"109217"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400185/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993428","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}
Yun-Yun Xu, Wen-Jing Chen, Yu-Jie Cai, Feng Lin, Zhi-Peng He
{"title":"Rosmarinic acid as a chemosensitizer in colorectal cancer: Targeting nuclear factor-kappa B pathway to overcome chemoresistance.","authors":"Yun-Yun Xu, Wen-Jing Chen, Yu-Jie Cai, Feng Lin, Zhi-Peng He","doi":"10.5306/wjco.v16.i8.108279","DOIUrl":"10.5306/wjco.v16.i8.108279","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is the third most common malignancy. However, the efficacy of current treatment strategies remains limited. In recent years, monomeric compounds from traditional Chinese medicine have received extensive attention in cancer therapy. Rosmarinic acid (RA), a natural phenolic acid, has multiple biological activities and exhibits anti-oncogenic effects in several cancers. Liu <i>et al</i> previously uncovered that RA could serve as a dual-action therapeutic agent in CRC. By suppressing nuclear factor-kappa B signaling <i>via</i> direct inhibition of inhibitory kappa B kinase beta, RA not only impedes tumor progression but also synergizes with first-line chemotherapeutics (5-fluorouracil/oxaliplatin) to reverse drug resistance. The authors demonstrate RA's capacity to downregulate nuclear factor-kappa B-driven oncogenes and enhance chemotherapeutic cytotoxicity <i>in vitro</i> through integrative approaches, including molecular docking, luciferase assays, and functional validation. While these findings position RA as a cost-effective adjuvant in precision oncology, critical clinical translational gaps remain, including optimizing RA's <i>in vivo</i> bioavailability, validating systemic safety in combinatorial regimens, and elucidating its immunomodulatory effects within the tumor microenvironment. This underscores the urgency of bridging phytochemistry and clinical oncology, advocating for biomarker-driven animal studies and phase I trials to translate RA's potential into actionable CRC therapies. By addressing these hurdles, RA could emerge as a paradigm-shifting agent, harmonizing natural product efficacy with modern therapeutic precision.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"108279"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400210/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993432","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}
{"title":"Appendiceal mucinous neoplasms: Optimizing treatment strategies based on clinical, histological, and molecular features.","authors":"Atsushi Mitamura, Shingo Tsujinaka, Fumiyoshi Fujishima, Kentaro Sawada, Makoto Hikage, Tomoya Miura, Yoh Kitamura, Yuuri Hatsuzawa, Toru Nakano, Chikashi Shibata","doi":"10.5306/wjco.v16.i8.109088","DOIUrl":"10.5306/wjco.v16.i8.109088","url":null,"abstract":"<p><p>Appendiceal mucinous neoplasms (AMNs) are rare tumors originating from mucin-producing epithelial cells of the appendix. They can exhibit both benign and malignant behavior. They are often incidentally discovered during appendectomy. Clinical presentation ranges from asymptomatic to mimicking acute appendicitis. Histologically, noninvasive AMNs are classified as low-grade AMNs (LAMNs) or high-grade AMNs (HAMNs), whereas invasive tumors are categorized as mucinous adenocarcinomas. Although LAMNs and HAMNs are generally nonmalignant, rupture can lead to pseudomyxoma peritonei (PMP). Surgical resection is the primary diagnostic and therapeutic approach, with intraoperative assessment to prevent rupture. Treatment strategies vary based on findings and include appendectomy, right hemicolectomy, and cytoreductive surgery with hyperthermic intraperitoneal chemotherapy. Histological diagnosis relies on mucin detection, and immunohistochemical markers such as cytokeratin 20 (diffusely positive), cytokeratin 7 (often negative), mucin 5AC, and special AT-rich sequence-binding protein 2 assist in characterization. Molecular profiling frequently identifies <i>KRAS</i>, <i>GNAS</i>, and <i>TP53</i> mutations. <i>KRAS</i> mutations are generally associated with a favorable prognosis, whereas <i>GNAS</i> and <i>TP53</i> mutations correlate with poorer survival outcomes. These findings highlight the potential role of molecular profiling in guiding treatment strategies for AMN and PMP.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"109088"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400231/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993674","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}
{"title":"Integrating tumor location into artificial intelligence-based prognostic models in cancer.","authors":"Chen Wang, Meng-Yan Chen, Yu-Gang Wang, Min Shi","doi":"10.5306/wjco.v16.i8.109934","DOIUrl":"10.5306/wjco.v16.i8.109934","url":null,"abstract":"<p><p>This letter is a commentary on the findings of Huang <i>et al</i>, who emphasize the prognostic value of tumor location in gastric cancer. Analyzing data from 3287 patients using Kaplan-Meier and multivariate Cox models, the authors found that the tumor location correlated with patient prognosis following surgery. Patients with tumors situated nearer to the stomach's proximal end were associated with shorter survival periods and poorer outcomes. Notably, gender-based differences in tumor markers, particularly carbohydrate antigen 72-4, further highlight the need for sex-specific influence on the tumor location. Despite increasing recognition of tumor location as a prognostic factor, its role remains unclear in clinical prediction models for various cancers. This letter highlights the potential of incorporating tumor location into artificial intelligence -based prognostic tools to enhance prognostic models. It also outlines a stepwise framework for developing these models, from retrospective training to prospective multicenter validation and clinical implementation. In addition, it addresses the technical, ethical, and interoperability challenges critical to successful real-world prognosis.</p>","PeriodicalId":23802,"journal":{"name":"World journal of clinical oncology","volume":"16 8","pages":"109934"},"PeriodicalIF":3.2,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12400222/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144993745","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}