1
|
Yu X, Chen X, Chen W, Han X, Xie Q, Geng D, Guo G, Zhou L, Tang S, Chen J, Huang X, Zhong X. TGFβ2 Promotes the Construction of Fibrotic and Immunosuppressive Tumor Microenvironment in Pancreatic Adenocarcinoma: A Comprehensive Analysis. Mol Biotechnol 2025; 67:2562-2575. [PMID: 39044066 DOI: 10.1007/s12033-024-01219-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/10/2024] [Indexed: 07/25/2024]
Abstract
Pancreatic adenocarcinoma (PAAD) was characterized by dense fibrotic stroma and immunosuppressive tumor microenvironment (TME). TGFβ signaling pathways are highly activated in human cancers. However, the role of TGFβ2 in TME of PAAD remains to be elucidated. In this study, we showed that TGFβ2 was expressed at a relatively high level in PAAD tissues or cancer cells. Moreover, its high expression predicted unfavorable prognosis. In PAAD, gene set enrichment analysis showed that TGFβ2 correlated positively with leukocyte transendothelial migration, but negatively with aerobic metabolism, including oxidative phosphorylation. Results in Tumor and Immune System Interaction Database showed that TGFβ2 correlated with the infiltration of tumor-associated macrophages (TAMs), which could be attributed to that TGFβ2 promote CCL2 expression in PAAD. Moreover, correlation analysis showed that TGFβ2 could trigger cancer-associated fibroblasts (CAFs) activation in PAAD. The drug sensitivity analysis may indicate that patients with TGFβ2 high expression have higher sensitivity to chemotherapeutics, but the sensitivity to targeted drugs is still controversial. TGFβ2 could promote expansion of CAFs and infiltration of TAMs, thus participating in the construction of a fibrotic and immunosuppressive TME in PAAD. Targeting TGFβ2 could be a promising therapeutic approach, which needs to be elucidated by clinical and experimental evidences.
Collapse
Affiliation(s)
- Xiaofen Yu
- Department of Medical Oncology, Nanchang Third Hospital, Nanchang, 330000, Jiangxi, China
| | - Xuefen Chen
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Wanxian Chen
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Xiaosha Han
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Qihu Xie
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Deyi Geng
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Genghong Guo
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Linsa Zhou
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Shijie Tang
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Jiasheng Chen
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China.
| | - Xin Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Pancreatobiliary Surgery, Sun Yat-Sen University Cancer Center, GuangzhouGuangdong, 510060, China.
| | - Xiaoping Zhong
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China.
| |
Collapse
|
2
|
Atasoy BM, Demirel B, Ekşi Özdaş FN, Devran B, Kılıç ZN, Gül D. The role of radiotherapy planning images in monitoring malnutrition and predicting prognosis in head and neck cancer patients: a pilot study. Radiat Oncol 2025; 20:70. [PMID: 40319283 PMCID: PMC12049786 DOI: 10.1186/s13014-025-02645-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Adaptive treatment planning can be made in radiotherapy of head and neck cancer patients for reasons such as changes in tumor volume or weight loss. This study aims to find the role of treatment planning images in monitoring radiotherapy-induced malnutrition and predicting the malnutrition-induced prognosis in head and neck cancer patients. METHODS For this study, we analyzed 30 patients who received radiotherapy in our clinic between September 2018 and September 2021. Those patients, both regular and completed weekly dietitian counseling notes during radiotherapy and available adaptive radiotherapy planning images, were included in the analysis. All patients had weekly nutritional interventions, including nutritional and anthropometric changes in weight, height, body mass index (BMI), and lean body mass (LBM). Skeletal muscle volume, called cervical muscle gauge (CMG), was measured from the simulation images of beginning and adaptive radiotherapy. Inflammatory parameters, including the neutrophil-lymphocyte ratio (NLR), the platelet-lymphocyte ratio (PLR), and the systemic inflammatory index (SII), were also calculated from weekly total blood counts. For the analysis, anthropometric measurements were compared at the beginning and adaptive treatment time. Progression-free (PFS) and overall (OS) survival were calculated according to weight and CMG changes. RESULTS The median weight loss percentage was 4.8% (0 to 24%). The mean percentage of weight changes, LBM, and CMG were 6.33%, 3.47%, and 9.28%, respectively. Results indicated that BMI (p = 006), weight (p < 0.001), LBM (p < 0.001), and CMG (p = 0.057) decreased during radiotherapy. Hemoglobin levels decreased (p = 0.005), and inflammatory markers increased. There were significant correlations between weight and LBM (p < 0.0001) and CMG (p = 0.005) loss. The median follow-up was 26 months. Loss of weight (PFS; 65.5% vs. 35.7%, p = 0.09, OS; 73.7% vs. 32.1%, p = 0.09), LBM (PFS; 75% vs. 41.1%, p = 0.118, OS; 65.6% vs. 52%, p = 0.221) and CMG (PFS; 56.3% vs. 47.1%, p = 0.516, OS;76.9% vs. 32.4%, p = 0.059) negatively affected three-year survival. CONCLUSIONS Cervical muscle volume measurement may help predict malnutrition in patients receiving radiotherapy for head and neck cancer. Our study shows adaptive planning images may be used for this approach. In addition, this method may help to predict prognosis due to malnutrition in patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Beste M Atasoy
- Department of Radiation Oncology, Marmara University School of Medicine, Istanbul, Türkiye.
- S.B.-M.Ü. Pendik Eğitim ve Arastırma Hastanesi Radyasyon Onkolojisi Klinigi, Fevzi Cakmak Mah. Muhsin Yaziciıoglu cad. No: 6, Pendik/Istanbul, 34899, Türkiye.
| | - Birsen Demirel
- Faculty of Health Science Department of Nutrition and Dietetics, Ondokuz Mayıs University, Samsun, Türkiye
| | - Feyza Nur Ekşi Özdaş
- Nutrition and Dietetic Division, MH-Marmara University Pendik Education and Research Hospital, Istanbul, Türkiye
| | - Bennur Devran
- Department of Radiation Oncology, Marmara University School of Medicine, Istanbul, Türkiye
| | | | - Dilek Gül
- Radiation Oncology Clinic, MH-Marmara University Pendik Education and Research Hospital, Istanbul, Türkiye
| |
Collapse
|
3
|
Guo Q, Wang B, Gao X, Zhao P, Lv S. Predicting prognosis of patients with triple‑negative breast cancer undergoing neoadjuvant chemotherapy based on inflammatory status at different time points: A propensity score matching analysis. Oncol Lett 2025; 29:252. [PMID: 40201033 PMCID: PMC11977454 DOI: 10.3892/ol.2025.14998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer with limited targeted treatment options, making the identification of reliable prognostic markers crucial for improving patient outcomes. The present study aimed to assess the predictive ability of pre-chemotherapy and pre-surgery inflammatory status on the prognosis of patients with TNBC undergoing neoadjuvant therapy. A total of 422 patients with TNBC who received neoadjuvant chemotherapy at the Inner Mongolia People's Hospital between January 2017 and December 2022 were selected for analysis. Fasting venous blood samples were collected 1 day prior to chemotherapy and 1 day prior to surgery to assess and calculate inflammatory markers, including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune-inflammation index (SII) and systemic inflammation response index (SIRI). The optimal cut-off values of the inflammatory markers were determined using receiver operating characteristic curves. Survival analysis was used to evaluate the differences in survival and significant prognostic factors. Propensity score matching (PSM) analysis was performed to further asses the prognostic value of the relevant factors. Survival analysis indicated that patients with high pre-chemotherapy and pre-surgery NLR, PLR, SII and SIRI scores exhibited shorter overall survival (OS) rates compared with those with low scores (all P<0.05). Multivariate analysis revealed that tumor-node-metastasis stage, pathological complete response and pre-surgery SII were independent prognostic factors for OS. Following PSM, the area under the curve for SII was 0.642 and patients with high SII scores exhibited shorter OS rates than those with low scores (χ2=8.452; P=0.004). Therefore, these results indicated that both pre-chemotherapy and pre-surgery inflammatory statuses are associated with the OS of patients with TNBC undergoing neoadjuvant chemotherapy, notably pre-surgery SII.
Collapse
Affiliation(s)
- Qian Guo
- Department of Breast Surgical Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region 010017, P.R. China
| | - Bingping Wang
- Department of Breast Surgical Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region 010017, P.R. China
| | - Xinran Gao
- Department of Breast Surgical Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region 010017, P.R. China
| | - Pu Zhao
- Department of Breast Surgical Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region 010017, P.R. China
| | - Shuang Lv
- Department of Medical Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region 010017, P.R. China
| |
Collapse
|
4
|
Varadharaj V, Petersen W, Batra SK, Ponnusamy MP. Sugar symphony: glycosylation in cancer metabolism and stemness. Trends Cell Biol 2025; 35:412-425. [PMID: 39462722 PMCID: PMC12032065 DOI: 10.1016/j.tcb.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024]
Abstract
Glycosylation is a complex co-translational and post-translational modification (PTM) in eukaryotes that utilizes glycosyltransferases to generate a vast array of glycoconjugate structures. Recent studies have highlighted the role of glycans in regulating essential molecular, cellular, tissue, organ, and systemic biological processes with significant implications for human diseases, particularly cancer. The metabolic reliance of cancer, spanning tumor initiation, disease progression, and resistance to therapy, necessitates a range of uniquely altered cellular metabolic pathways. In addition, the intricate interplay between cell-intrinsic and -extrinsic mechanisms is exemplified by the communication between cancer cells, cancer stem cells (CSCs), cancer-associated fibroblasts (CAFs), and immune cells within the tumor microenvironment (TME). In this review article, we explore how differential glycosylation in cancer influences the metabolism and stemness features alongside new avenues in glycobiology.
Collapse
Affiliation(s)
- Venkatesh Varadharaj
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wyatt Petersen
- Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, NE, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, NE, USA.
| |
Collapse
|
5
|
Lv X, Zhang PB, Zhang EL, Yang S. Predictive factors and prognostic models for Hepatic arterial infusion chemotherapy in Hepatocellular carcinoma: a comprehensive review. World J Surg Oncol 2025; 23:166. [PMID: 40287734 PMCID: PMC12034129 DOI: 10.1186/s12957-025-03765-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/23/2025] [Indexed: 04/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent and lethal cancer, often diagnosed at advanced stages where traditional treatments such as surgical resection, liver transplantation, and locoregional therapies provide limited benefits. Hepatic arterial infusion chemotherapy (HAIC) has emerged as a promising treatment modality for advanced HCC, enhancing anti-tumor efficacy through targeted drug delivery while minimizing systemic side effects. However, the heterogeneous nature of HCC leads to variable responses to HAIC, highlighting the necessity for reliable predictive indicators to tailor personalized treatment strategies. This review explores the factors influencing HAIC success, including patient demographics, tumor characteristics, biomarkers, genomic profiles, and advanced imaging techniques such as radiomics and deep learning models. Additionally, the synergistic potential of HAIC combined with immunotherapy and molecular targeted therapies is examined, demonstrating improved survival outcomes. Prognostic scoring systems and nomograms that integrate clinical, molecular, and imaging data are discussed as superior tools for individualized prognostication compared to traditional staging systems. Understanding these predictors is essential for optimizing HAIC efficacy and enhancing survival and quality of life for patients with advanced HCC. Future research directions include large-scale prospective studies, integration of multi-omics data, and advancements in artificial intelligence to refine predictive models and further personalize treatment approaches.
Collapse
Affiliation(s)
- Xing Lv
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Peng-Bo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Er-Lei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - S Yang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
6
|
Huo Z, Chong F, Luo S, Li N, Tong N, Lu Z, Guo J, Zhang L, Lin X, Zhang M, Zhang H, Shi M, He X, Liu J, Song C, Shi H, Xu H. Potential framework of the Global Leadership Initiative in Sarcopenia (GLIS) criteria based on muscle mass and/or strength for predicting survival in cancer patients: A nationwide multicenter cohort study. Clin Nutr 2025; 49:187-201. [PMID: 40344922 DOI: 10.1016/j.clnu.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 04/03/2025] [Accepted: 04/20/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND The Global Leadership Initiative on Sarcopenia (GLIS) was proposed recently by creating a widely recognized conceptual definition of sarcopenia, however, the diagnostic framework of GLIS in cancer patients remains unclear. This study aims to evaluate the potential framework of GLIS based on muscle mass and/or strength in cancer patients. METHODS We performed a multicenter cohort study spanning from November 2012 to May 2020. Potential covariates were identified through univariate and multivariate analyses. The association between low muscle mass (LMM) and/or low muscle strength (LMS) with survival was estimated using Kaplan-Meier curves and Cox models. LMM was identified by lean mass index (LMI) or calf circumference (CC) while LMS was identified by hand grip strength (HGS). RESULTS A total of 6471 cancer patients were included, with a median follow-up of 50.0 months. Both LMM-LMI or LMS (HR = 1.56; 95%CI: 1.42, 1.71; p < 0.001) and LMM-LMI plus LMS (HR = 2.01; 95%CI: 1.65, 2.44; p < 0.001) were associated with a lower overall survival (OS) compared with patients without sarcopenia. Similarly, both LMM-CC or LMS group (HR = 1.51; 95%CI: 1.37, 1.67; p < 0.001) and LMM-CC plus LMS group (HR = 1.45; 95%CI: 1.28, 1.63; p < 0.001) were associated with a lower OS. Age, alcohol, Nutritional Risk Screening 2002 (NRS2002) score, Karnofsky Performance Status (KPS) score, Tumor Node Metastasis (TNM) stage, cancer category, albumin, direct bilirubin, anticancer therapy plus sex were introduced as covariates in fully-adjusted Cox model. Multivariable-adjusted Cox models revealed that LMM-LMI or LMS was an independent prognosis factor for cancer patients (HR = 1.18; 95%CI: 1.07, 1.31; p = 0.001). LMM-LMI plus LMS also was an independent predictor for survival among cancer patients (HR = 1.58; 95 % CI: 1.30, 1.94; p < 0.001). CONCLUSION The potential framework of GLIS based on muscle mass and/or strength was associated with survival in Chinese cancer patients. This research provides a simplified, clinically outcome-driven potential framework of sarcopenia, and offers new insights for the development of an operational definition of GLIS in the future.
Collapse
Affiliation(s)
- Zhenyu Huo
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Feifei Chong
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Siyu Luo
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Na Li
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Ning Tong
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Zongliang Lu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Jing Guo
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Ling Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Xin Lin
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Mengyuan Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Hongmei Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Muli Shi
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Xiumei He
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China
| | - Jie Liu
- Department of Clinical Nutrition, The Thirteenth People's Hospital of Chongqing, Chongqing, 400053, China
| | - Chunhua Song
- Department of Epidemiology, College of Public Health, Zhengzhou University, Henan, 450001, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China.
| | - Hongxia Xu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Chongqing Municipal Health Commission Key Laboratory of Intelligent Clinical Nutrition and Transformation, Chongqing, 400042, China.
| |
Collapse
|
7
|
Weber F, Reese KL, Pantel K, Smit DJ. Cancer-associated fibroblasts as a potential novel liquid biopsy marker in cancer patients. J Exp Clin Cancer Res 2025; 44:127. [PMID: 40259388 PMCID: PMC12010557 DOI: 10.1186/s13046-025-03387-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are tissue residing cells within the tumor microenvironment (TME). Stromal CAFs have been shown to be associated with poor prognosis and tumor progression in several solid tumor entities. Although the molecular mechanisms are not fully understood yet, a critical role within the TME through direct interaction with the tumor cells as well as other cells has been proposed. While most studies on CAFs focus on stromal CAFs, recent reports highlight the possibility of detecting circulating CAFs (cCAFs) in the blood. In contrast to invasive tissue biopsies for stromal CAF characterization, liquid biopsy allows a minimally invasive isolation of cCAFs. Furthermore, liquid biopsy methods could enable continuous monitoring of cCAFs in cancer patients and therefore may present a novel biomarker for solid tumors. In this work, we present an overview of cCAF studies currently available and summarize the liquid biopsy techniques for cCAF isolation and detection. Moreover, the future research directions in the emerging field are highlighted and the potential applications of cCAFs as novel biomarkers for solid tumor patients discussed.
Collapse
Affiliation(s)
- Franziska Weber
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Kim-Lea Reese
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Martinistraße 52, 20246, Hamburg, Germany
| | - Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
8
|
Wu J, Dong Y, Zhu W, Meng J, Zhang H, Fang C, Lin L. Capecitabine metronomic chemotherapy for metastatic colorectal cancer patients reaching NED: A protocol for a prospective, randomized, controlled trial. PLoS One 2025; 20:e0320591. [PMID: 40258007 PMCID: PMC12011264 DOI: 10.1371/journal.pone.0320591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 02/19/2025] [Indexed: 04/23/2025] Open
Abstract
INTRODUCTION An increasing number of patients with metastatic colorectal cancer (mCRC) have achieved no evidence of diseases (NED) status after surgery or other treatments. However, the latest guidelines for colorectal cancer do not recommend an appropriate treatment for patients with mCRC who achieve NED status. Capecitabine metronomic chemotherapy has the advantages of significant efficacy and minimal adverse reactions, it is a potential effective method for maintenance treatment for mCRC, but no RCTs have been reported. Therefore, we designed a randomized controlled trial to evaluate the efficacy and safety of capecitabine metronomic chemotherapy for mCRC patients who achieve NED. METHODS/DESIGN This study is a prospective, randomized controlled study that evaluates the efficacy and safety of capecitabine metronomic chemotherapy for patients with mCRC who achieve NED status. 240 eligible participants will be randomly assigned to either a capecitabine metronomic chemotherapy group or a "watch and wait" group at a 1:1 allocation ratio. Eligible patients diagnosed with stage IV mCRC, both the primary tumor and the metastases, are those who have achieved R0 resection (or complete destruction by ablation) and reached NED. Participants who are enrolled in the capecitabine group will receive capecitabine (500 mg/m2 body surface area twice daily) for 2 years. Meanwhile, those who are assigned to the control group will receive regular imaging examination and follow-up only. All participants will follow up for 1 year after receiving 2 years of intervention. The primary outcomes will be disease-free survival (DFS) from randomization, stratified by preoperative chemotherapy, metastatic organs, number of metastases, lenght of previous systemic treatment, response to previous chemotherapy. Secondary outcomes will include overall survival (OS), 1-year,2-year,3-year survival rate and adverse reactions. DISCUSSION As a potentially effective treatment, low-dose capecitabine metronomic chemotherapy has been explored in clinical practice. The results of this trial will provide evidence on the efficacy and safety of capecitabine metronomic chemotherapy for patients with mCRC who have reached NED status. TRIAL REGISTRATION Chinese Clinical Trial Registry (ChiCTR2100047149, protocol version number F2.0).
Collapse
Affiliation(s)
- Jiaming Wu
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Dong
- Clinical Research and Big Data Laboratory, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wanshan Zhu
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Jincheng Meng
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Huatang Zhang
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Cantu Fang
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
| | - Lizhu Lin
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
9
|
Tsutsumi C, Ohuchida K, Imamura M, Tan B, Shimada Y, Son K, Kosai T, Katayama N, Mochida Y, Hayashida S, Iwamoto C, Torata N, Horioka K, Shindo K, Mizuuchi Y, Ikenaga N, Nakata K, Oda Y, Nakamura M. Prognostic nutrition index reveals LAG3 in cytotoxic CD8+ T cells and MHC class II in gastric cancer cells. Cancer Immunol Immunother 2025; 74:176. [PMID: 40252096 PMCID: PMC12009253 DOI: 10.1007/s00262-025-04037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/25/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND The prognostic nutrition index (PNI) has recently been highlighted as a predictor of immune checkpoint (IC) inhibitor efficacy in gastric cancer (GC). Although LAG3, an IC molecule, has gained considerable attention, its association with PNI remains unexplored. MATERIALS AND METHODS We retrospectively analyzed clinical data from 796 GC patients who underwent radical gastrectomy to identify which previously reported nutritional index had the greatest impact on prognosis. Single-cell RNA sequencing was performed on 38 GC tissues, and multiplex immunofluorescence staining was conducted on 59 GC tissues to evaluate the relationship between nutritional indices and IC molecule expression in cytotoxic CD8-positive T cells. RESULTS A low preoperative PNI was identified as the strongest predictor of poor prognosis among the nutritional indices in GC patients. The expression of not only PDCD1 (encoding PD1) but also LAG3 in cytotoxic CD8-positive T cells was significantly higher in GC with low PNI compared to those with high PNI. Among cytotoxic CD8-positive T cells, the proportion of LAG3-positive cells was greater than that of PDCD1-positive cells, particularly in GC with low PNI, and most LAG3-positive cells did not co-express PDCD1. Additionally, the expression of MHC class II, a ligand for LAG3, was higher in GC cells with high levels of epithelial-mesenchymal transition-related molecules in GC with low PNI compared to those with high PNI. CONCLUSIONS PNI can reflect LAG3 expression in cytotoxic CD8-positive T cells and MHC class II expression in GC cells.
Collapse
Affiliation(s)
- Chikanori Tsutsumi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masaki Imamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Bryan Tan
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Shimada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiwa Son
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takaaki Kosai
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Katayama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Mochida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sayuri Hayashida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chika Iwamoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nobuhiro Torata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shindo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yusuke Mizuuchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
10
|
Roncati L. Adjuvant Metronomic Chemotherapy After Surgery in pT1-T2 N0 M0 HER2-Positive and ER/PR-Positive Breast Cancer Plus Targeted Therapy, Anti-Hormonal Therapy, and Radiotherapy, with or Without Immunotherapy: A New Operational Proposal. Cancers (Basel) 2025; 17:1323. [PMID: 40282499 PMCID: PMC12025911 DOI: 10.3390/cancers17081323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Breast cancer is the most common and deadly female-specific malignancy in the world. Four immunohistochemical subtypes are distinguished: luminal A, luminal B, HER2-positive, and triple-negative. In turn, the HER2-positive subtype presents two variants depending on the status of the hormone receptors. The variant that expresses them can benefit from both anti-HER2 and anti-hormonal therapy. Today, MCTP finds application in maintenance therapy after standard of care and in advanced breast cancer when the patient's clinical condition is already seriously compromised by metastatic disease; in this context, it is used as a first-line treatment, in pre-treated subjects, or as a rescue treatment. Here, the use of adjuvant oral MCTP after surgery at an early stage in HER-2 and hormone-positive local breast cancer is proposed, where effective treatment options are available, such as anti-HER2 therapy (e.g., trastuzumab, pertuzumab), anti-hormonal therapy (e.g., tamoxifen, letrozole), radiotherapy, and, in case of strong PD-1 positivity, immunotherapy.
Collapse
Affiliation(s)
- Luca Roncati
- Department of Life Sciences, Health, and Health Care Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
11
|
Ma Y, Zhu M, Zhang J, Jiao D, Hou Y, Chen X, Liu Z. Efficacy of adjuvant capecitabine in triple-negative breast cancer with residual disease after neoadjuvant therapy: a real-world study. Breast 2025; 81:104477. [PMID: 40250161 PMCID: PMC12036065 DOI: 10.1016/j.breast.2025.104477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE To determine the beneficiaries of capecitabine in patients with triple-negative breast cancer (TNBC) who failed to achieve pathological complete response (pCR) by analyzing the efficacy of the drug in different HER2 statuses and TNM stages. METHODS The Kaplan-Meier survival curve was plotted to estimate the effect of capecitabine therapy on disease-free survival (DFS) and overall survival (OS). Furthermore, the Cox proportional hazards model was used to analyze the factors that influence DFS and OS. RESULTS A total of 296 patients with TNBC who had non-pCR after undergoing neoadjuvant therapy (NAT) were included in this study. There were 152 patients (51.4 %) in the capecitabine group and 144 patients (48.6 %) in the no-capecitabine group. The 3-year DFS and OS rates of the capecitabine group were better than those of the no-capecitabine group (DFS 80.0 % vs. 68.0 % p = 0.012, OS 95.9 % vs. 86.9 % p = 0.011). In addition, the capecitabine group exhibited significantly better DFS and OS than the no-capecitabine group in the HER2-low (DFS p = 0.004, OS p = 0.009) and stage III (DFS p = 0.004, OS p = 0.008) populations but not in the HER2-0 or stage II population. CONCLUSION Adjuvant capecitabine therapy significantly improved the prognosis of patients with TNBC who had residual disease after NAT, and the improvements in the outcomes were significant in patients with HER2-low expression and stage III disease. Other effective treatment methods should be explored for patients with HER2-0 expression or stage II disease.
Collapse
Affiliation(s)
- Youzhao Ma
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Mingda Zhu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Jingyang Zhang
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Dechuang Jiao
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Yangyang Hou
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Xiuchun Chen
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China.
| |
Collapse
|
12
|
Zhang L, Li Q, Wu M, Feng X, Dai W, Chen P, Chen D, Zheng Z, Lin X, Wei G. TRIM22 governs tumorigenesis and protects against endometrial cancer-associated cachexia by inhibiting inflammatory response and adipose thermogenic activity. Cancer Metab 2025; 13:17. [PMID: 40200303 PMCID: PMC11980105 DOI: 10.1186/s40170-025-00386-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Endometrial cancer (EC) is one of the most common cancers in women, with a short overall survival and poor prognosis. Besides the biologically aggressive EC properties, Cancer-associated cachexia is the main factor. However, the detailed mechanism underlying EC-related cachexia and its harmful effects on EC progression and patient prognosis remains unclear. METHODS For clinical specimen and the vitro experiment, we detected TRIM22 expression level, EC patients' survival time, EC cell functional change, and adipose thermogenic changes to identify the function of TRIM22 in EC progression, EC-associated cachexia, and their molecular mechanisms. Then, for the vivo experiment, we exploited the xenografts in mice to identify the function of TRIM22 again, and to screen the drug therapeutic schedule. RESULTS Herein, we demonstrated that TRIM22 inhibited EC cell growth, invasion, and migration. Interleukin (IL)-6 mediated brown adipose tissue activation and white adipose tissue browning which induced EC-related cachexia. TRIM22 suppressed the EC cells' secretion of IL-6, and IL-6 mediated EC-related cachexia. Mechanistically, TRIM22 inhibited EC progression by suppressing the nucleotide-binding oligomerization domain 2(NOD2)/nuclear factor-kappaB (NF-κB) signaling pathway, with the purpose of impeding the production of IL-6. Moreover, we revealed that TRIM22 inhibited EC-associated cachexia by suppressing the IL-6/IL-6 receptor (IL-6R) signaling pathway. Therapeutically, we demonstrated that combination treatment with a TRIM22 inducer (progesterone) and a thermogenic inhibitor (IL-6R antibody) synergistically augmented the antitumor efficacy of carbotaxol (carboplatin and paclitaxel), in vivo. CONCLUSION Our data reveals that TRIM22-EC-IL-6-cachexia cross-communication has important clinical relevance and that the use of combined therapy holds great promise for enhancing the efficacy of anti-ECs. (Fig. graphical abstract).
Collapse
Affiliation(s)
- Liping Zhang
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Quanrong Li
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Meiting Wu
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiushan Feng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Weichao Dai
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Peifang Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Dezhao Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Zhiqun Zheng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiaoyan Lin
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
13
|
Liang CL, Li WD, Wang J. Preoperative immune-inflammation index in predicting the diagnosis and adverse pathological features of prostate cancer. Front Oncol 2025; 15:1537542. [PMID: 40248204 PMCID: PMC12003118 DOI: 10.3389/fonc.2025.1537542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Background Studies have reported that the systemic immune-inflammation index (SII) is positively correlated with genitourinary cancers. This study aims to explore the predictive value of preoperative immune-inflammation index for the diagnosis of prostate cancer and its adverse clinical characteristics. Methods This study analyzed patients who underwent their first prostate biopsy in the Urology Department of the Affiliated Hospital of Guangdong Medical University from January 2020 to January 2024. The predictive ability of SII for prostate cancer was evaluated, and the correlation between SII and localized prostate cancer and metastatic prostate cancer was explored. Results The SII in the PCa group was significantly higher than in the BPH group (558.14 vs. 515.06, P = 0.022), and SII independently predicted PCa risk (OR = 1.001, P = 0.013). Metastatic PCa patients exhibited higher SII compared to localized cases (694.80 vs. 437.95, P < 0.001), with multivariate analysis confirming SII, neutrophils, lymphocytes, and PSA as independent predictors of metastasis (OR = 1.000, P = 0.044). ROC analysis revealed limited predictive power of SII alone (AUC = 0.559), but its combination with PSA significantly improved accuracy (AUC = 0.791). A comprehensive model integrating SII, age, uric acid, and PSA achieved an AUC of 0.823, outperforming PSA alone (AUC = 0.777). Conclusions SII enhances the accuracy of PCa diagnosis and metastatic risk prediction when combined with PSA, demonstrating significant clinical utility. Although SII alone has limited predictive value, its cost-effectiveness and accessibility make it a valuable tool for stratified PCa management. Prospective studies are needed to validate its long-term prognostic significance.
Collapse
Affiliation(s)
| | | | - Jian Wang
- Department of Urology, Affiliated Hospital of Guangdong Medical University, First Clinical Medical College, Guangdong Medical University, Guangdong, China
| |
Collapse
|
14
|
Xu Y, Shen P, Zhu J, Qian D, Gu K, Mao Y, Ji S, Yang B, Zhao Y. Comprehensive malnutritional index for predicting clinical outcomes in locally advanced rectal cancer receiving neoadjuvant chemoradiotherapy. BIOMOLECULES & BIOMEDICINE 2025; 25:1079-1091. [PMID: 39508747 PMCID: PMC11984360 DOI: 10.17305/bb.2024.11188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024]
Abstract
The objective of this investigation was to assess the prognostic significance of the comprehensive malnutritional index (CNI) in patients with locally advanced rectal cancer (LARC) who underwent neoadjuvant chemoradiotherapy (nCRT) followed by surgery. A total of 240 LARC patients were recruited. The CNI was calculated using principal components analysis based on hemoglobin (Hb), total lymphocyte count (TLC), albumin (ALB), body mass index (BMI), and usual body weight percentage (UBW%). The patients were then categorized into two groups based on the median CNI value. Cox regression and survival analyses were performed. The CNI-low (120 cases) and CNI-high (120 cases) groups were classified based on the median CNI value. The results indicated that the CNI demonstrated superior predictive ability for disease-free survival (DFS) and overall survival (OS) compared to other malnutritional indexes. LARC patients in the CNI-high group had significantly longer DFS and OS compared to those in the CNI-low group. Multivariate analysis revealed that the CNI was an independent prognostic factor for DFS (hazard ratio [HR] = 0.49; 95% confidence interval [CI], 0.29-0.83; P = 0.008) and OS (HR = 0.30; 95% CI, 0.16-0.58; P < 0.001). Additionally, the CNI-high group benefited from postoperative chemotherapy (DFS: P = 0.029, OS: P = 0.024), while the CNI-low group did not show such benefits (DFS: P = 0.448, OS: P = 0.468). These findings suggest that the CNI could serve as a valuable prognostic indicator for LARC patients who undergo nCRT followed by surgery. Preoperative nutrition optimization is important for LARC patients.
Collapse
Affiliation(s)
- Yu Xu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Cancer Center, Wuxi, China
| | - Peipei Shen
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Cancer Center, Wuxi, China
| | - Jiahao Zhu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Cancer Center, Wuxi, China
| | - Danqi Qian
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Cancer Center, Wuxi, China
| | - Ke Gu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Cancer Center, Wuxi, China
| | - Yong Mao
- Wuxi Clinical Cancer Center, Wuxi, China
- Department of Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| | - Bo Yang
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Cancer Center, Wuxi, China
| | - Yutian Zhao
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Cancer Center, Wuxi, China
| |
Collapse
|
15
|
Wang F, Gong B, Chen L, Cao Y, Zhu L, Chai B, Wang J, Zhou G, Zheng C. Prognostic value of sarcopenia in patients with unresectable colorectal liver metastases after drug-eluting beads transcatheter arterial chemoembolization: a single center retrospective study. Sci Rep 2025; 15:11097. [PMID: 40169848 PMCID: PMC11962110 DOI: 10.1038/s41598-025-95782-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Sarcopenia is a prevalent condition in tumor patients and can potentially impact the prognosis of tumor treatment. This retrospective study aimed to evaluate the correlations between sarcopenia and the prognosis of patients with unresectable colorectal liver metastases (CRLM) received drug-eluting beads transcatheter arterial chemoembolization (DEB-TACE) therapy. From December 2018 to December 2023, unresectable CRLM patients who had already received second-line therapy from the Wuhan Union Hospital were involved in our study. Skeletal muscle mass was evaluated on CT at the L3 vertebra, and the optimal cut-off point for skeletal muscle index classification was determined using x-tile software. Overall survival (OS) and progression-free survival (PFS) were estimated using Kaplan-Meier analysis and Cox regression analysis. Seventy-one patients were included in the study, 34 with sarcopenia (sarcopenia group) and 37 without sarcopenia (non-sarcopenia group), respectively. The median PFS and OS was elevated in the non-sarcopenia group compared with the sarcopenia group (6.1 months versus 4.3 months, p = 0.012; 14.8 months versus 10.2 months, p < 0.001). The multivariate Cox regression analysis revealed that sarcopenia, extrahepatic metastases, and neutrophil-to-lymphocyte ratio (NLR) ≥ 5 were identified as independent risk factors for both PFS and OS. The advantages of non-sarcopenia in terms of OS were consistent across all subgroups examined. Additionally, the sarcopenia group exhibited a higher incidence of vomiting/nausea, fatigue, and abdominal pain following the DEB-TACE operation compared to the non-sarcopenia group. Sarcopenia demonstrated a substantial predictive value for both PFS and OS in unresectable CRLM patients who underwent DEB-TACE treatments. Besides, NLR > 5 and extrahepatic metastases were independent risk factors linked to a poorer prognosis. Furthermore, patients with sarcopenia may face an increased likelihood of experiencing adverse events following DEB-TACE treatments.
Collapse
Affiliation(s)
- Fuquan Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Licheng Zhu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Bin Chai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jihua Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Guofeng Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
16
|
Chen Y, Zhang B, Wang X, Chen Y, Anwar M, Fan J, Ma B. Prognostic value of preoperative modified Glasgow prognostic score in predicting overall survival in breast cancer patients: A retrospective cohort study. Oncol Lett 2025; 29:180. [PMID: 39990808 PMCID: PMC11843409 DOI: 10.3892/ol.2025.14926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/07/2025] [Indexed: 02/25/2025] Open
Abstract
The modified Glasgow prognostic score (mGPS), based on C-reactive protein and albumin levels, is an inflammation-based prognostic tool used in various cancers. However, related research in breast cancer is limited. The present study evaluated the prognostic value of the preoperative mGPS in predicting overall survival (OS) of patients with breast cancer undergoing surgery. A retrospective cohort study was conducted involving 300 patients with breast cancer with up to 10 years of follow-up. Patients were categorized into three groups based on mGPS scores of 0, 1 and 2, and their clinical and pathological data were collected. Kaplan-Meier survival analysis and Cox proportional hazards models were used to assess survival outcomes and identify risk factors associated with higher mGPS scores. A prognostic nomogram was developed based on multivariate analysis to predict 5- and 10-year OS. Patients with high mGPS scores showed significantly poor survival outcomes. The 5- and 10-year survival rates for mGPS 0, 1 and 2 were 80, 70 and 55%, and 71, 55 and 22%, respectively (P<0.001). Multivariate Cox analysis identified the mGPS, age, smoking, PAM50 and TNM stage as independent predictors of OS. The nomogram based on the mGPS demonstrated good predictive accuracy (concordance index: 0.81) and calibration. The preoperative mGPS is an independent prognostic factor for OS of patients with breast cancer. It is a simple, cost-effective tool that can aid in risk stratification and guide treatment strategies. Further validation in larger cohorts is recommended.
Collapse
Affiliation(s)
- Yi Chen
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Boxiang Zhang
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Xiaoli Wang
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Yanyan Chen
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Munawar Anwar
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Jingjing Fan
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Binlin Ma
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| |
Collapse
|
17
|
Lin HY, Chu PY. Mitochondrial calcium uniporter as biomarker and therapeutic target for breast cancer: Prognostication, immune microenvironment, epigenetic regulation and precision medicine. J Adv Res 2025; 70:445-461. [PMID: 38663838 PMCID: PMC11976406 DOI: 10.1016/j.jare.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/24/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
INTRODUCTION Mitochondrial calcium uniporter (MCU) is a central subunit of MCU complex that regulate the levels of calcium ions within mitochondria. A comprehensive understanding the implications of MCU in clinical prognostication, biological understandings and therapeutic opportunity of breast cancer (BC) is yet to be determined. OBJECTIVES This study aims to investigate the role of MCU in predictive performance, tumor progression, epigenetic regulation, shaping of tumor immune microenvironment, and pharmacogenetics and the development of anti-tumor therapy for BC. METHODS The downloaded TCGA datasets were used to identify predictive ability of MCU expressions via supervised learning principle. Functional enrichment, mutation landscape, immunological profile, drug sensitivity were examined using bioinformatics analysis and confirmed by experiments exploiting human specimens, in vitro and in vivo models. RESULTS MCU copy numbers increase with MCU gene expression. MCU expression, but not MCU genetic alterations, had a positive correlation with known BC prognostic markers. Higher MCU levels in BC showed modest efficacy in predicting overall survival. In addition, high MCU expression was associated with known BC prognostic markers and with malignancy. In BC tumor and sgRNA-treated cell lines, enrichment pathways identified the involvement of cell cycle and immunity. miR-29a was recognized as a negative epigenetic regulator of MCU. High MCU levels were associated with increased mutation levels in oncogene TP53 and tumor suppression gene CDH1, as well as with an immunosuppressive microenvironment. Sigle-cell sequencing indicated that MCU mostly mapped on to tumor cell and CD8 T-cells. Inter-databases verification further confirmed the aforementioned observation. miR-29a-mediated knockdown of MCU resulted in tumor suppression and mitochondrial dysfunction, as well as diminished metastasis. Furthermore, MCU present pharmacogenetic significance in cellular docetaxel sensitivity and in prediction of patients' response to chemotherapeutic regimen. CONCLUSION MCU shows significant implication in prognosis, outcome prediction, microenvironmental shaping and precision medicine for BC. miR-29a-mediated MCU inhibition exerts therapeutic effect in tumor growth and metastasis.
Collapse
Affiliation(s)
- Hung-Yu Lin
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Pei-Yi Chu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan; Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
18
|
He S, Meng J, Liang C, Wang Y, Qin X, Huang L, Wang R, Huang W. Prognostic implications of alternative splicing events and key splicing factors in head and neck squamous cell carcinoma. Discov Oncol 2025; 16:430. [PMID: 40159586 PMCID: PMC11955442 DOI: 10.1007/s12672-025-02214-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
The incidence of head and neck squamous cell carcinoma (HNSCC) remains high, accompanied by low 5-year survival rates. Identifying prognostic factors is essential for advancing personalized treatment approaches. Increasing evidence implicates aberrant alternative splicing (AS) plays a key role in tumor progression. Utilizing data from TCGA and TCGA SpliceSeq, prognosis-associated AS events were identified through Cox regression analysis. A prognostic risk model was developed via multivariate Cox and LASSO regression, with validation conducted using Kaplan-Meier survival analysis and ROC curve analysis. The correlation between splicing factors (SFs) and prognosis-associated AS events was analyzed using Pearson's method, followed by the construction of an SF-AS regulatory network. Key splicing factors (KSFs) were identified using Cytoscape software. Expression of KSFs in HNSCC was confirmed by quantitative PCR and Western blotting. SiRNA-mediated knockdown in HNSCC cell lines (HONE1, HN4, SAS) demonstrated effects on cell proliferation, invasion, and migration, as assessed by CCK8, colony formation, Transwell, and wound healing assays. Tumor growth was further evaluated in a subcutaneous tumor model in vivo. A total of 2347 survival-related AS events were identified, of which eleven were used to construct the prognostic model. Patients in the low-risk group exhibited significantly improved outcomes (P = 0e + 00), underscoring the model's predictive accuracy. Notably, DDX39B and PRPF39 emerged as key splicing factors, exhibiting high expression in HNSCC and correlating with poor prognosis, positioning them as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Siyi He
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Jiali Meng
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Chunyan Liang
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Yiru Wang
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Xinling Qin
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China
| | - Lulu Huang
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China
| | - Rensheng Wang
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China.
| | - Weimei Huang
- Department of Radiation Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi, China.
| |
Collapse
|
19
|
Jogiat UM, Bédard A, Baracos V, Turner SR, Eurich D, Filafilo H, Bédard EL. Thoracic muscle mass predicts survival among patients with locally advanced esophageal cancer. Clin Nutr 2025; 49:90-97. [PMID: 40253811 DOI: 10.1016/j.clnu.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND & AIMS There is limited literature evaluating muscle mass at the fourth thoracic (T4) vertebrae using computed tomography (CT) images, with no studies evaluating T4 muscle mass in esophageal cancer. METHODS In this retrospective cohort study, body composition analysis using skeletal muscle index (SMI) was conducted at T4 and L3. Overall survival (OS) and disease-free survival (DFS) were evaluated using Kaplan-Meier curves and log-rank tests, as well as multivariable cox proportional hazards models. Correlation analysis and evaluation of fixed and proportional bias was conducted. Low muscle mass was defined by the lowest quartile of the SMI distribution from the post-neoadjuvant CT: <30.4 cm2/m2 (females) and <42.2 cm2/m2 (males) for L3, and <35.4 cm2/m2 (females) and <52.6 cm2/m2 (males) for T4. RESULTS Of the 120 patients included, eight (8.2 %) patients had T4-low muscle mass at the staging CT which increased to 25 (25.8 %) at the post-neoadjuvant CT. On multivariable analysis, T4-low muscle mass was associated with worse overall survival (OS) (HR 2.51, 95 % CI 1.47-4.29, p = 0.001) and disease-free survival (DFS) (HR 1.88, 95 % CI 1.09-3.24, p = 0.022). T4-SMI was higher than L3-SMI at both the staging (65.4 ± 13.6 cm2/m2 versus 51.1 ± 10.0 cm2/m2, p < 0.001) and post-neoadjuvant (57.8 ± 12.7 cm2/m2 versus 45.8 ± 9.3 cm2/m2, p < 0.001) CT scans. The correlation (R-value) between T4 and L3 SMI was greater than 0.6 (0.62-0.81) for all staging intervals. CONCLUSION Our findings support using low muscle mass at T4 as a prognostic indicator for OS and DFS. These findings can be extrapolated to tumor groups, such as lung cancer, where L3-low muscle mass status is not routinely available.
Collapse
Affiliation(s)
- Uzair M Jogiat
- Division of Thoracic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Alexandre Bédard
- Division of Thoracic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Vickie Baracos
- Division of Palliative Care Medicine, Department of Oncology, University of Alberta, Edmonton, Canada
| | - Simon R Turner
- Division of Thoracic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Dean Eurich
- School of Public Health, University of Alberta, Edmonton, Canada
| | - Heather Filafilo
- Division of Thoracic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Eric Lr Bédard
- Division of Thoracic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada.
| |
Collapse
|
20
|
Liu C, Sun L, Niu N, Hou P, Chen G, Wang H, Zhang Z, Jiang X, Xu Q, Zhao Y, Wang Y, Shi Y, Liu M, Yang Y, Qian W, Wang J, Liu C. Molecular classification of hormone receptor-positive /HER2-positive breast cancer reveals potential neoadjuvant therapeutic strategies. Signal Transduct Target Ther 2025; 10:97. [PMID: 40133264 PMCID: PMC11937365 DOI: 10.1038/s41392-025-02181-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Significant heterogeneity exists in hormone receptor (HR)-positive/HER2-positive (HR+/HER2+) breast cancer, contributing to suboptimal pathological complete response rates with conventional neoadjuvant treatment regimens. Overcoming this challenge requires precise molecular classification, which is pivotal for the development of targeted therapies. We conducted molecular typing on a cohort of 211 patients with HR+/HER2+ breast cancer and performed a comprehensive analysis of the efficacy of various neoadjuvant treatment regimens. Our findings revealed four distinct molecular subtypes, each exhibiting unique characteristics and therapeutic implications. The HER2-enriched subtype, marked by activation of the HER2 signaling and hypoxia-inducible factor 1 (HIF-1) pathway, may benefit from intensified anti-HER2-targeted therapy. Estrogen receptor (ER)-activated subtype demonstrated potential sensitivity to combined therapeutic strategies targeting both ER and HER2 pathways. Characterized by high immune cell infiltration, the immunomodulatory subtype showed sensitivity to HER2-targeted antibody-drug conjugates (ADCs) and promise for immune checkpoint therapy. The highly heterogeneous subtype requires a multifaceted therapeutic approach. Organoid susceptibility assays suggested phosphoinositide 3-kinase inhibitors may be a potential treatment option. These findings underscore the importance of molecular subtyping in HR+/HER2+ breast cancer, offering a framework for developing precise and personalized treatment strategies. By addressing the heterogeneity of the disease, these approaches have the potential to optimize therapeutic outcomes and improve patient care.
Collapse
Affiliation(s)
- Chao Liu
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lisha Sun
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Nan Niu
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Pengjie Hou
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guanglei Chen
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hao Wang
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhan Zhang
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaofan Jiang
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qianshi Xu
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yafei Zhao
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yimin Wang
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Shi
- Department of Breast Surgery, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, China
| | - Mingxin Liu
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongliang Yang
- Shanghai General Medical Center, School of Clinical Medicine, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Wei Qian
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Jiandong Wang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Caigang Liu
- Cancer Stem Cell and Translation Medicine Lab, Shengjing Hospital of China Medical University, Shenyang, China.
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
21
|
Carvalho E, Canberk S, Schmitt F, Vale N. Molecular Subtypes and Mechanisms of Breast Cancer: Precision Medicine Approaches for Targeted Therapies. Cancers (Basel) 2025; 17:1102. [PMID: 40227634 PMCID: PMC11987866 DOI: 10.3390/cancers17071102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer remains one of the most prevalent diseases worldwide, primarily affecting women. Its heterogeneous nature poses a significant challenge in the development of effective and targeted treatments. Molecular characterization has enabled breast cancer to be classified into four main subtypes: luminal A, luminal B, HER2-positive, and triple-negative breast cancer, based on hormone receptor expression and HER2 status. A deeper understanding of these molecular markers and their associated signaling pathways, such as MAPK and PI3K/AKT, is essential for improving prognosis and optimizing treatment strategies. Currently, several therapeutic agents are utilized in neoadjuvant and adjuvant therapies, often in combination with surgical interventions. However, emerging evidence highlights the growing challenge of drug resistance, which significantly limits the efficacy of existing treatments. Addressing this issue may require innovative approaches, including combination therapies and precision medicine strategies, tailored to the molecular profile of each patient. Therefore, a comprehensive understanding of the pathophysiologic mechanisms driving breast cancer progression and resistance is crucial for the development of advanced targeted therapies with greater precision and efficacy. This review aims to explore recent advancements in molecular research related to breast cancer subtypes and provide a critical analysis of current therapeutic approaches within the framework of precision medicine.
Collapse
Affiliation(s)
- Eduarda Carvalho
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
| | - Sule Canberk
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Fernando Schmitt
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
22
|
Zeng XY, Li Y, Ma J, Zuo ZC, Jiang MJ, Liang ZG, Chen KH, Li L, Qu S, Liu Y, Zhu XD. Prognostic nomogram for synchronous metastatic nasopharyngeal carcinoma: a retrospective multicentre study. Radiat Oncol 2025; 20:42. [PMID: 40114228 PMCID: PMC11927208 DOI: 10.1186/s13014-025-02602-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Patients with synchronous metastatic nasopharyngeal carcinoma (smNPC) exhibit significant heterogeneity, and clinical prognostic models suitable for this cohort remain limited. We aimed to develop a prognostic prediction tool to facilitate personalised prognostic assessments and inform treatment decisions for these patients. METHODS This retrospective multicentre study enrolled 556 patients with smNPC. The training cohort comprised 386 patients from Guangxi Medical University Cancer Hospital, while the external validation cohort comprised 170 patients from Wuzhou Red Cross Hospital and Xiangtan Central Hospital. We applied the Cox proportional hazards model to determine factors associated with overall survival (OS). A nomogram prognostic model was developed to predict OS based on the identified prognostic factors. The model's predictive performance was evaluated for discrimination and calibration, and patients were stratified based on their calculated prognostic risk scores. Kaplan-Meier survival curves were employed to assess prognostic differences across the stratified groups. RESULTS Multivariate analysis identified that M classification, primary tumour radiotherapy, and immunotherapy were significantly associated with OS. A prognostic nomogram integrating these variables exhibited good discrimination (C-index: 0.743) and calibration, which was validated in an external validation cohort. Patients stratified by the model-derived risk scores into high- and low-risk groups showed a significant difference in survival disparity. CONCLUSIONS We established a nomogram prognostic model that effectively facilitated individualised prognostic prediction and risk stratification in patients with smNPC, thereby assisting clinicians in treatment decision-making.
Collapse
Affiliation(s)
- Xiao-Yi Zeng
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
- Department of Radiation Oncology, Wuzhou Red Cross Hospital, Wuzhou, People's Republic of China
| | - Ye Li
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Jie Ma
- Medical Imaging Department, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Zhi-Chao Zuo
- Department of Radiology, Xiangtan Central Hospital, Xiangtan, People's Republic of China
| | - Meng-Jie Jiang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Zhong-Guo Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Kai-Hua Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Ling Li
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, People's Republic of China
| | - Song Qu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, People's Republic of China
| | - Yang Liu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China.
| | - Xiao-Dong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China.
- Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, People's Republic of China.
- Department of Oncology, Affiliated Wu-Ming Hospital of Guangxi Medical University, Nanning, People's Republic of China.
| |
Collapse
|
23
|
Zheng L, Cai W, Ke Y, Hu X, Yang C, Zhang R, Wu H, Liu D, Yu H, Wu C. Cancer‑associated fibroblasts: a pivotal regulator of tumor microenvironment in the context of radiotherapy. Cell Commun Signal 2025; 23:147. [PMID: 40114180 PMCID: PMC11927177 DOI: 10.1186/s12964-025-02138-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND In the course of tumor treatment, radiation therapy (RT) not only kills cancer cells, but also induces complex biological effects in non-malignant cells around cancer cells. These biological effects such as angiogenesis, changes in stromal composition and immune cell infiltration remodel the tumor microenvironment (TME). As one of the major components of the TME, Cancer‑associated fibroblasts (CAFs) are not only involved in tumorigenesis, progression, recurrence, and metastasis but also regulate the tumor-associated immune microenvironment. CAFs and tumor cells or immune cells have complex intercellular communication in the context of tumor radiation. MAIN CONTENT Different cellular precursors, spatial location differences, absence of specific markers, and advances in single-cell sequencing technology have gradually made the abundant heterogeneity of CAFs well known. Due to unique radioresistance properties, CAFs can survive under high doses of ionizing radiation. However, radiation can induce phenotypic and functional changes in CAFs and further act on tumor cells and immune cells to promote or inhibit tumor progression. To date, the effect of RT on CAFs and the effect of irradiated CAFs on tumor progression and TME are still not well defined. CONCLUSION In this review, we review the origin, phenotypic, and functional heterogeneity of CAFs and describe the effects of RT on CAFs, focusing on the mutual crosstalk between CAFs and tumor or immune cells after radiation. We also discuss emerging strategies for targeted CAFs therapy.
Collapse
Affiliation(s)
- Linhui Zheng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Wenqi Cai
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Xiaoyan Hu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Chunqian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Runze Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Huachao Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Dong Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Haijun Yu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China.
| | - Chaoyan Wu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
| |
Collapse
|
24
|
Carrilho LAO, Juliani FL, Moreira RCDL, Dias Guerra L, Santos FS, Padilha DMDH, Branbilla SR, Horita VN, Novaes DML, Antunes-Correa LM, Lima CSP, Mendes MCS, Carvalheira JBC. Adipose tissue characteristics as a new prognosis marker of patients with locally advanced head and neck cancer. Front Nutr 2025; 12:1472634. [PMID: 40161297 PMCID: PMC11949816 DOI: 10.3389/fnut.2025.1472634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Background Patients with head and neck cancer (HNC) are at increased risk of malnutrition due to the presence of tumor and treatments. Body composition is a prognostic factor in these patients. However, the relationship between adipose tissue characteristics and survival in HNC is still unclear. Objective To evaluate the associations of adiposity, the radiodensity of adipose tissue and muscularity with the prognosis of patients with locally advanced HNC undergoing to chemoradiotherapy. Methods This retrospective study included 132 patients diagnosed with locally advanced HNC. Body composition assessment was performed using computed tomography (CT) images at the level of the third cervical vertebra (C3). The total adipose tissue radiodensity (TATR), the total adipose tissue index (TATI) and skeletal muscle index (SMI) were evaluated. The primary outcome was overall survival (OS). Results Patients in the highest TATI tertile had a lower risk of mortality when compared to those in the lowest tertile, HR: 0.56, 95% confidence Interval (CI): 0.32-0.96; p = 0.039. The highest TATR tertile was not associated with death. Patients with greater adiposity had a higher median survival compared to patients with medium and lower TATI (p = 0.0193). Individuals with lower TATI had lower energy intake than patients with higher TATI (p = 0.03). Additionally, patients with low muscularity had worse OS in the multivariable analysis (HR: 1.77, 95% CI: 1.01-3.07; p = 0.044). Conclusion In patients with locally advanced HNC, our findings underscore the significance of elevated adiposity, beyond maintained muscularity, as independent protective factors for overall survival. Our study highlights the critical importance of assessing body composition and initiating early nutritional interventions to improve the prognosis of these patients.
Collapse
Affiliation(s)
- Larissa Ariel Oliveira Carrilho
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Fabiana Lascala Juliani
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Rafaella Caroline de Lellis Moreira
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Livia Dias Guerra
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Fernanda Silva Santos
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Daniela Morais de Holanda Padilha
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
- Nestlé Health Science, Vevey, Switzerland
| | - Sandra Regina Branbilla
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Vivian Naomi Horita
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Davi Magalhães Leite Novaes
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | | | - Carmem Silvia Passos Lima
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Maria Carolina Santos Mendes
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - José Barreto Campello Carvalheira
- Department of Radiology and Oncology, Division of Oncology, School of Medical Sciences, State University of Campinas (UNICAMP), São Paulo, Brazil
| |
Collapse
|
25
|
Liu F, Ma C, Chen M, Chen K, Zhu L, Li L, Zhu X, Qu S, Yan C. A Nomogram Based on Tumor Response to Induction Chemotherapy and Plasma Epstein-Barr Virus DNA Level after Induction Chemotherapy to Explore Individualized Treatment of Patients with Locally Advanced Nasopharyngeal Carcinoma. J Inflamm Res 2025; 18:3677-3693. [PMID: 40099001 PMCID: PMC11913028 DOI: 10.2147/jir.s507926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose To explore the influence of Epstein-Barr virus (EBV) DNA levels before and after induction chemotherapy (IC), tumor response to IC, and baseline factors on overall survival (OS) in patients with locally advanced nasopharyngeal carcinoma (LA-NPC). A nomogram was subsequently constructed to explore the individualized optimal cumulative cisplatin dose (CCD) in concurrent chemoradiotherapy (CCRT). Methods A total of 581 LA-NPC patients were included, randomly divided into training and validation cohorts in a 7:3 ratio. In the training cohort, a nomogram was subsequently established based on multivariate Cox regression analysis and then validated. Subsequently, patients were classified into different risk groups based on the nomogram, and the impact of different levels of CCD on survival outcomes was evaluated. Results EBV DNA levels after IC, tumor response to IC, age, and LDH were independent prognostic factors of OS. Schoenfeld residual analysis indicated overall satisfaction of the proportional hazards assumption for the Cox regression model. The C-index of the nomogram was 0.758 (95% CI: 0.695-0.821) for the training cohort and 0.701 (95% CI: 0.589-0.813) for the validation cohort. Calibration curves demonstrated good correlation between the nomogram and actual survival outcomes. DCA confirmed the clinical utility enhancement of the nomogram over the TNM staging system. For OS, patients in the medium/high-risk group with a CCD > 200 mg/m² had better outcomes than those with CCD ≤ 200 mg/m², although the difference was not statistically significant (p = 0.097). No significant difference was observed in local relapse-free survival (LRFS), distant metastasis-free survival (DMFS), and progression-free survival (PFS) across various levels of CCD in different risk subgroups (p > 0.05). Conclusion The nomogram based on EBV DNA levels after IC, tumor response, LDH, and age effectively predicts OS in LA-NPC patients, aids in risk stratification, and may guide treatment decisions.
Collapse
Affiliation(s)
- Fushuang Liu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Chengxian Ma
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Meiwen Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Kaihua Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Liru Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Ling Li
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, 530199, People's Republic of China
| | - Song Qu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Chang Yan
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| |
Collapse
|
26
|
Liao XW, Gao JB, Sun H, Chen HD, Zheng MH, Han L, Chen XG, Su YN, Pan DL, Wu M, Cai SL, Lin X, Chen GZ. Prediction of neoadjuvant chemotherapy efficacy and prognostic biomarker analysis in patients with triple-negative breast cancer. Front Pharmacol 2025; 16:1553831. [PMID: 40144666 PMCID: PMC11937642 DOI: 10.3389/fphar.2025.1553831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Background Neoadjuvant chemotherapy has become a common and effective treatment modality for triple-negative breast cancer (TNBC). The primary goal is to reduce the size of the primary tumor, enabling breast-conserving surgery, axillary preservation, and a transition to operability, thereby providing patients with more therapeutic options. Although neoadjuvant chemotherapy (NAC) has demonstrated favorable outcomes in clinical practice, predicting its efficacy and prognostic value in TNBC remains a key challenge in current clinical research. Methods This study included 248 TNBC patients who received NAC at two breast cancer treatment centers. By employing a modeling validation approach, we aim to explore predictors of treatment efficacy and potential prognostic biomarkers associated with NAC. Results In the multivariable analysis of the training set, the factors predicting the pathological complete response (pCR) to NAC in TNBC patients include high biopsy-sTILs expression, biopsy-Ki67 > 20%, and positive expression of biopsy-androgen receptor (AR). The factors predicting disease-free survival (DFS) are ypN3, high postoperative sTIL expression, receipt of postoperative radiotherapy, and effective NAC. The factors predicting overall survival (OS) include ypN2, ypN3, high postoperative sTIL expression, postoperative Ki67 > 20%, receipt of postoperative radiotherapy, and effective NAC. The C-indices in the training and validation sets for the prediction of pCR using the nomogram were 0.729 and 0.816, respectively. The C-indices for predicting DFS were 0.895 and 0.865, respectively. The C-indices for predicting OS were 0.899 and 0.860, respectively. Conclusion This study established and validated a nomogram model predicting the pCR, DFS, and OS in TNBC patients undergoing NAC. This model demonstrates good discrimination and accuracy.
Collapse
Affiliation(s)
- Xiao-Wen Liao
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jia-Bin Gao
- General Surgery Department of Pengyang County People’s Hospital, Guyuan, Ningxia, China
| | - Hong Sun
- Department of Pharmacy, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Hong-Dan Chen
- First Department of Cadre Clinic, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Min-Hui Zheng
- Oral and Maxillofacial Surgery Thyroid and Hernia Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Lei Han
- Department of Breast Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Xiao-Geng Chen
- Department of Breast Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yu-Nan Su
- Department of Emergency, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Ding-Long Pan
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Min Wu
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shuang-Long Cai
- Department of Breast Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Xiuquan Lin
- Department for Chronic and Noncommunicable Disease Control and Prevention, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian, China
| | - Guo-Zhong Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
27
|
Su H, Chen L, Wu J, Cheng Z, Li J, Ren Y, Xu J, Dang Y, Zheng M, Cao Y, Gao J, Dai C, Hu X, Xie H, Chen J, Luo T, Zhu J, Wu C, Sha W, Chen C, Liu H. Proteogenomic characterization reveals tumorigenesis and progression of lung cancer manifested as subsolid nodules. Nat Commun 2025; 16:2414. [PMID: 40069142 PMCID: PMC11897189 DOI: 10.1038/s41467-025-57364-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Lung adenocarcinoma (LUAD) radiologically displayed as subsolid nodules (SSNs) is prevalent. Nevertheless, the precise clinical management of SSNs necessitates a profound understanding of their tumorigenesis and progression. Here, we analyze 66 LUAD displayed as SSNs covering 3 histological stages including adenocarcinoma in situ (AIS), minimally invasive adenocarcinoma (MIA) and invasive adenocarcinoma (IAC) by incorporating genomics, proteomics, phosphoproteomics and glycoproteomics. Intriguingly, cholesterol metabolism is aberrantly regulated in the preneoplastic AIS stage. Importantly, target ablation of proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the initiation of LUAD. Furthermore, sustained endoplasmic reticulum stress is demonstrated to be a hallmark and a reliable biomarker of AIS progression to IAC. Consistently, target promotion of ER stress profoundly retards LUAD progression. Our study provides comprehensive proteogenomic landscape of SSNs, sheds lights on the tumorigenesis and progression of SSNs and suggests preventive and therapeutic strategies for LUAD.
Collapse
Affiliation(s)
- Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Li Chen
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhongyi Cheng
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jing Li
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Junfang Xu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yifang Dang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Mengge Zheng
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yajuan Cao
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jiani Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Tao Luo
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jun Zhu
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Wei Sha
- Department of tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Haipeng Liu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| |
Collapse
|
28
|
Migliorelli A, Manuelli M, Ciorba A, Stomeo F, Pelucchi S, Bianchini C. The Role of Blood Inflammatory Markers in Salivary Gland Carcinoma: A Scoping Review. J Clin Med 2025; 14:1762. [PMID: 40095867 PMCID: PMC11901287 DOI: 10.3390/jcm14051762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: Malignant carcinomas of the salivary glands account for approximately 1 to 7% of all head and neck malignancies and approximately 0.3% of all malignant neoplasms. Recently, the scientific community has focused on finding biomarkers that could tailor the treatment for patients with this type of cancer. The neutrophil-lymphocyte ratio (NLR) was the first marker studied and it is one of the most widely used; the platelet-lymphocyte ratio (PLR), the systemic immune inflammation index (SII) and the systemic inflammatory response index (SIRI) have recently emerged as important biomarkers. The aim of this scoping review is to evaluate the role of inflammatory biomarkers in the management of salivary gland malignancies. Methods: A review of the English literature on inflammatory blood markers in major salivary gland cancer was performed using PubMed, Scopus, and Google Scholar databases. The literature review was performed using the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines for scoping reviews. Results: Eleven full-text articles were included in this review, for a total of 1356 patients in which the role of inflammatory biomarkers (NLR, PLR, SII or SIRI) for the diagnosis and prognosis of salivary gland cancer was assessed. NLR (i) was evaluated in all the studies; (ii) it contributed to the diagnosis and prognosis of both adult and pediatric patients and (iii) can be considered the main biomarker, even if a universal cut-off range is not available yet. PLR, SII and SIRI were introduced more recently and were evaluated only in some studies. Conclusions: The findings of this study suggest that elevated NLR values, regardless of age, are more frequently associated with malignancy and a poor prognosis. Further studies are necessary to evaluate the role of biomarkers other than NLR, and to identify universal and practical cut-off values.
Collapse
Affiliation(s)
| | | | | | | | | | - Chiara Bianchini
- ENT & Audiology Unit, Department of Neurosciences, University Hospital of Ferrara, 44100 Ferrara, Italy
| |
Collapse
|
29
|
Xie J, Yang Z, Li Z, Zhang T, Chen H, Chen X, Dai Z, Chen T, Hou J. Triple-positive breast cancer: navigating heterogeneity and advancing multimodal therapies for improving patient outcomes. Cancer Cell Int 2025; 25:77. [PMID: 40045297 PMCID: PMC11881339 DOI: 10.1186/s12935-025-03680-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
Triple-positive breast cancer (TPBC), a unique subtype of luminal breast cancer, is characterized by concurrent positivity for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Owing to the crosstalk between the ER and HER2 signaling pathways, the standard of care and drug resistance of this particular subtype are difficult challenges. Recent research and clinical trials have indicated a shift in the treatment paradigm for TPBC from single-target therapies to multi-pathway, multitarget strategies aiming to comprehensively modulate intricate signaling networks, thereby overcoming resistance and enhancing therapeutic outcomes. Among multiple strategies, triple-drug therapy has emerged as a promising treatment modality, demonstrating potential efficacy in patients with TPBC. Moving forward, there is a critical need to perform in-depth analyses of specific mechanisms of cancer pathogenesis and metastasis, decipher the complex interactions between different genes or proteins, and identify concrete molecular targets, thus paving the way for the development of tailored therapeutic strategies to combat TPBC effectively.
Collapse
Affiliation(s)
- Jie Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Zhihui Yang
- Zunyi Medical University, No.6 Xuefu West Road, Zunyi, 563006, Guizhou Province, China
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zhuolin Li
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Tianyu Zhang
- Urology Department, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Huan Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Xueru Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zehua Dai
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Tao Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Jing Hou
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China.
| |
Collapse
|
30
|
Köşeci T, Seyyar M, Kıdı MM, Biter S, Eser K, Kefeli U, Nayır E, Duman BB, Mete B, Demirhindi H, Çil T. Prognostic Significance of the Combined Albumin-To-Alkaline Phosphatase Ratio (AAPR) and Haemoglobin-Albumin-Lymphocyte-Platelet (HALP) Score in Patients with Metastatic Renal Cell Carcinoma Treated by Targeted Therapy: A New Prognostic Combined Risk Scoring. J Clin Med 2025; 14:1742. [PMID: 40095849 PMCID: PMC11900617 DOI: 10.3390/jcm14051742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/16/2025] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: Renal cell carcinoma (RCC) accounts for 2-3% of all cancers, with approximately 25% of patients being detected at the metastatic stage. This study aimed to determine the prognostic significance of co-evaluating two risk parameters: one, the HALP score based on haemoglobin, albumin, lymphocyte, and platelet counts, and the other, albumin-to-alkaline phosphatase ratio (AAPR) in patients with metastatic RCC treated with targeted therapy. Methods: This retrospective cohort study included 147 patients with metastatic RCC. The HALP score and AAPR values were calculated from pre-treatment blood test results, and followingly, the cut-off value was determined by using ROC analysis. Patients were categorised into three groups with a low, moderate or high combined risk score based on this cut-off value. The effect of these risk groups on survival was evaluated. Results: The mean age of patients was 64.1 ± 11.9. The median follow-up time was 38.3 months, and the mortality rate was 53.7% in all groups. Kaplan-Meier survival analysis showed a statistically significant difference between the combined scores of the risk groups: the median survival time was 51.6 months in the low-risk group, 20.7 months in the medium-risk group, and 10.4 months in the high-risk group (p < 0.001), with 5-year survival rates being 38.8% in the low-risk group, 30% in the intermediate-risk group, and 19% in the high-risk group. When compared to the low-risk group, Cox regression analysis revealed that the mortality risk, i.e., HR (hazard ratio), was 2.42 times higher in the intermediate-risk group and 3.76 times higher in the high-risk group. A nephrectomy operation decreased the mortality risk (HR = 0.24) by 4.16 times. Conclusions: This new combined risk scoring, obtained from co-evaluating the HALP score and AAPR, was found to be an independent prognostic factor in metastatic RCC patients. This combined risk scoring is expected to help clinicians in treatment decisions.
Collapse
Affiliation(s)
- Tolga Köşeci
- Department of Medical Oncology, Faculty of Medicine, Çukurova University, 01330 Adana, Türkiye; (M.M.K.); (S.B.)
| | - Mustafa Seyyar
- Department of Medical Oncology, Gaziantep City Hospital, 27470 Gaziantep, Türkiye;
| | - Mehmet Mutlu Kıdı
- Department of Medical Oncology, Faculty of Medicine, Çukurova University, 01330 Adana, Türkiye; (M.M.K.); (S.B.)
| | - Sedat Biter
- Department of Medical Oncology, Faculty of Medicine, Çukurova University, 01330 Adana, Türkiye; (M.M.K.); (S.B.)
| | - Kadir Eser
- Department of Oncology, Mersin University Hospital, 33240 Mersin, Türkiye;
| | - Umut Kefeli
- Department of Medical Oncology, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Türkiye;
| | - Erdinç Nayır
- Department of Medical Oncology, Mersin Medical Park Hospital, 33200 Mersin, Türkiye;
| | - Berna Bozkurt Duman
- Department of Medical Oncology, Adana City Training and Research Hospital, University of Health Sciences, 01370 Adana, Türkiye; (B.B.D.); (T.Ç.)
| | - Burak Mete
- Department of Public Health, Faculty of Medicine, Çukurova University, 01330 Adana, Türkiye;
| | - Hakan Demirhindi
- Department of Public Health, Faculty of Medicine, Çukurova University, 01330 Adana, Türkiye;
| | - Timuçin Çil
- Department of Medical Oncology, Adana City Training and Research Hospital, University of Health Sciences, 01370 Adana, Türkiye; (B.B.D.); (T.Ç.)
| |
Collapse
|
31
|
Wang J, Shao F, Yu QX, Ye L, Wusiman D, Wu R, Tuo Z, Wang Z, Li D, Cho WC, Wei W, Feng D. The Common Hallmarks and Interconnected Pathways of Aging, Circadian Rhythms, and Cancer: Implications for Therapeutic Strategies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0612. [PMID: 40046513 PMCID: PMC11880593 DOI: 10.34133/research.0612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/17/2025]
Abstract
The intricate relationship between cancer, circadian rhythms, and aging is increasingly recognized as a critical factor in understanding the mechanisms underlying tumorigenesis and cancer progression. Aging is a well-established primary risk factor for cancer, while disruptions in circadian rhythms are intricately associated with the tumorigenesis and progression of various tumors. Moreover, aging itself disrupts circadian rhythms, leading to physiological changes that may accelerate cancer development. Despite these connections, the specific interplay between these processes and their collective impact on cancer remains inadequately explored in the literature. In this review, we systematically explore the physiological mechanisms of circadian rhythms and their influence on cancer development. We discuss how core circadian genes impact tumor risk and prognosis, highlighting the shared hallmarks of cancer and aging such as genomic instability, cellular senescence, and chronic inflammation. Furthermore, we examine the interplay between circadian rhythms and aging, focusing on how this crosstalk contributes to tumorigenesis, tumor proliferation, and apoptosis, as well as the impact on cellular metabolism and genomic stability. By elucidating the common pathways linking aging, circadian rhythms, and cancer, this review provides new insights into the pathophysiology of cancer and identifies potential therapeutic strategies. We propose that targeting the circadian regulation of cancer hallmarks could pave the way for novel treatments, including chronotherapy and antiaging interventions, which may offer important benefits in the clinical management of cancer.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Fanglin Shao
- Department of Rehabilitation,
The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qing Xin Yu
- Department of Pathology,
Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
- Department of Pathology,
Ningbo Medical Centre Lihuili Hospital, Ningbo, Zhejiang 315040, China
| | - Luxia Ye
- Department of Public Research Platform,
Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA,
Army Medical University, Chongqing, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - William C. Cho
- Department of Clinical Oncology,
Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
- Division of Surgery and Interventional Science,
University College London, London W1W 7TS, UK
| |
Collapse
|
32
|
Xia J, Hou Y, Wang J, Zhang J, Wu J, Yu X, Cai H, Yang W, Xu Y, Mou S. Repair of Isoaspartyl Residues by PCMT1 and Kidney Fibrosis. J Am Soc Nephrol 2025:00001751-990000000-00577. [PMID: 40036072 DOI: 10.1681/asn.0000000652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
Background Kidney fibrosis, the excessive accumulation and dysregulated remodeling of the extracellular matrix, is the principal pathophysiologic process in CKD. Protein L-isoaspartyl/D-aspartyl methyltransferase (PCMT1) is crucial in repairing post-translational modifications of L-isoaspartyl residues, which are important for extracellular matrix proteins because of their low turnover rate and susceptibility to accelerating factors. This study aimed to reveal a novel role of PCMT1 in kidney fibrosis. Methods Kidney tissues from mice and humans were evaluated for PCMT1 expression and its association with fibrosis and kidney function. PCMT1's effects on the TGF-β1/Smad signaling were analyzed, and its functional role was assessed in tubule-specific Pcmt1 knockout murine models of kidney fibrosis. The ability of secreted PCMT1 to repair L-isoaspartyl residues on the ectodomain of TGF-β receptor 2 (TGFBR2) was investigated using immunoprecipitation, gene lentivirus overexpression or knockout, and post-translational modification mass spectrometry. Results PCMT1 expression was decreased in the tubules of human kidney biopsies from patients with CKD and murine fibrosis models. Renal tubule-specific PCMT1 deficiency in murine kidney fibrosis models worsened tubular injury, extracellular matrix protein deposition, myofibroblast activation, and TGF-β1/Smad signaling overactivation. Mechanistically, PCMT1 was unconventionally secreted and enzymatically inhibited TGF-β1–induced extracellular matrix protein deposition in vitro. PCMT1 interacted with TGFBR2, reversing N63 deamidation on its ectodomain, which triggered TGFBR2 ubiquitination and degradation. PCMT1 supplementation in kidneys decreased TGFBR2 levels, attenuated TGF-β1/Smad overactivation, and impeded the profibrotic process. Conclusions Our study highlights the importance of PCMT1 in maintaining extracellular matrix homeostasis and mitigating kidney fibrosis by regulating TGFBR2 deamidation and its protein stability, suppressing the TGF-β1/Smad signaling.
Collapse
Affiliation(s)
- Jia Xia
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yutong Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jie Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jiahui Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jiajia Wu
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xiang Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Hong Cai
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Wen Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Shan Mou
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
33
|
Zhang X, Liu J, Cao Y, Wang W, Lin H, Yu Y. Multi-omic and machine learning analysis of mitochondrial RNA modification genes in lung adenocarcinoma for prognostic and therapeutic implications. Transl Oncol 2025; 53:102306. [PMID: 39908965 PMCID: PMC11847145 DOI: 10.1016/j.tranon.2025.102306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related deaths, driven by complex pathogenesis and poor prognosis. Recognizing the pivotal role of mitochondrial RNA modifications (MRM) in cancer progression, this study aims to provide a comprehensive analysis of MRM-related genes and their clinical relevance in lung adenocarcinoma (LUAD). Integrating multi-omic datasets, we systematically explored the molecular features of MRM-related genes across various cancers and identified distinct expression patterns and prognostic associations. Single-cell analysis further reveals MRM-driven cell-cell interactions and pathway activation, particularly in cycling and epithelial cells. Using advanced machine learning techniques, we developed a novel prognostic signature-the Mitochondrial RNA Modification-related Signature (MRMS)-comprising nine genes: TXN, LDHA, HMGA1, SFTPB, KRT8, ALG3, S100A16, HSPD1, and ALDOA. The MRMS demonstrates superior predictive performance for LUAD survival compared to previously reported models. Our findings uniquely link MRMS to increased tumor mutational burden, genetic instability, and an immunosuppressive tumor microenvironment characterized by reduced immune cell infiltration and elevated tumor purity. Additionally, MRMS is associated with immunotherapy-related features, suggesting its potential in predicting treatment response. Experimental validation identified ALG3 as an oncogenic driver in LUAD, influencing tumor cell proliferation, migration, and invasion. In conclusion, this study establishes MRMS as a robust prognostic biomarker and highlights its dual role in shaping the tumor immune microenvironment and guiding therapeutic strategies. These findings provide novel insights into mitochondrial RNA modifications and their potential applications in personalized treatment for LUAD.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Jiatao Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Yaolin Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Haoran Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Yue Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
34
|
García-Saenz JÁ, Rodríguez-Lescure Á, Cruz J, Albanell J, Alba E, Llombart A. Second-Line Treatment Options for Patients with Metastatic Triple-Negative Breast Cancer: A Review of the Clinical Evidence. Target Oncol 2025; 20:191-213. [PMID: 39806129 PMCID: PMC11933194 DOI: 10.1007/s11523-024-01125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Metastatic triple-negative breast cancer has a poor prognosis and poses significant therapeutic challenges. Until recently, limited therapeutic options have been available for patients with advanced disease after failure of first-line chemotherapy. The aim of this review is to assess the current evidence supporting second-line treatment options in patients with metastatic triple-negative breast cancer. Evidence was reviewed from controlled clinical trials in which eribulin, vinorelbine, capecitabine, gemcitabine, gemcitabine plus carboplatin, fam-trastuzumab-deruxtecan, sacituzumab govitecan, olaparib, and talazoparib were used in the second-line treatment for metastatic breast cancer, either as study drugs or as comparators. The benefit of treatment was evaluated using the European Society for Medical Oncology-Magnitude of Clinical Benefit Scale. Based on the evidence review, sacituzumab govitecan was identified as the preferred second-line treatment option for patients with metastatic triple-negative breast cancer, supported by clinical evidence and consensus across international clinical guidelines. Olaparib and talazoparib are of use in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer and germline BRCA1/2 mutations. Exploratory data for fam-trastuzumab-deruxtecan suggest a survival benefit in human epidermal growth factor receptor 2-low, hormone-receptor-negative patients, but further solid evidence is required. Other chemotherapies with lower European Society for Medical Oncology-Magnitude of Clinical Benefit Scale scores may continue to be useful in highly selected patients.
Collapse
Affiliation(s)
- José Ángel García-Saenz
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Calle Profesor Martín Lagos, S/N, 28040, Madrid, Spain.
| | | | - Josefina Cruz
- Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Joan Albanell
- Hospital del Mar Research Institute, Barcelona, Spain
- Pompeu Fabra University, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| | - Emilio Alba
- Medical Oncology Unit, Universitary Hospital Virgen de la Victoria, CIBERONC, IBIMA, Malaga, Spain
| | | |
Collapse
|
35
|
Rahimian S, Mirkazemi K, Kamalinejad A, Doroudian M. Exosome-based advances in pancreatic cancer: The potential of mesenchymal stem cells. Crit Rev Oncol Hematol 2025; 207:104594. [PMID: 39732301 DOI: 10.1016/j.critrevonc.2024.104594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/28/2024] [Accepted: 12/08/2024] [Indexed: 12/30/2024] Open
Abstract
Pancreatic cancer, especially pancreatic ductal adenocarcinoma (PDAC), is one of the most challenging clinical conditions due to its late-stage diagnosis and poor survival rates. Mesenchymal stem cells (MSCs), used for targeted therapies, are being explored as a promising treatment because of their tumor-homing properties and potential contributions to the pancreatic cancer microenvironment. Understanding these interactions is crucial for developing effective treatments. In this study, we investigated how MSCs exhibit tropism towards tumors, influence the microenvironment through paracrine effects, and serve as potential drug delivery vehicles. We also examined their role in progression and therapeutic resistance in pancreatic cancer therapy. The cytotoxic effects of certain compounds on tumor cells, the use of genetically modified MSCs as drug carriers, and the potential of exosomal biomarkers like miRNAs and riRNAs for diagnosis and monitoring of pancreatic cancer were analyzed. Overall, MSC-based therapies, coupled with insights into tumor-stromal interactions, offer new avenues for improving outcomes in pancreatic cancer treatment. Additionally, the use of MSC-based therapies in clinical trials is discussed. While MSCs show promising potential for pancreatic cancer monitoring, diagnosis, and treatment, results so far have been limited.
Collapse
Affiliation(s)
- Sana Rahimian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Kimia Mirkazemi
- Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Armita Kamalinejad
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
36
|
Zhou J, Yang F, Zheng C, Chen Y, Chen M, Lin Q, Chang C, Cai S, Sun Z, Li H, Qin L, Chen Y. OncomiR mdv1-miR-M7-5p promotes avian lymphomatosis by modulating the BCL2/Bax mitochondrial apoptosis signaling pathway. Vet Microbiol 2025; 302:110417. [PMID: 39889336 DOI: 10.1016/j.vetmic.2025.110417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Marek's disease virus (MDV) is an oncogenic alphaherpesvirus that infects poultry and causes fatal lymphomas in infected chickens. Notably, the mdv1-miR-M7-5p, a pivotal oncomiR encoded by MDV, is closely associated with viral replication and latency. Here, mdv1-miR-M7-5p was transfected into the chicken lymphoma cell line MSB1, which resulted in the inhibition of lymphoma cell apoptosis and an increase in lymphoma cell proliferation and migration. Additionally, the expression of the tumor suppressor genes p53 and ARRDC3 were significantly downregulated, while the MDV latency-associated genes such as ICP4 and ICP27 were significantly upregulated. The BCL2/Bax ratio was increased while the expression of genes involved in the apoptotic signaling pathway were decreased. Furthermore, our mitochondrial function experiments in MSB1 cells demonstrated that mdv1-miR-M7-5p enhanced mitochondrial ATP release and altered the mitochondrial membrane potential, thereby affecting mitochondrial function and inhibiting lymphoma cell apoptosis. Dual-luciferase assays revealed that mdv1-miR-M7-5p binds to caspase-6. For the in vivo study, a cholesterol-modified inhibitor of mdv1-miR-M7-5p was administered to chickens. Inhibition of mdv1-miR-M7-5p resulted in a lower mortality rate than that in the control groups. Furthermore, the expression levels of the cytokines interferon-gamma (IFN-γ), interleukin (IL)-4, and IL-17 in the plasma of MDV-infected chickens were significantly increased. A marked increase was observed in apoptosis in the spleen tissues, and the expression of apoptosis-related genes including caspase-3 and tumor suppressor gene PTEN in immune organs, including the spleen, bursa of Fabricius, and thymus, were markedly upregulated. In summary, the oncogenic miRNA mdv1-miR-M7-5p promotes MDV latency and may facilitate lymphoma formation by mediating the BCL2/CytC signaling pathway. This mediation enhances mitochondrial function and inhibits lymphoma cell apoptosis, thereby contributing to lymphoma development.
Collapse
Affiliation(s)
- Jun Zhou
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Fan Yang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Congsen Zheng
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Yanting Chen
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Meiting Chen
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Qiaoer Lin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Chuanzhe Chang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Shikai Cai
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Zhaoyang Sun
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Limei Qin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China.
| | - Yanfeng Chen
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China.
| |
Collapse
|
37
|
Hushmandi K, Alimohammadi M, Heiat M, Hashemi M, Nabavi N, Tabari T, Raei M, Aref AR, Farahani N, Daneshi S, Taheriazam A. Targeting Wnt signaling in cancer drug resistance: Insights from pre-clinical and clinical research. Pathol Res Pract 2025; 267:155837. [PMID: 39954370 DOI: 10.1016/j.prp.2025.155837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/22/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Cancer drug resistance, encompassing both acquired and intrinsic chemoresistance, remains a significant challenge in the clinical management of tumors. While advancements in drug discovery and the development of various small molecules and anti-cancer compounds have improved patient responses to chemotherapy, the frequent and prolonged use of these drugs continues to pose a high risk of developing chemoresistance. Therefore, understanding the primary mechanisms underlying drug resistance is crucial. Wnt proteins, as secreted signaling molecules, play a pivotal role in transmitting signals from the cell surface to the nucleus. Aberrant expression of Wnt proteins has been observed in a variety of solid and hematological tumors, where they contribute to key processes such as proliferation, metastasis, stemness, and immune evasion, often acting in an oncogenic manner. Notably, the role of the Wnt signaling pathway in modulating chemotherapy response in human cancers has garnered significant attention. This review focuses on the involvement of Wnt signaling and its related molecular pathways in drug resistance, highlighting their associations with cancer hallmarks, stemness, and tumorigenesis linked to chemoresistance. Additionally, the overexpression of Wnt proteins has been shown to accelerate cancer drug resistance, with regulation mediated by non-coding RNAs. Elevated Wnt activity reduces cell death in cancers, particularly by affecting mechanisms like apoptosis, autophagy, and ferroptosis. Furthermore, pharmacological compounds and small molecules have demonstrated the potential to modulate Wnt signaling in cancer therapy. Given its impact, Wnt expression can also serve as a prognostic marker and a factor influencing survival outcomes in human cancers.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Teimour Tabari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
38
|
Marshall M, Lajara S, Quiroga-Garza G, Korentzelos D, Mohebnasab M, Khader S. Primary tumor characteristics and immunohistochemical profile of renal cell carcinoma in serous fluid cytology. J Am Soc Cytopathol 2025; 14:123-131. [PMID: 39690083 DOI: 10.1016/j.jasc.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) involves serosal surfaces in 2%-3% of cases, and thus few papers describe serous fluid cytology (SFC) involvement by RCC. This diagnosis is challenging, given its rarity, nondescript cytomorphologic features and infrequent expression of widely used epithelial markers MOC31 and BerEP4. We describe our institutional experience with RCC in SFC specimens. MATERIALS AND METHODS Our institutional laboratory information system was queried for SFC specimens from patients with RCC between 2013 and 2023. Cases signed out as "Suspicious for Malignant Cells" and "Positive for Malignant Cells" were included. Patient demographics, immunohistochemical results, primary tumor characteristics, and molecular data were recorded. RESULTS Sixty-one cases, 50 pleural, and 11 peritoneal fluid specimens were identified. Fifty (50) were signed out as positive for malignancy and 11 were signed out as suspicious for malignancy. MOC31 and BerEP4 were positive in 59% and 55% of cases, respectively. PAX-8, CA9, CD10, and RCC were positive in 85%, 82%, 73%, and 29% of cases, respectively. Primary tumor histologic subtypes included 39 clear cell, 6 papillary, 1 chromophobe, and 15 were not further subclassified. Fifty-nine percent (59%) of cases had a nuclear grade of 4%, and 37% had sarcomatoid or rhabdoid differentiation. Seventy-one percent (71%) of cases had stage 3 or 4 disease. CONCLUSIONS RCC metastases to serosal surfaces are more likely to be seen in patients with higher disease stage, high nuclear grade, and sarcomatoid or rhabdoid differentiation. MOC31 and BerEP4 performed poorly in this setting. We recommend the addition of cytokeratins, PAX-8, CD10, and CA-9 to confirm metastatic involvement.
Collapse
Affiliation(s)
- Mason Marshall
- Cytopathology Center of Excellence, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| | - Sigfred Lajara
- Cytopathology Center of Excellence, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Gabriela Quiroga-Garza
- Genitourinary Pathology Center of Excellence, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Dimitrios Korentzelos
- Genitourinary Pathology Center of Excellence, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Maedeh Mohebnasab
- Molecular and Genomic Pathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Samer Khader
- Cytopathology Center of Excellence, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
39
|
Zhu JM, Chen SH, Xu YC, Gao RC, Cai H, Zheng QS, Sun XL, Xue XY, Wei Y, Xu N. ALB inhibits tumor cell proliferation and invasion by regulating immune microenvironment and endoplasmic reticulum stress in clear cell renal cell carcinoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167672. [PMID: 39862995 DOI: 10.1016/j.bbadis.2025.167672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE The aim of this work is to identify putative hub genes for the advancement of clear cell renal cell carcinoma (ccRCC) and determine the fundamental mechanisms. METHODS We employed multiple bioinformatics techniques to screen hub genes. Key hub gene expression levels in ccRCC were assessed. A plethora of functional experiments were carried out to explore the biological role of hub gene. Based on genome-wide association studies, a Mendelian randomization research was conducted to ascertain the causative relationship between albumin (ALB) and ccRCC. RESULTS ALB was low expression in ccRCC tissues and cell lines. It was an independent predictor of progression-free survival following treatment and the overall survival of ccRCC patients. ALB overexpression exhibited the reverse effects of ALB knockdown, which increased cell proliferation, migration, and invasion while inhibiting cell death. Similarly, ALB overexpression inhibited the growth of ccRCC tumors in vivo. Consistent with functional enrichment analysis, ALB overexpression activates the endoplasmic reticulum stress (ERS) in vitro and vivo. The Mendelian randomization showed ALB was associated with the risk of ccRCC. Additionally, ALB was causally associated with γδT cells infiltrates in ccRCC. CONCLUSION ALB plays an important effect in ccRCC via activation of the ERS and regulating immune microenvironment.
Collapse
Affiliation(s)
- Jun-Ming Zhu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Shao-Hao Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yi-Cheng Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Rui-Cheng Gao
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Hai Cai
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xiong-Lin Sun
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.
| |
Collapse
|
40
|
Jiang M, Zhang C. Higher dietary vegetable and fruit intake along with their biomarkers is inversely associated with all-cause mortality among cancer survivors. Nutr Res 2025; 135:141-157. [PMID: 40056790 DOI: 10.1016/j.nutres.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/08/2025] [Accepted: 02/08/2025] [Indexed: 03/10/2025]
Abstract
The association between dietary vegetable and fruit (VF) intake and survival outcomes among cancer survivors remains unclear, with limited research available on the association between VF-related biomarkers and mortality in this population. We hypothesized that there were inverse relationships between dietary VF intake, serum carotenoids, serum vitamin C, composite biomarker score and both all-cause and cancer-specific mortality among cancer survivors. This study analyzed cancer survivors from the National Health and Nutrition Examination Survey (NHANES) 1999 to 2018 cycles for VF intake (n = 4326), and from NHANES III, 2003 to 2006, and 2017 to 2018 cycles for serum carotenoids (n = 2187), serum vitamin C (n = 2267), and composite biomarker score (n = 2131). Weighted multivariable Cox proportional hazards regression models were used to assess the associations between exposures and mortality, computing hazard ratio (HR) and 95% CI. Dietary VF intake was significantly associated with a lower risk of all-cause mortality (HR = 0.80, 95% CI: 0.67-0.96) over a median follow-up of 6.9 years. For serum biomarkers, higher concentration of total carotenoids (median follow-up: 10.0 years), vitamin C (median follow-up: 9.4 years) and higher composite biomarker score (median follow-up: 10.1 years) were associated with lower risks of all-cause mortality, with HRs (95% CIs) of 0.73 (0.58-0.92), 0.73 (0.56-0.95), and 0.73 (0.57-0.95), respectively. Additionally, only vitamin C was linked to cancer-specific mortality, with HRs (95% CIs) of 0.55 (0.37-0.81). Higher VF intake and their associated biomarkers are associated with a lower risk of all-cause mortality among U.S. cancer survivors.
Collapse
Affiliation(s)
- Mianmian Jiang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, PR China
| | - Caixia Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
41
|
Vitale E, Maistrello L, Rizzo A, Brunetti O, Massafra R, Mollica V, Massari F, Santoni M. Nutritional conditions and PFS and OS in cancer immunotherapy: the MOUSEION-010 meta-analysis. Immunotherapy 2025; 17:269-281. [PMID: 40134096 PMCID: PMC12013447 DOI: 10.1080/1750743x.2025.2483656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The MOUSEION-010 Meta-Analysis assessed the association between nutritional status and clinical outcomes such as Progression Free Survival (PFS) and Overall Survival (OS) among cancer patients treated with immune checkpoint inhibitors (ICIs). METHODS Nutritional status was assessed based on the Prognostic Nutrition Index (PNI), Geriatric Nutritional Risk Index (GNRI) and Controlling Nutritional Status (CONUT) indexes. Databases consulted were: Embase, PubMed, Scopus and Web of Science. RESULTS PNI and GNRI indexes did not show a significant association with both PFS and OS, while CONUT index displayed a significant difference in PFS between the two groups, in favor of the control group (Z = 4.04; p < 0.01) also without any publication bias (β= -1.27; 95% CI = [-2.13; -0.42]; p = 0.10]). The same trend was recorded in OS, too (Z = 4.24; p < 0.01). However, publication bias was present (β = 1.89; 95% CI = [1.26; 2.54]; p = 0.028]) and the numerosity of the studies did not reveal the sufficient statistical power to obtain reliable results. CONCLUSION Malnutrition could negatively impact cancer patients, especially in advanced phases. Our findings could be associated with the reduction of physical ability and daily activity performance, lower compliance with treatment protocols, and shorter survival outcomes.
Collapse
Affiliation(s)
- Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | | | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | |
Collapse
|
42
|
Song J, Liu T, Huang Q, Lv Y, Wen Y, Wang R, Bie J. Prognostic value of prognostic nutritional index in patients with nasopharyngeal carcinoma treated with endostar and concurrent chemoradiotherapy. Support Care Cancer 2025; 33:226. [PMID: 40011250 DOI: 10.1007/s00520-025-09280-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
PURPOSE This study aimed to evaluate the prognostic value of the pre-treatment prognostic nutritional index (PNI) in patients with locally advanced nasopharyngeal carcinoma (LANPC) treated with endostar combined with concurrent chemoradiotherapy (ECCRT). METHODS Clinical data from 92 patients with LANPC who underwent ECCRT between May 2015 and December 2020 were retrospectively analyzed. The PNI was calculated using peripheral blood samples taken 1 week before treatment. The optimal cut-off value for PNI was determined via receiver operating characteristic (ROC) curve analysis based on overall survival (OS). Patients were categorized into high PNI and low PNI groups. The Kaplan-Meier method assessed the impact of PNI on survival, while univariate and multivariate Cox regression analyses identified independent risk factors affecting patient survival. RESULTS The optimal cut-off value of PNI was 50.05. The 3-year OS, progression-free survival (PFS), distant metastasis-free survival (DMFS), and local recurrence-free survival (LRRFS) rates were 91.07% vs. 75.00% (P = 0.002), 83.93% vs. 66.67% (P = 0.015), 89.29% vs. 69.44% (P = 0.004), and 94.64% vs. 91.67% (P = 0.668) in the high PNI and low PNI groups, respectively. A low PNI was associated with shorter OS (HR = 3.592, P = 0.004), PFS (HR = 2.890, P = 0.017), and DMFS (HR = 3.826, P = 0.008). Multivariate analysis revealed that PNI was an independent prognostic factor for OS, PFS, and DMFS. CONCLUSIONS The PNI may serve as a valuable prognostic predictor for patients with LANPC receiving ECCRT, aiding clinicians in selectively providing multimodal interventions to optimize survival outcomes.
Collapse
Affiliation(s)
- JunMei Song
- Department of Oncology, Beijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), 637000, Nanchong, Sichuan, China
- The Second Clinical Medical College of North, Sichuan Medical College, 637000, Nanchong, Sichuan, China
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Ting Liu
- Second Division of Department of Radiation Oncology, Guangxi Academy of Medical, Sciences & the People's Hospital of Guangxi Zhuang Autonomous Region, 530021, Nanning, Guangxi, China
| | - Qiulin Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - YuQing Lv
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - YaJing Wen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - RenSheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China.
| | - Jun Bie
- Department of Oncology, Beijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), 637000, Nanchong, Sichuan, China.
- The Second Clinical Medical College of North, Sichuan Medical College, 637000, Nanchong, Sichuan, China.
| |
Collapse
|
43
|
Önder T, Öner İ, Karaçin C, Ateş Ö. Valuable predictive power of prognostic nutritional index in metastatic breast cancer patients treated with CDK4/6 inhibitors. Jpn J Clin Oncol 2025:hyaf036. [PMID: 39997162 DOI: 10.1093/jjco/hyaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
AIMS AND OBJECTIVES The prognostic value of nutritional status in HR+/HER2- metastatic breast cancer (mBC) patients treated with CDK4/6 inhibitors (CDK4/6is) and endocrine therapy (ET) is unclear. METHODS/MATERIALS The effect of PNI values before starting CDK 4/6i on patient prognosis was retrospectively analyzed. RESULTS A total of 431 patients were evaluated. After 35.7 months of follow-up, the median overall survival (mOS) was 46.3 months (95% CI, 29.7-62.8). The PNI-low group had decreased progression-free survival compared to the PNI-high group [16.6 vs. 30.5 months; univariate HR = 1.640, 95% confidence interval (CI): 1.281-2.099, P < .001]. The PNI-low group's mOS was noticeably shorter than the PNI-high group (35.0 months vs. not reached; multivariate-adjusted HR: 2.082, 95% CI: 1.398-3.102, P < .001). When stratified by CDK4/6i line: In patients using CDK4/6i as the first line, mPFS for the PNI-low and PNI-high group was 24.6 vs. 35.6 months (P = .026), and survival probabilities at 24, 36, and 48 months in the PNI-low and PNI-high groups were 75%, 62%, 57%, and 88%, 80%, and 72%, respectively (P = .002). In patients using CDK4/6i as the second line and after, mPFS was 8.2 vs.12.0 months (P = .014), and mOS was 18.6 vs. 39.6 months (P = .001) for the PNI-low and PNI-high group, respectively. The ORR and DCR were significantly lower in the low-PNI group than in the high-PNI group (P = .018 and P = .017, respectively). The incidence of grade 3-4 side effects due to CDK4/6is (39.8% vs. 30.7%, P = .046) was significantly greater in the PNI-low group than in the PNI-high group. CONCLUSIONS This study's results suggest that PNI is an easily measured and reliable indicator of prognosis in mBC patients treated with CDK4/6i and ET.
Collapse
Affiliation(s)
- Tuğba Önder
- Health Sciences University, Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Mehmet Akif Ersoy Neighborhood, 13th Street No: 56 Demetevler Yenimahalle, Ankara 06200, Türkiye
| | - İrem Öner
- Health Sciences University, Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Mehmet Akif Ersoy Neighborhood, 13th Street No: 56 Demetevler Yenimahalle, Ankara 06200, Türkiye
| | - Cengiz Karaçin
- Health Sciences University, Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Mehmet Akif Ersoy Neighborhood, 13th Street No: 56 Demetevler Yenimahalle, Ankara 06200, Türkiye
| | - Öztürk Ateş
- Health Sciences University, Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Mehmet Akif Ersoy Neighborhood, 13th Street No: 56 Demetevler Yenimahalle, Ankara 06200, Türkiye
| |
Collapse
|
44
|
de Souza MM, Gini ALR, Moura JA, Scarim CB, Chin CM, dos Santos JL. Prodrug Approach as a Strategy to Enhance Drug Permeability. Pharmaceuticals (Basel) 2025; 18:297. [PMID: 40143076 PMCID: PMC11946379 DOI: 10.3390/ph18030297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025] Open
Abstract
Absorption and permeability are critical physicochemical parameters that must be balanced to achieve optimal drug uptake. These key factors are closely linked to the maximum absorbable dose required to provide appropriate plasma levels of drugs. Among the various strategies employed to enhance drug solubility and permeability, prodrug design stands out as a highly effective and versatile approach for improving physicochemical properties and enabling the optimization of biopharmaceutical and pharmacokinetic parameters while mitigating adverse effects. Prodrugs are compounds with reduced or no activity that, through bio-reversible chemical or enzymatic processes, release an active parental drug. The application of this technology has led to significant advancements in drug optimization during the design phase, and it offers broad potential for further development. Notably, approximately 13% of the drugs approved by the U.S. Food and Drug Administration (FDA) between 2012 and 2022 were prodrugs. In this review article, we will explore the application of prodrug strategies to enhance permeability, describing examples of market drugs. We also describe the use of the prodrug approach to optimize PROteolysis TArgeting Chimeras (PROTACs) permeability by using conjugation technologies. We will highlight some new technologies in prodrugs to enrich permeability properties, contributing to developing new effective and safe prodrugs.
Collapse
Affiliation(s)
- Mateus Mello de Souza
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Ana Luísa Rodriguez Gini
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Jhonnathan Alves Moura
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| | - Cauê Benito Scarim
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Chung Man Chin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Union of the Colleges of the Great Lakes (UNILAGO), School of Medicine, Advanced Research Center in Medicine (CEPAM), Sao Jose do Rio Preto 15030-070, SP, Brazil
| | - Jean Leandro dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| |
Collapse
|
45
|
Tan Q, Xiang C, Zhang H, Yuan Y, Gong S, Zheng Z, Wang X, Liu X, Chen Y, Tan C. YAP promotes fibrosis by regulating macrophage to myofibroblast transdifferentiation and M2 polarization in chronic pancreatitis. Int Immunopharmacol 2025; 148:114087. [PMID: 39818090 DOI: 10.1016/j.intimp.2025.114087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
Chronic pancreatitis (CP) is a clinical entity characterized by progressive inflammation and irreversible fibrosis of the pancreas, which ultimately leads to exocrine and/or endocrine insufficiency as well as an increased risk of pancreatic cancer. Currently, there are no specific or effective approved therapies for CP. Herein, we show that macrophage to myofibroblast transdifferentiation (MMT) and M2 macrophage polarization are associated with both human CP and CP experimental mouse models. In addition, we show YAP is activated in macrophages during CP. Furthermore, we used the YAP agonist XMU-MP-1 (XMU) and the YAP inhibitor Verteporfin (VP) to modulate YAP expression levels. In vitro experiments revealed that XMU upregulated YAP expression, thereby promoting MMT and enhancing M2 macrophage polarization; conversely, VP downregulated YAP expression, inhibiting these effects. In vivo studies indicated that XMU exacerbated acinar cell atrophy and interstitial fibrosis in caerulein-induced mouse models of CP, while VP mitigated these adverse effects associated with CP. These findings provide new insights into the pathogenic mechanisms underlying CP, and offer potential therapeutic targets for CP.
Collapse
Affiliation(s)
- Qingquan Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China
| | - Chengzhi Xiang
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China
| | - Haoqi Zhang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China
| | - Yuan Yuan
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland
| | - Songlin Gong
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland
| | - Zhenjiang Zheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China
| | - Xing Wang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China
| | - Xubao Liu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China
| | - Yonghua Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China.
| | - Chunlu Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan CN 610041, China.
| |
Collapse
|
46
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
47
|
Hunter NB, Peterson LM, Specht JM, Mankoff DA, Muzi M, Chen DL, Gwin WR, Vinayak S, Davidson NE, Linden HM. Fluoroestradiol (FES) and Fluorodeoxyglucose (FDG) PET imaging in patients with ER+, HER2-positive or HER2-negative metastatic breast cancer. Breast Cancer Res 2025; 27:23. [PMID: 39962532 PMCID: PMC11834562 DOI: 10.1186/s13058-025-01975-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND 18F-Fluorodeoxyglucose (FDG) and 18F-Fluorestradiol (FES) have been FDA approved for measuring tumor glycolytic activity and estrogen receptor (ER) uptake, respectively, in clinical positron emission tomography (PET) imaging for patients with hormone-receptor (HR) positive metastatic breast cancer (MBC), but little is known about its utility in patients with breast tumors that overexpress human epidermal growth factor 2 (HER2). We hypothesize that comparing patterns of FDG and FES uptake in patients with HER2-positive versus HER2-negative MBC can guide further biologic and clinical studies into the HR/HER2-positive phenotype. METHODS We conducted a retrospective study examining uptake in matched lesions for FES and FDG-PET scans, assessing these parameters in 213 patients with ER-positive/HER2-positive (n = 33) versus ER-positive/HER2-negative MBC (n = 180). We employed log-rank and t-tests to assess the association of HER2 status with outcome variables and the hypotheses that patients expressing HER2-positive disease lived longer than patient with HER2-negative disease. RESULTS No difference in FES or FDG avidity was observed between patients with HER2-negative or HER2-positive tumor status. Limited data also suggests that patients with HER2-positive disease had better overall survival (p = 0.024), than those with HER2-negative disease, but not time-to-progression between the same patient cohorts. CONCLUSION This retrospective analysis suggests that there is a possible role for future trials using FES-PET in helping to select patients with ER+/HER2-positive primary tumors who retain ER expression at all sites of disease and may benefit from endocrine therapy.
Collapse
Affiliation(s)
- Natasha B Hunter
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lanell M Peterson
- University of Washington, Seattle, WA, USA.
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Jennifer M Specht
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Mark Muzi
- University of Washington, Seattle, WA, USA
| | - Delphine L Chen
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - William R Gwin
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shaveta Vinayak
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nancy E Davidson
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hannah M Linden
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
48
|
Ge W, Gao M, Dai Y, Zheng G, Yang L, Zuo W, Tian X. Comprehensive network pharmacology and experimental study to investigate the effect and mechanism of solasonine on breast carcinoma treatment. Cancer Cell Int 2025; 25:49. [PMID: 39962568 PMCID: PMC11834262 DOI: 10.1186/s12935-025-03665-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/26/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Ferroptosis is a therapeutic strategy for breast carcinoma (BC). Solasonine (SS) was linked to ferroptosis as a tumor suppressor. However, whether SS could treat BC by activating ferroptosis and its underlying mechanisms has not been reported. METHODS We obtained the intersection of genes targeting SS and BC disease through network pharmacology. Bioinformatics analysis revealed that the intersection genes were primarily enriched in the extracellular signal-regulated kinase 2/mitogen-activated protein kinase (ERK2/MAPK) signaling pathway. The interaction modes of SS with ERK2 and epidermal growth factor receptor (EGFR) were simulated by molecular docking. We further detected the expressions of ERK2 and p-ERK2 in BC patients and the correlation between ERK2/p-ERK2 and ferroptosis. The effects and mechanism of SS on ferroptosis in BC were validated by mutation plasmids construction, immunohistology, wound healing, transwell assay, and western blotting using in vitro and in vivo models. RESULTS ERK2 and p-ERK2 were up-regulated in BC patients, and the ERK2/p-ERK2 ratio was negatively correlated with ferroptosis. Molecular docking indicated that SS could bind to ERK2 and EGFR to inhibit the activity of the ERK2/MAPK pathway. In vitro and in vivo experiments confirmed that SS induced ferroptosis by inhibiting the ERK2/MAPK pathway, inhibiting proliferation, migration, and invasion of BC cells. CONCLUSION SS could inactivate the ERK2/MAPK pathway, thereby inducing ferroptosis and further inhibiting BC cell proliferation, migration, and invasion. This study clarified the potential mechanism of SS in BC and provided a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Wenkai Ge
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Min Gao
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yingqi Dai
- Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Gang Zheng
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Li Yang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wenshu Zuo
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xingsong Tian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
49
|
Topkan E, Somay E, Selek U. Comment on "Prevalence and Association of Sarcopenia with Mortality in Patients with Head and Neck Cancer: A Systematic Review and Meta-analysis". Ann Surg Oncol 2025:10.1245/s10434-025-17023-3. [PMID: 39953351 DOI: 10.1245/s10434-025-17023-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Affiliation(s)
- Erkan Topkan
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana, Turkey
| | - Efsun Somay
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Baskent University, Ankara, Turkey.
| | - Uğur Selek
- Department of Radiation Oncology, Faculty of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
50
|
Láinez Ramos-Bossini AJ, Gámez Martínez A, Luengo Gómez D, Valverde-López F, Morillo Gil AJ, González Flores E, Salmerón Ruiz Á, Jiménez Gutiérrez PM, Melguizo C, Prados J. Computed Tomography-Based Sarcopenia and Pancreatic Cancer Survival-A Comprehensive Meta-Analysis Exploring the Influence of Definition Criteria, Prevalence, and Treatment Intention. Cancers (Basel) 2025; 17:607. [PMID: 40002202 PMCID: PMC11853262 DOI: 10.3390/cancers17040607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/16/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Sarcopenia has been associated with poor outcomes in pancreatic cancer (PC). However, published results are heterogeneous in terms of study design, oncological outcomes, and sarcopenia measurements. This meta-analysis aims to evaluate the impact of computed tomography (CT)-based sarcopenia on overall survival (OS) and progression-free survival (PFS) in patients with PC, considering potential confounders such as the CT-based method and thresholds used to define sarcopenia, as well as treatment intention. Methods: We systematically searched databases for observational studies reporting hazard ratios (HRs) for OS and PFS in PC patients stratified by CT-based sarcopenia status. Random-effects models were used to calculate pooled crude and adjusted HRs (cHRs and aHRs, respectively), with subgroup analyses based on sarcopenia measurement methods, cutoff values, sarcopenia prevalence, and treatment intention. Heterogeneity was assessed using the I2 and τ2 statistics, and publication bias was evaluated using funnel plots and Egger's test. Results: Data from 48 studies were included. Sarcopenia was significantly associated with worse OS (pooled cHR = 1.58, 95% CI: 1.38-1.82; pooled aHR = 1.39, 95% CI: 1.16-1.66) and worse PFS (pooled cHR = 1.55, 95% CI: 1.29-1.86; pooled aHR = 1.31, 95% CI: 1.11-1.55). Subgroup analyses revealed significantly different, stronger associations in studies using stricter sarcopenia cutoffs (<50 cm2/m2 for males) and in patients undergoing curative treatments. Heterogeneity was substantial across analyses (I2 > 67%), but with generally low τ2 values (0.01-0.25). Egger's test indicated potential publication bias for OS (p < 0.001), but no significant bias was observed for PFS (p = 0.576). Conclusions: Sarcopenia determined by CT is an independent predictor of poor OS and PFS in PC, but this association varies depending on the cutoff used for its definition as well as on the treatment intention. Therefore, its routine assessment in clinical practice could provide valuable prognostic information, but future research should focus on standardizing sarcopenia assessment methods.
Collapse
Affiliation(s)
- Antonio Jesús Láinez Ramos-Bossini
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
| | - Antonio Gámez Martínez
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
| | - David Luengo Gómez
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Francisco Valverde-López
- Department of Gastroenterology and Hepatology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain;
| | - Antonio Jesús Morillo Gil
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
| | | | - Ángela Salmerón Ruiz
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | | | - Consolación Melguizo
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - José Prados
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| |
Collapse
|