1
|
Guo Y, Lian J, Chen Y, Quan L, Guo X, Zhang J, Liu Z, Liu A. Factors affecting refractoriness or recurrence in diffuse large B-cell lymphoma: development and validation of a novel predictive nomogram. Hematology 2025; 30:2445395. [PMID: 39722597 DOI: 10.1080/16078454.2024.2445395] [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: 12/10/2023] [Accepted: 12/08/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Relapsed/Refractory (R/R) diffuse large B-cell lymphoma (DLBCL) represents a subgroup with a high incidence and dismal prognosis. Currently, there is a lack of robust models for predicting R/R DLBCL. Therefore, we conducted a retrospective study to identify key determinants to be incorporated into a novel nomogram to enhance the identification of DLBCL patients at elevated risk of refractoriness/recurrence. METHODS We included 293 newly-diagnosed DLBCL patients from Harbin Medical University Cancer Hospital, collected from 2008-2017. Patients were randomly divided into a training cohort (n = 206) and a validation cohort (n = 87) at a 7:3 ratio. The training cohort underwent univariable analysis to select variables for a binary logistic regression model. These variables were also prioritized using a random forest algorithm. The developed nomogram was evaluated with the receiver-operator characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA) for its clinical utility. RESULTS Univariable analysis pinpointed several factors significantly associated with refractoriness/recurrence, including pathological subtype, lactate dehydrogenase (LDH), International Prognostic Index (IPI), treatment, absolute lymphocyte count (ALC), lymphocyte/monocyte ratio (LMR), and prognostic nutritional index (PNI). Binary logistic regression highlighted pathological subtype, LDH, treatment, and ALC as key predictors, which were incorporated into the nomogram. The nomogram showed excellent calibration and accuracy in both cohorts, and comparative DCA and ROC analysis demonstrated its superior net benefit and area under the curve (AUC) compared to traditional indexes like IPI, R-IPI, and NCCN-IPI. CONCLUSION This nomogram serves as a valuable tool for predicting the likelihood of refractoriness or recurrence in DLBCL patients.
Collapse
Affiliation(s)
- Yiwei Guo
- Hematology Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Jie Lian
- Outpatient Chemotherapy Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Yao Chen
- Hematology Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Lina Quan
- Hematology Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Xiuchen Guo
- Hematology Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Jingbo Zhang
- Hematology Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Zhiqiang Liu
- Hematology Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Aichun Liu
- Hematology Department, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| |
Collapse
|
2
|
Yanes-Díaz J, Palao-Suay R, Camacho-Castañeda FI, Riestra-Ayora J, Aguilar MR, Sanz-Fernández R, Sánchez-Rodríguez C. In vivo antitumor activity of PHT-427 inhibitor-loaded polymeric nanoparticles in head and neck squamous cell carcinoma. Drug Deliv 2025; 32:2449376. [PMID: 39789884 PMCID: PMC11727052 DOI: 10.1080/10717544.2024.2449376] [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: 07/01/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Recent studies on head and neck squamous cell carcinoma (HNSCC) tumorigenesis have revealed several dysregulated molecular pathways. The phosphatidylinositol-3-kinase (PI3K) signaling pathway is frequently activated in HNSCC, making it an attractive target for therapies. PHT-427 is a dual inhibitor of PI3K and the mammalian target of AKT/PDK1. This study evaluates the anticancer efficacy of the inhibitor PHT-427 loaded into polymeric nanoparticles (NP) based on α-TOS (NP-427) administered by intratumoral injection into a hypopharyngeal squamous cell carcinoma (FaDu cells) heterotopic xenograft mouse model. The nanocarrier system, based on block copolymers of N-vinylpyrrolidone (VP) and a methacrylic derivative of α-TOS (MTOS), was synthesized, and PHT-427 was loaded into the delivery system. First, we evaluated the effect of NP-427 on tumor growth by measuring tumor volume, mouse weight, survival, and the development of tumor ulceration and necrosis. In addition, we measured PI3KCA/AKT/PDK1 gene expression, PI3KCA/AKT/PDK1 protein levels, Epidermal Growth Factor Receptor (EGFR), and angiogenesis in the tumor tissue. PHT-427 encapsulation increased drug efficacy and safety, as demonstrated by decreased tumor volume, reduced PI3K/AKT/PDK1 pathway expression, and improved antitumor activity and necrosis induction in the mouse xenograft model. EGFR and angiogenesis marker (Factor VIII) expression were significantly lower in the NP-427 group compared to other experimental groups. Administration of encapsulated PHT-427 at the tumor sites proves promising for HNSCC therapy.
Collapse
Affiliation(s)
- Joaquín Yanes-Díaz
- Otolaryngology Department, Hospital Universitario de Getafe, Madrid, Spain
| | - Raquel Palao-Suay
- Department of Polymeric Nanomaterials and Biomaterials Institute of Polymer Science and Technology, ICTP-CSIC, Madrid, Spain
- CIBER-BBN, Networking Biomedical Research Centre in Bioengineering Biomaterials, and Nanomedicine, Madrid, Spain
| | - Francisca Inmaculada Camacho-Castañeda
- Pathology Department, Hospital Universitario de Getafe, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Juan Riestra-Ayora
- Otolaryngology Department, Hospital Universitario de Getafe, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - María Rosa Aguilar
- Department of Polymeric Nanomaterials and Biomaterials Institute of Polymer Science and Technology, ICTP-CSIC, Madrid, Spain
- CIBER-BBN, Networking Biomedical Research Centre in Bioengineering Biomaterials, and Nanomedicine, Madrid, Spain
| | - Ricardo Sanz-Fernández
- Otolaryngology Department, Hospital Universitario de Getafe, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Carolina Sánchez-Rodríguez
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|
3
|
Zhou T, Luo Y, Xiong W, Meng Z, Yu NX, Zhang J. Latent profiles of problem-solving skills and their association with depressive symptoms in parents of children with cancer: A cross-sectional study. Asia Pac J Oncol Nurs 2025; 12:100633. [PMID: 39759502 PMCID: PMC11699806 DOI: 10.1016/j.apjon.2024.100633] [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: 09/26/2024] [Accepted: 11/27/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE Depressive symptoms are prevalent among parents of children with cancer, significantly impacting their well-being. Problem-solving skills, strongly linked to depressive symptoms, offer a promising avenue for intervention. This study aimed to identify latent profiles of parental problem-solving skills and evaluate differences in depressive symptoms across these profiles. METHODS A cross-sectional survey was conducted with 318 parents of children with cancer in mainland China. Self-reported data on demographics, problem-solving skills, and depressive symptoms were collected. Latent profile analysis was used to classify parental problem-solving skills into distinct profiles, and multiple logistic regression identified predictors of profile membership. RESULTS Three profiles of problem-solving skills were identified: (1) problem-oriented and constructive (n = 94, 29.6%), (2) impulsivity-oriented and irrational (n = 76, 23.9%), and (3) emotion-oriented and avoidant (n = 148, 46.5%). Parents with higher education, greater income, and urban residency were more likely to belong to the problem-oriented group. Fathers predominated in the impulsivity-oriented group, while mothers were more represented in the emotion-oriented group. Significant differences in depressive symptoms were observed across profiles, with the problem-oriented group reporting the lowest levels. CONCLUSIONS This study highlights the heterogeneity of problem-solving skills among parents of children with cancer and underscores the need for tailored interventions. Addressing specific characteristics of each profile can improve parental well-being and provide targeted support for this vulnerable population. TRIAL REGISTRATION ChiCTR2300071828.
Collapse
Affiliation(s)
- Tianji Zhou
- Xiangya School of Nursing, Central South University, Changsha, Hunan, China
- Department of Social and Behavioural Sciences, City University of Hong Kong, Hong Kong, China
| | - Yuanhui Luo
- Xiangya School of Nursing, Central South University, Changsha, Hunan, China
| | - Wenjin Xiong
- Xiangya School of Nursing, Central South University, Changsha, Hunan, China
| | - Zhenyu Meng
- Xiangya School of Nursing, Central South University, Changsha, Hunan, China
| | - Nancy Xiaonan Yu
- Department of Social and Behavioural Sciences, City University of Hong Kong, Hong Kong, China
| | - Jingping Zhang
- Xiangya School of Nursing, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Wysocki O, Mak S, Frost H, Graham DM, Landers D, Aslam T. Translating the machine; An assessment of clinician understanding of ophthalmological artificial intelligence outputs. Int J Med Inform 2025; 201:105958. [PMID: 40349525 DOI: 10.1016/j.ijmedinf.2025.105958] [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: 03/27/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Advances in artificial intelligence offer the promise of automated analysis of optical coherence tomography (OCT) scans to detect ocular complications from anticancer drug therapy. To explore how such AI outputs are interpreted in clinical settings, we conducted a survey-based interview study with 27 clinicians -comprising 10 ophthalmic specialists, 10 ophthalmic practitioners, and 7 oncologists. Participants were first introduced to core AI concepts and realistic clinical scenarios, then asked to assess AI-generated OCT analyses using standardized Likert-scale questions, allowing us to gauge their understanding, trust, and readiness to integrate AI into practice. METHODS We developed a questionnaire through literature review and consultations with ophthalmologists, computer scientists, and AI researchers. A single investigator interviewed 27 clinicians across three specialties and transcribed their responses. Data were summarized as medians (ranges) and compared with Mann-Whitney U tests (α = 0.05). RESULTS We noted important differences in the impact of various explainability methods on trust, depending on the clinical or AI scenario nature and the staff expertise. Explanations of AI outputs increased trust in the AI algorithm when outputs simply reflected ground truth expert opinion. When clinical scenarios were complex with incorrect AI outcomes, a mixed response to explainability led to correctly reduced trust in experienced clinicians but mixed feedback amongst less experienced clinicians. All clinicians had a general consensus on lack of current knowledge in interacting with AI and desire more training. CONCLUSIONS Clinicians' trust in AI algorithms are affected by explainability methods and factors, including AI's performance, personal judgments and clinical experience. The development of clinical AI systems should consider the above and these responses ideally be factored into real-world assessments. Use of this study's findings could help improve the real world validity of medical AI systems by enhancing the human-computer interactions, with preferred explainability techniques tailored to specific situations.
Collapse
Affiliation(s)
- Oskar Wysocki
- Cancer Research UK Manchester Institute, University of Manchester, Oxford Rd, Manchester M13 9PL, United Kingdom; Idiap Research Institute, National University of Sciences, Rue Marconi 19, CH - 1920 Martigny, Switzerland
| | - Sammie Mak
- St.Jame's University Hospital, Beckett St, Harehills, Leeds LS9 7TF, United Kingdom; Leeds Teaching Hospitals NHS Trust, Great George St, Leeds LS13EX, United Kingdom
| | - Hannah Frost
- Cancer Research UK Manchester Institute, University of Manchester, Oxford Rd, Manchester M13 9PL, United Kingdom
| | - Donna M Graham
- Cancer Research UK Manchester Institute, University of Manchester, Oxford Rd, Manchester M13 9PL, United Kingdom; The Christie HNS Foundation Trust, Wilmslow Rd, Manchester M204BX, United Kingdom
| | - Dónal Landers
- DeLondra Oncology Ltd, 38, Carlton Avenue, Wilmslow SK9 4EP, United Kingdom
| | - Tariq Aslam
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom; School of Health Sciences, University of Manchester, Oxford Road, Manchester M139PL, United Kingdom.
| |
Collapse
|
5
|
Zhou C, Wang J, Zhou L, Li H, Liu X, Wang S, Zhang X, Ye X, Ren H, Zeng K, Li X, Wang D, Ji J. The novel Piperine derivative YL-1-9 exhibits anti-breast Cancer effects by inducing apoptosis via the p53/p21 pathway. Bioorg Med Chem Lett 2025; 123:130231. [PMID: 40204112 DOI: 10.1016/j.bmcl.2025.130231] [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: 12/23/2024] [Revised: 03/25/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
The tumor suppressor protein p53 plays a crucial role in the pathogenesis of breast cancer; however, its function is often compromised due to MDM2 overexpression or mutations in the p53 gene, which occurs in approximately 30-35 % of breast cancer cases. Piperine, a natural bioactive compound, has shown potential in inhibiting breast cancer cell growth by upregulating p53 expression. However, its clinical application is hindered by poor bioavailability, potential toxicity, and the risk of undesirable drug interactions. In the present study, a novel derivative of Piperine, YL-1-9, was synthesized and evaluated for its anticancer activity against breast cancer. YL-1-9, a bicyclic amide derivative of Piperine, was evaluated for antitumor effects both in vitro and in vivo using MTT assays and the chick embryo chorioallantoic membrane (CAM) model. Further investigations into its effects on breast cancer cell clonogenicity, adhesion, invasion, and migration were conducted through colony formation assays, EdU assays, cell adhesion and invasion studies, and wound healing experiments. Western blot analysis was performed to elucidate the effects of YL-1-9 on the cell cycle and apoptosis, which were further validated using YO-PRO-1 and propidium iodide dual staining. YL-1-9 significantly inhibited breast cancer cell proliferation, adhesion, invasion, and migration, while inducing cell cycle arrest and promoting apoptosis. Mechanistically, YL-1-9 downregulated critical proteins in the CDK4/6-cyclin D-Rb-E2F pathway and the Caspase 3/Bax/Bcl-2 apoptosis signaling pathway. These findings position YL-1-9 as a promising candidate for breast cancer therapy; however, further clinical studies are necessary to fully assess its therapeutic potential.
Collapse
Affiliation(s)
- Chongyun Zhou
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Jiayun Wang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Lili Zhou
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Hanxue Li
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xing Liu
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Sen Wang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xingyu Zhang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xiaoqing Ye
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Hongyu Ren
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Kaile Zeng
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xiuming Li
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Dan Wang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Jing Ji
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China; College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, Jiangsu 225300, PR China.
| |
Collapse
|
6
|
Tang J, Li X, Tang N, Lin X, Du Y, Zhang S, Li Q, Zhang Y, Zhang Y, Hang H, Qiu T, Qiu Y, Cheng H, Dai Z, Hong H, Wei W, He J, Yan C. CD44 identified as a diagnostic biomarker for highly malignant CA19-9 negative pancreatic cancer. Cancer Lett 2025; 622:217713. [PMID: 40216152 DOI: 10.1016/j.canlet.2025.217713] [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: 02/26/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited diagnostic biomarkers. Carbohydrate antigen 19-9 (CA19-9) is a widely used clinical biomarker and is generally considered to correlate with PDAC malignancy. However, the relationship between CA19-9 expression levels and tumor aggressiveness remains underexplored. In this study, we report a biphasic relationship between CA19-9 expression levels and PDAC malignancy, where both negative (<5 U/mL) and high (>37 U/mL) CA19-9 levels are associated with increased tumor aggressiveness. We defined CA19-9 negative PDAC as tumors that lack CA19-9 expression intracellulary, on the cell membrane, and in secreted form. In PDAC cell lines and patient-derived organoids, CA19-9 negativity, confirmed by immunofluorescence, flow cytometry and ELISA, correlated with more aggressive behaviors. In PDAC patients, tumors from those with serum CA19-9 levels below 5 U/mL exhibited stronger metabolically activity, more immunosuppressive tumor microenvironment, and worse survival than CA19-9 positive tumors, with over 90 % showing absent CA19-9 expression by immunohistochemistry (IHC). Glycoproteomics profiling identified CD44 as a highly expressed biomarker in CA19-9 negative PDAC. Elevated CD44 expression effectively distinguished CA19-9 negative PDAC from both CA19-9 positive PDAC and CA19-9 negative benign pancreatic diseases, suggesting its potential as a diagnostic tool. Furthermore, we developed a radionuclide-labeled CD44 antibody 89Zr-1M2E3, which specifically recognized CA19-9 negative PDAC tumors in preclinical models using PET-CT imaging. These findings highlight CD44 as a promising biomarker and therapeutic target for diagnosing and treating CA19-9 negative PDAC.
Collapse
Affiliation(s)
- Jiatong Tang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaoyang Li
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Neng Tang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiawen Lin
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixiang Du
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China
| | - Shuo Zhang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qi Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yifan Zhang
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hexing Hang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tongtong Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hao Cheng
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhan Dai
- Nanjing Okay Biotechnology Co., Ltd, Nanjing, Jiangsu Provinve, China
| | - Hao Hong
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Wei Wei
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
7
|
Liu X, Lv M, Feng B, Gong Y, Min Q, Wang Y, Wu Q, Chen J, Zhao D, Li J, Zhang W, Zhan Q. SQLE amplification accelerates esophageal squamous cell carcinoma tumorigenesis and metastasis through oncometabolite 2,3-oxidosqualene repressing Hippo pathway. Cancer Lett 2025; 621:217528. [PMID: 39924077 DOI: 10.1016/j.canlet.2025.217528] [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: 10/18/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent cancers worldwide, characterized by a dismal prognosis and elusive therapeutic targets. Dysregulated cholesterol metabolism is a critical hallmark of cancer cells, facilitating tumor progression. Here, we used whole genome sequencing data from several ESCC cohorts to identify the important role of squalene epoxidase (SQLE) in promoting ESCC tumorigenesis and metastasis. Specifically, our findings highlight the significance of 2,3-oxidosqualene, an intermediate metabolite of cholesterol biosynthesis, synthesized by SQLE and metabolized by lanosterol synthase (LSS), as a key regulator of ESCC progression. Mechanistically, the interaction between 2,3-oxidosqualene and vinculin enhances the nuclear accumulation of Yes-associated protein 1 (YAP), thereby increasing YAP/TEAD-dependent gene expression and accelerating both tumor growth and metastasis. In a 4-nitroquinoline 1-oxide (4-NQO)-induced ESCC mouse model, overexpression of Sqle resulted in accelerated tumorigenesis compared to wild-type controls, highlighting the pivotal role of SQLE in vivo. Furthermore, elevated SQLE expression in ESCC patients correlates with a poorer prognoses, suggesting potential therapeutic avenues for treatment. In conclusion, our study elucidates the oncogenic function of 2,3-oxidosqualene as a naturally occurring metabolite and proposes modulation of its levels as a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Xuesong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Peking University International Cancer Institute, Beijing, 100191, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Mengzhu Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Bicong Feng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Ying Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jie Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Dongyu Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jinting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Peking University International Cancer Institute, Beijing, 100191, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Soochow University Cancer Institute, Suzhou, 215127, China; Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China.
| |
Collapse
|
8
|
Pflug C, Müller F, Koseki JC, Petersen C, Nienstedt JC, Tribius S. Objective dysphagia is very common after radiotherapy in oropharyngeal cancer patients. Oral Surg Oral Med Oral Pathol Oral Radiol 2025; 140:54-63. [PMID: 40169338 DOI: 10.1016/j.oooo.2025.02.008] [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: 08/08/2024] [Revised: 11/30/2024] [Accepted: 02/08/2025] [Indexed: 04/03/2025]
Abstract
OBJECTIVE Dysphagia is one of the most serious adverse events in the treatment of head and neck cancer. This cross-sectional study aimed to assess pharyngeal residue, and penetration/aspiration in oropharyngeal cancer patients (OPC) after radiotherapy using flexible endoscopic evaluation of swallowing (FEES). METHODS A total of 35 OPC patients who had received radio-(chemo) therapy (R(C)T), including 8 patients with primary R(C)T), were included and examined by FEES to determine the swallowing status and were asked to indicate their swallowing ability on a visual scale to reflect the problem perceived by the patient. During FEES the patients were given three standardized bolus consistencies and four test pills. Penetration, aspiration, and residue were evaluated and classified. RESULT Relevant dysphagia was present in 23/35 (66%) patients. Almost half of all patients (15/35) showed aspiration (53% (8/15) silent). Residue occurred in 91% but without correlation to aspiration. A significant association between dysphagia and impaired pill swallowing was found (P = .003) occurring in 20 of 35 patients. Even in patients with small tumors and without prior surgery severe dysphagia was found. CONCLUSIONS Severe dysphagia is frequent after R(C)T affecting more than half of the patients with OPC. The frequent impaired pill swallowing ability should be considered Therefore, regular dysphagia diagnostics in the follow-up setting are advisable to initiate appropriate treatment and raise patients' quality of life, prevent aspiration pneumonia, and improve overall outcomes after tumor therapy.
Collapse
Affiliation(s)
- Christina Pflug
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Frank Müller
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana-Christiane Koseki
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Petersen
- Department of Radiotherapy and Radiation Oncology, Center for Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julie Cläre Nienstedt
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Silke Tribius
- Department of Radiation Oncology, Asklepios Hospital St. Georg, Hamburg, Germany; Hermann-Holthusen Institute for Radiation Oncology, Asklepios Hospital St. Georg, Hamburg, Germany
| |
Collapse
|
9
|
Sun C, He Q, Yang X, Wang J, Xia D, Xia T, Liao H, Xiong X, Liao Y, Shen H, Sun Q, Yuan Y, He Y, Liu L. A novel NIR-dependent IDO-inhibiting ethosomes treatment melanoma through PTT/PDT/immunotherapy synergy. Colloids Surf B Biointerfaces 2025; 251:114565. [PMID: 39999696 DOI: 10.1016/j.colsurfb.2025.114565] [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: 12/26/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Phototherapy is a treatment method that uses the characteristics of different bands of light to treat diseases. Tumor immunotherapy, on the other hand, treats tumors by regulating the body's immune system. The combination of phototherapy and immunotherapy can significantly enhance the treatment of melanoma. In this study, we prepared and characterized INEs, a novel ethosome composed of the photosensitizer IR251 and the Indoleamine-2, 3-dioxygenase (IDO) inhibitor NLG919. INEs demonstrated excellent phototherapeutic properties, strong phototoxicity, and a notable ability to inhibit IDO. Under 808 nm laser irradiation, INEs effectively induced immunogenic cell death (ICD) in melanoma cells. In vivo experiments demonstrated that INEs injection into primary tumors triggered ICD, promoted maturation of DC cells, and activated naive T cells, leading to the production of effector T cells (specifically CD4+ and CD8+ T cells) that targeted and killed tumor cells. Both primary and distant tumors were treated simultaneously with favorable biosafety. In conclusion, INEs represent a promising strategy for melanoma treatment by a combination of phototherapy and immunotherapy with high safety. This study provides new insights and a theoretical basis for the clinical treatment of melanoma.
Collapse
Affiliation(s)
- Changzhen Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, Luzhou Key Laboratory of Research and Development of Medical Institution Preparations and Large-scale Health Products, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Qingqing He
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xun Yang
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jianv Wang
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Dengmei Xia
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Tong Xia
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Hongye Liao
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yongmei Liao
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Hongping Shen
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, Luzhou Key Laboratory of Research and Development of Medical Institution Preparations and Large-scale Health Products, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Qin Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, Luzhou Key Laboratory of Research and Development of Medical Institution Preparations and Large-scale Health Products, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yuan Yuan
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, Luzhou Key Laboratory of Research and Development of Medical Institution Preparations and Large-scale Health Products, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yuanmin He
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| | - Li Liu
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
10
|
Li S, Zhang J, Wei W, Zhang Z, Huang W, Xia L. The important role of myeloid-derived suppressor cells: From hepatitis to liver cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189329. [PMID: 40262654 DOI: 10.1016/j.bbcan.2025.189329] [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: 09/26/2024] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
Liver homeostasis is coordinated by crosstalk between resident and infiltrating inflammatory cells. Liver disease creates a dynamic inflammatory microenvironment characterized by aberrant metabolism and continuous hepatic regeneration, making it an important risk factor for hepatocellular carcinoma (HCC) as well as liver failure. Recent studies have revealed a critical heterogeneous population of myeloid-derived suppressor cells (MDSCs), which influence liver disease progression and malignancy by dynamically regulating the immune microenvironment. MDSCs play an important role in preventing excessive immune responses in the liver. However, MDSCs are also associated with the promotion of liver injury and liver cancer progression. The plasticity of MDSCs in liver disease is a unique challenge for therapeutic intervention strategies and requires a deeper understanding of the underlying mechanisms. Here, we review the role of MDSCs in the establishment and progression of liver disease and highlight the evidence for MDSCs as a priority target for current and future therapeutic strategies. We explore the fate of MDSCs from hepatitis to liver cancer, providing recent insights into potential targets for clinical intervention.
Collapse
Affiliation(s)
- Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Wang Wei
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhicheng Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
11
|
Hu X, Chang H, Guo Y, Yu L, Li J, Zhang B, Zhao H, Xu J, Pan G, Zhang K, Lü M, Cui H. Mori Folium ethanol extracts induce ferroptosis and suppress gastric cancer progression by inhibiting the AKT/GSK3β/NRF2 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156789. [PMID: 40344847 DOI: 10.1016/j.phymed.2025.156789] [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: 12/31/2024] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Mori Folium, the leaf of Morus alba L., is a traditional Chinese medicine (TCM) known for its diverse pharmacological activities, including anti-inflammatory and immunomodulatory effects. While the Morus alba itself has been reported to contain various bioactive compounds with anticancer properties, the anticancer activity of Mori Folium and its underlying mechanisms remain insufficiently understood. PURPOSE This study aimed to investigate the effects of Mori Folium ethanol extracts (MFEE) on gastric cancer (GC) and to elucidate its underlying mechanisms. METHODS To investigate the anti-GC properties of MFEE, CCK-8, colony formation, EdU, flow cytometry, and soft agar, scratch, and transwell assays were employed. Western blot was employed to analyze the expression of ferroptotic proteins, while ferroptotic cellular events were also assessed, including iron accumulation, GSH levels, reactive oxygen species (ROS) production, mitochondrial changes, and lipid peroxidation. The chemical profile of MFEE was characterized using a UPLC-ESI-MS/MS system. Additionally, network pharmacology analysis was performed to investigate the potential anti-GC mechanisms of MFEE. Finally, the in vivo anti-cancer effects of MFEE were evaluated using a subcutaneous mouse model, with hematoxylin and eosin (H&E) and immunohistochemistry (IHC) staining to assess histopathological and molecular changes. RESULTS This study demonstrated that MFEE suppresses GC cell proliferation, blocks the G1-S cell cycle transition, and inhibits migration and invasion by promoting Fe²⁺ accumulation, increasing MDA levels and ROS, depleting GSH, and downregulating the expression of xCT and GPX4, thereby inducing ferroptosis. Chemical analysis identified 1596 phytochemicals, including 35 bioactive compounds. The induction of ferroptosis by MFEE was associated with the inhibition of the PI3K/AKT signaling pathway, modulating the AKT/GSK3β/NRF2 axis. Activation of AKT by SC79 was found to mitigate MF-induced ferroptosis. Notably, MFEE enhanced the chemosensitivity of GC cells to cisplatin, potentially through ferroptosis induction. CONCLUSION This study revealed that MFEE induces ferroptosis in GC cells by modulating the PI3K/AKT signaling pathway, enhancing chemosensitivity to cisplatin, and providing a potential therapeutic strategy for GC.
Collapse
Affiliation(s)
- Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Hongbo Chang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yan Guo
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Lang Yu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Jing Li
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Bili Zhang
- School of Life Science, Chongqing University, Chongqing 400044, China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jingyang Xu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Guangzhao Pan
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China.
| | - Kui Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Muhan Lü
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
12
|
Zhang C, Yin X, Jiang J, Wang P, Wang Y. Downregulation of GFPT2 enhances cisplatin chemotherapy sensitivity in STK11/KRAS mutant non-small cell lung cancer by regulating the hexosamine biosynthesis pathway, resisting tumor growth. Cytokine 2025; 191:156943. [PMID: 40253947 DOI: 10.1016/j.cyto.2025.156943] [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: 01/03/2025] [Revised: 03/21/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE To explore the role of GFPT2 in the sensitivity of STK11/KRAS lung cancer cells to cisplatin chemotherapy, and its underlying mechanism. MATERIALS & METHODS A549 and H460 cells were used to analyze the effect of GFPT2 on cisplatin chemotherapy sensitivity, as both carry KRAS mutations and H460 has LKB1 inactivation mutations, meeting the requirements of a KRAS/LKB1 co mutant tumor model. The levels of UDP-GlcNAc, OGT, OGA, and O-GlcNAc in the HBP pathway were also determined. To verify the potential role of HBP, we added OGT inhibitors. In vivo, we constructed a nude mouse model bearing A549 tumor to further validate the results of in vitro cell experiments. RESULTS GFPT2 silencing can significantly inhibit cell proliferation, invasion, and migration, promote cell apoptosis, and enhance the effect of cisplatin (p < 0.05). After OSMI-1 processing, GFPT2 enhances O-GlcNAc modification levels via the OGT-mediated HBP, thereby decreasing the sensitivity of STK11/KRAS mutant cells to cisplatin chemotherapy. In addition, GFPT2 silencing enhances the chemotherapy sensitivity of cisplatin and inhibits tumor growth, while overexpression of GFPT2 weakens this effect (p < 0.05). The above results provide new targets and combination therapy options for the clinical treatment of KRAS/LKB1 mutant lung cancer. CONCLUSION Our study found that inhibiting GFPT2 can enhance the chemotherapy sensitivity of cisplatin to STK11/KRAS/LKB1 mutant NSCLCs cells through the OGT mediated HBP pathway, filling a key gap in the chemotherapy resistance mechanism of KRAS/LKB1 mutant lung cancer.
Collapse
Affiliation(s)
- Cheng Zhang
- Thoracic Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Xuelei Yin
- Yantai Keyou Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Jun Jiang
- Key Laboratory of Genetics Research and Evaluation of the National Drug Administration, Shandong Food and Drug Inspection and Research Institute, Jinan, Shandong, China
| | - Peng Wang
- Ministry of Scientific and Technological Innovation, Yantai Hi-tech Industrial Development Zone, Yantai, Shandong, China
| | - Yirong Wang
- Department of Radiotherapy, Yantaishan Hospital, Yantai, Shandong, China.
| |
Collapse
|
13
|
Luo XR, Huang LZ, Yin J, Xiong ZM, Li WX, Liao C, Lin ML, Huang W, Zhang S. FSTL3 promotes colorectal cancer by activating the HIF1 pathway. Gene 2025; 954:149435. [PMID: 40154584 DOI: 10.1016/j.gene.2025.149435] [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: 01/06/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Follistatin-like 3 (FSTL3) is a glycoprotein known to promote tumor growth, invasion, and angiogenesis in various cancers. However, its role in Colorectal Cancer (CRC), particularly concerning the hypoxia-inducible factor 1α (HIF1α) signaling pathway, remains unclear. The HIF1α pathway is critical in CRC progression, enabling tumor cells to adapt to hypoxia through angiogenesis, Epithelial-Mesenchymal Transition (EMT), and metabolic reprogramming. Analysis of The Cancer Genome Atlas (TCGA) and GSE39582 datasets revealed that FSTL3 is significantly upregulated in CRC tissues and correlates with poor Overall Survival (OS), Progression-Free Survival (PFS), and aggressive features such as venous, lymphatic, and perineural invasion. In vitro experiments demonstrated that FSTL3 overexpression in HCT15 and HCT116 cells promoted proliferation, migration, and cell cycle progression, whereas knockdown in LOVO and Caco2 cells suppressed these processes and induced apoptosis. Transcriptome sequencing and western blot analysis indicated that FSTL3 activated the HIF1α pathway by upregulating HIF1α, ANGPT2, and HK3, which are key regulators of angiogenesis and glycolysis. Importantly, treatment with the HIF1α inhibitor KC7F2 reversed the oncogenic effects of FSTL3 overexpression both in vitro and in vivo. In xenograft and tail vein metastasis models, KC7F2 suppressed tumor growth, reduced pulmonary metastasis, and restored lung tissue integrity, further downregulating FSTL3 and HIF1α expression. These findings suggest that FSTL3 promotes CRC progression via the HIF1α pathway and highlight its potential as a prognostic biomarker and therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Xiang-Rong Luo
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China; The Central Hospital of Shaoyang, No. 36, Hongqi Road, Shaoyang City, Hunan Province 422000, PR China
| | - Li-Zhe Huang
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Jie Yin
- The Central Hospital of Shaoyang, No. 36, Hongqi Road, Shaoyang City, Hunan Province 422000, PR China
| | - Zu-Ming Xiong
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Wen-Xin Li
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Cun Liao
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Ming-Lin Lin
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Wei Huang
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China
| | - Sen Zhang
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, PR China.
| |
Collapse
|
14
|
Tang M, Song K, Xie D, Yuan X, Wang Y, Li Z, Lu X, Guo L, Zhu X, Xiong L, Zhou W, Lin J. PSAT1 promotes the progression of colorectal cancer by regulating Hippo-YAP/TAZ-ID1 axis via AMOT. Mol Cell Biochem 2025; 480:3647-3668. [PMID: 39739271 PMCID: PMC12095340 DOI: 10.1007/s11010-024-05194-8] [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: 08/13/2024] [Accepted: 12/14/2024] [Indexed: 01/02/2025]
Abstract
Colorectal cancer (CRC) ranks third for morbidity and second for mortality among all digestive malignant tumors worldwide, but its pathogenesis remains not entirely clear. Bioinformatic analyses were performed to find out important biomarkers for CRC. For validation, reverse transcription-quantitative PCR, western blotting, and immunohistochemistry were performed. Then, cell transfection, gain- and loss-of-function assays, immunofluorescence, cell line RNA-sequencing and analyses, and in vivo tumorigenesis assay were also performed to further explore the mechanism. We prioritized phosphoserine aminotransferase 1 (PSAT1) as an important biomarker in CRC. PSAT1 expression was gradually up-regulated as the CRC disease progresses and may relate to poor prognosis. PSAT1 promoted the malignant behaviors of CRC cells. Although PSAT1 is an enzyme essential to serine biosynthesis, an exogenous supplement of serine did not completely rescue the malignant behaviors in PSAT1-knockdown CRC cells. Interestingly, PSAT1 inhibited the Hippo tumor-suppressor pathway by promoting the nucleus-localization of YAP/TAZ and increasing the expression of ID1 in CRC cells. Furthermore, AMOT, a vascular-related molecule that molecularly interacts with YAP/TAZ, was up-regulated upon PSAT1 knockdown in CRC cells. Knocking down AMOT partially rescued the inhibition of proliferation and the reduced nuclear localization of YAP/TAZ caused by PSAT1 knockdown in CRC cells. Moreover, PSAT1 was closely related to vascular-related pathways, in which AMOT might act as a mediator. Finally, PSAT1 promoted CRC proliferation by negatively regulating AMOT in vivo. PSAT1 could enhance the progression of colorectal cancer by regulating Hippo-YAP/TAZ-ID1 axis via AMOT, which is independent of the metabolic function of PSAT1.
Collapse
Affiliation(s)
- Minshan Tang
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Kai Song
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Danning Xie
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Xinyu Yuan
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Yaxuan Wang
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Zhiyang Li
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Xiansheng Lu
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Liang Guo
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Xiaotong Zhu
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Le Xiong
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Wenqian Zhou
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
15
|
Shiono A, Imai H, Endo S, Okazaki S, Abe T, Mouri A, Kaira K, Masubuchi K, Kobayashi K, Minato K, Kato S, Kagamu H. Clinical significance of post‑progression survival after chemoradiotherapy on overall survival in limited‑disease small cell lung cancer. Mol Clin Oncol 2025; 22:58. [PMID: 40322546 PMCID: PMC12046617 DOI: 10.3892/mco.2025.2853] [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: 11/25/2024] [Accepted: 03/19/2025] [Indexed: 05/08/2025] Open
Abstract
Overall survival (OS) among patients with limited-disease small-cell lung cancer (LD-SCLC) receiving chemoradiotherapy can be significantly influenced by subsequent treatments. Thus, the present study aimed to examine the interplay between progression-free survival (PFS), post-progression survival (PPS) and OS in patients with LD-SCLC undergoing chemoradiotherapy. This study retrospectively analyzed 84 patients with relapsed LD-SCLC who received chemoradiotherapy between April 2007 and June 2021. The correlations between PFS and OS as well as PPS and OS post-chemoradiotherapy were analyzed at the individual patient level. Spearman's rank correlation and linear regression analyses revealed a robust correlation between PPS and OS (r=0.76; P<0.05; R2=0.85). PFS was moderately correlated with OS (r=0.57; P<0.05; R2=0.25). Furthermore, the presence of liver metastases upon relapse and the administration of platinum re-challenge chemotherapy were significantly associated with PPS (P<0.05). The analysis of relationships between OS and PFS as well as OS and PPS in this patient cohort revealed that OS was more strongly correlated with PPS than PFS. These findings suggest that factors such as liver metastases at relapse and the administration of platinum re-challenge chemotherapy may influence PPS.
Collapse
Affiliation(s)
- Ayako Shiono
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
| | - Hisao Imai
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
| | - Satoshi Endo
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
| | - Shohei Okazaki
- Division of Radiation Oncology, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
| | - Takanori Abe
- Department of Radiation Oncology, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
| | - Atsuto Mouri
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
| | - Kyoichi Kaira
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
| | - Ken Masubuchi
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
| | - Kunihiko Kobayashi
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
| | - Koichi Minato
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
- Division of Health Evaluation and Promotion, SUBARU Health Insurance Society, Ota Memorial Hospital, Ota, Gunma 373-0055, Japan
| | - Shingo Kato
- Department of Radiation Oncology, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
| | - Hiroshi Kagamu
- Department of Respiratory Medicine, Comprehensive Cancer Center, International Medical Center, Saitama Medical University, Hidaka, Saitama 350-1298, Japan
| |
Collapse
|
16
|
Mostafavi M, Ghazi F, Mehrabifard M, Alivirdiloo V, Hajiabbasi M, Rahimi F, Mobed A, Taheripak G, Ramezani Farani M, Huh YS, Bakhtiyari S, Alipourfard I. State-of-the-art application of nanoparticles in radiotherapy: a platform for synergistic effects in cancer treatment. Strahlenther Onkol 2025; 201:577-588. [PMID: 39367110 PMCID: PMC12119707 DOI: 10.1007/s00066-024-02301-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/24/2024] [Indexed: 10/06/2024]
Abstract
Radiotherapy (RT) is a gold standard cancer treatment worldwide. However, RT has limitations and many side effects. Nanoparticles (NPs) have exclusive properties that allow them to be used in cancer therapy. Consequently, the combination of NP and RT opens up a new frontier in cancer treatment. Among NPs, gold nanoparticles (GNPs) are the most extensively studied and are considered ideal radiosensitizers for radiotherapy due to their unique physicochemical properties and high X‑ray absorption. This review analyzes the various roles of NPs as radiosensitizers in radiotherapy of glioblastoma (GBS), prostate cancer, and breast cancer and summarizes recent advances. Furthermore, the underlying mechanisms of NP radiosensitization, including physical, chemical, and biological mechanisms, are discussed, which may provide new directions for next-generation GNP optimization and clinical transformation.
Collapse
Affiliation(s)
- Mehrnaz Mostafavi
- Faculty of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhood Ghazi
- Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Vahid Alivirdiloo
- Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | | | - Fatemeh Rahimi
- Division of Clinical Laboratory, Zahra Mardani Azar Children Training Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Taheripak
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon, Korea (Republic of)
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon, Korea (Republic of)
| | - Salar Bakhtiyari
- Department of Clinical Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Iraj Alipourfard
- Iraj Alipourfard, Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
17
|
Cazzaniga ME, Huober J, Tamma A, Emde A, Thoele K, O'Shaughnessy J. Oral Anticancer Therapies: Addressing Nonadherence in Patients With Breast Cancer. Clin Breast Cancer 2025; 25:307-324. [PMID: 39800641 DOI: 10.1016/j.clbc.2024.12.011] [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: 03/26/2024] [Revised: 11/28/2024] [Accepted: 12/15/2024] [Indexed: 05/25/2025]
Abstract
This review aims to investigate the issue of treatment nonadherence and to present the available strategies to improve adherence to oral treatments in breast cancer. A literature search was conducted to contextualise the issue of nonadherence, investigate the reasons behind nonadherence, and demonstrate strategies to address treatment nonadherence in breast cancer. Findings indicate that adherence rates decrease while discontinuation rates increase with increasing lengths of breast cancer treatment course. Lack of adherence is proven to be detrimental to treatment outcomes. Patients struggle to adhere to treatment due to inadequate relationships with healthcare providers, lack of information, psychological distress, and side effects. Healthcare providers should evaluate patient's experience to provide the necessary support. Following this assessment, healthcare providers may recommend interventions addressing patient knowledge, psychological distress or side effects. Treatment adherence remains an issue for oral therapeutics in breast cancer. After patient assessment, healthcare providers can offer personalised strategies to improve treatment adherence. The most crucial interventions address patient knowledge, psychological distress, and side effects.
Collapse
Affiliation(s)
- M E Cazzaniga
- Scientific Department, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
| | - J Huober
- Chief Physician, Breast Center, St. Gallen, Switzerland
| | - A Tamma
- Lilly Oncology Breast Cancer, Eli Lilly and Company, Indianapolis, IN
| | - A Emde
- Lilly Oncology Breast Cancer, Eli Lilly and Company, Indianapolis, IN
| | - K Thoele
- Lilly Oncology Breast Cancer, Eli Lilly and Company, Indianapolis, IN
| | | |
Collapse
|
18
|
Ji Y, Liu L, Liu Y, Ma Y, Ji Z, Wu X, Zhu Q. Exploring gene biomarkers and targeted drugs for ferroptosis and cuproptosis in osteosarcoma: A bioinformatic approach. ENVIRONMENTAL TOXICOLOGY 2025; 40:891-901. [PMID: 38546286 PMCID: PMC12069744 DOI: 10.1002/tox.24250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 05/14/2025]
Abstract
Osteosarcoma predominantly affects adolescents and young adults and is characterized as a malignant bone tumor. In recent decades, substantial advancements have been achieved in both diagnosing and treating osteosarcoma. Resulting in enhanced survival rates. Despite these advancements, the intricate relationship between ferroptosis and cuproptosis genes in osteosarcoma remains inadequately understood. Leveraging TARGET and GEO datasets, we conducted Cox regression analysis to select prognostic genes from a cohort of 71 candidates. Subsequently, a novel prognostic model was engineered using the LASSO algorithm. Kaplan-Meier analysis demonstrated that patients stratified as low risk had a substantially better prognosis compared with their high-risk counterparts. The model's validity was corroborated by the area under the receiver operating characteristic (ROC) curve. Additionally, we ascertained independent prognostic indicators, including clinical presentation, metastatic status, and risk scores, and crafted a clinical scoring system via nomograms. The tumor immune microenvironment was appraised through ESTIMATE, CIBERSORT, and single-sample gene set enrichment analysis. Gene expression within the model was authenticated through PCR validation. The prognostic model, refined by Cox regression and the LASSO algorithm, comprised two risk genes. Kaplan-Meier curves confirmed a significantly improved prognosis for the low-risk group in contrast to those identified as high-risk. For the training set, the ROC area under the curve (AUC) values stood at 0.636, 0.695, and 0.729 for the 1-, 3-, and 5-year checkpoints, respectively. Although validation set AUCs were 0.738, 0.668, and 0.596, respectively. Immune microenvironmental analysis indicated potential immune deficiencies in high-risk patients. Additionally, sensitivity to three small molecule drugs was investigated in the high-risk cohort, informing potential immunotherapeutic strategies for osteosarcoma. PCR analysis showed increased mRNA levels of the genes FDX1 and SQLE in osteosarcoma tissues. This study elucidates the interaction of ferroptosis and cuproptosis genes in osteosarcoma and paves the way for more targeted immunotherapy.
Collapse
Affiliation(s)
- Yingnan Ji
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Lv Liu
- Benxi Central HospitalBenxiChina
| | - Yu Liu
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Yudong Ma
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Zhenhua Ji
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Xiaodan Wu
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Qi Zhu
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| |
Collapse
|
19
|
Yuk JS, Lee JL, Shin Y, Han GH, Yoon SH, Kim MH, Noh JH, Lee Y, Kim J, Yoon SY, Cho H, Yang K, Bae BN, Kim KW, Gwak G. The effect of menopausal hormone therapy on thyroid cancer survivors from the National Health Insurance Database in South Korea cohort. Eur J Obstet Gynecol Reprod Biol 2025; 310:113983. [PMID: 40273642 DOI: 10.1016/j.ejogrb.2025.113983] [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: 01/20/2025] [Revised: 03/04/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE There is a lack of research on how menopausal hormone therapy (MHT) affects the prognosis of postmenopausal thyroid cancer (TC) survivors. This study aimed to evaluate the association between MHT and the risk of death in postmenopausal TC survivors. METHODS This retrospective cohort study used the data of women with the diagnostic code for TC from the Korean National Health Insurance Database between 2007 and 2021. The postmenopausal women with TC who received radical treatment such as surgery or/and radioactive iodine therapy were divided into two groups - MHT group and non-MHT group - according to whether or not they received MHT after TC treatment. The risk of death in TC survivors was analysed according to the type and duration of the MHT regimen. RESULTS Among postmenopausal TC survivors, the risk of death did not differ between the women with MHT duration > 5 years and the non-MHT group, with a hazard ratio (HR) of 0.964 [95 % confidence interval (CI) 0.697-1.334] (p = 0.826). However, MHT duration < 5 years was associated with increased risk of death, with the MHT group having an HR of 1.744 (95 % CI 1.105-2.753) (p = 0.017) compared with the non-MHT group. A trend was observed towards increased risk of death in women with MHT duration ≤ 6 months, with an HR of 1.703 (95 % CI 0.98-2.962) (p = 0.059). The risk of death did not differ between women with MHT duration > 6 months and women in the non-MHT group (HR 1.205, 95 % CI 0.79-1.839) (p = 0.064). In analyses by MHT regimen, no increase in the risk of death was observed for any MHT regimen for any duration. CONCLUSION The risk of death in postmenopausal TC survivors who used MHT for > 5 years did not differ from that of their counterparts who did not use MHT. However, postmenopausal TC survivors who used MHT for < 5 years, particularly those who used MHT for < 6 months, had a higher risk of death than their counterparts who did not use MHT.
Collapse
Affiliation(s)
- Jin-Sung Yuk
- Department of Obstetrics and Gynaecology, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Jin Li Lee
- Department of Obstetrics and Gynaecology, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Yeonjin Shin
- Department of Obstetrics and Gynaecology, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Gwan Hee Han
- Department of Obstetrics and Gynaecology, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Sang-Hee Yoon
- Department of Obstetrics and Gynaecology, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Myoung Hwan Kim
- Department of Obstetrics and Gynaecology, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Ji Hyun Noh
- Department of Obstetrics and Gynaecology, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Yujin Lee
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Jungbin Kim
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Sam-Youl Yoon
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Hyunjin Cho
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Keunho Yang
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Byung Noe Bae
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Ki Whan Kim
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea
| | - Geumhee Gwak
- Department of Surgery, Sanggye Paik Hospital, School of Medicine, Inje University, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Herichová I, Reis R, Vanátová D. Differences in the role of Gper1 in colorectal cancer progression depending on sex. Oncol Lett 2025; 29:305. [PMID: 40321663 PMCID: PMC12046377 DOI: 10.3892/ol.2025.15051] [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: 01/23/2025] [Accepted: 02/13/2025] [Indexed: 05/08/2025] Open
Abstract
To evaluate the role of 17β-oestradiol (E2) in the sex-dependent progression of colorectal cancer (CRC), the present study focused on E2 signalling mediated via the nuclear receptors [oestrogen receptor (ESR)1 and ESR2] and the membrane G protein-coupled oestrogen receptor 1 (Gper1) in males and females diagnosed with CRC. This study also investigated Gper1 signalling in the CRC cell lines DLD1 and LoVo, which differ in the p53 pathway. In cancer tissue, Gper1 becomes by far the most abundant E2 receptor due to an increase in Gper1 and a decrease in ESR2 expression. These changes are more prominent in males than in females. More pronounced differences in Gper1 expression between cancer and adjacent tissues were observed in males in lower stages compared with those in higher stages of disease and females. High expression of Gper1 was associated with worse survival in males without nodal involvement but not in females. The expression of E2 receptors in the CRC cell lines DLD1 and LoVo resembles that of human cancer tissue. Silencing of Gper1 (siGper1) caused an increase in the rate of metabolism in LoVo cells with wild-type tp53. In DLD1 cells with the mutated form of tp53, siGper1 did not exert this effect. High levels of Gper1 were associated with worse survival and could contribute to sex-dependent changes in the CRC prognosis. Tumour suppressor effects of Gper1 were, at least to some extent, dependent on signalling downstream of p53, which was more frequently deficient in males than in females. Overall, this suggests that up-regulation of Gper1 (or administration of a Gper1 agonist) would be more beneficial for patients with wild-type tp53.
Collapse
Affiliation(s)
- Iveta Herichová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| | - Richard Reis
- First Surgery Department, University Hospital, Comenius University in Bratislava, 81107 Bratislava, Slovak Republic
| | - Denisa Vanátová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| |
Collapse
|
21
|
Cobbs C, Chesnut GT, Shafi AA. Understanding Racial Disparities in Prostate Cancer: A Multifaceted Approach. Cancer Med 2025; 14:e70979. [PMID: 40444484 PMCID: PMC12123386 DOI: 10.1002/cam4.70979] [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: 01/14/2025] [Revised: 05/05/2025] [Accepted: 05/12/2025] [Indexed: 06/02/2025] Open
Abstract
Prostate cancer (PCa) remains a significant public health challenge in the United States, disproportionately affecting African American (AA) men, who face higher incidence rates, more aggressive disease, and elevated mortality compared to Caucasian American (CA) men. This review explores the multifactorial underpinnings of these disparities, integrating genomic, socioeconomic, environmental, and systemic contributors. Genomic analyses reveal that AA men harbor distinct molecular alterations, including higher frequencies of FOXA1, BRAF, and CHD1 mutations, as well as DNA damage repair defects, highlighting the critical need for population-specific precision medicine. Immune-oncologic pathways and stromal interactions within the tumor microenvironment further underscore biological differences driving aggressive disease phenotypes. Concurrently, adverse social determinants-including limited access to care, lower PSA screening rates, delayed treatment, medical mistrust, and underrepresentation in clinical trials-contribute to poorer outcomes. Despite these challenges, evidence from equal-access healthcare systems indicates that when provided equitable treatment, AA men can achieve outcomes comparable to or better than their CA counterparts. This review emphasizes actionable strategies to reduce disparities, including increasing AA representation in clinical trials, enhancing culturally competent patient-provider communication, improving access to early detection and high-quality care, and expanding community-based outreach initiatives. A holistic, interdisciplinary approach is essential to dismantle systemic barriers and achieve health equity in prostate cancer outcomes.
Collapse
Affiliation(s)
- Charles Cobbs
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of SurgeryUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Gregory T. Chesnut
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of SurgeryUniformed Services University of the Health SciencesBethesdaMarylandUSA
- Urology Service, Department of SurgeryWalter Reed National Military Medical CenterBethesdaMarylandUSA
| | - Ayesha A. Shafi
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of SurgeryUniformed Services University of the Health SciencesBethesdaMarylandUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc.BethesdaMarylandUSA
| |
Collapse
|
22
|
Chen X, Beilman B, Gibbs HD, Hamilton JL, Parker N, Bur AM, Caudell JJ, Gan GN, Hamilton-Reeves JM, Jim HSL, Kirtane K, Lominska C, Crowder SL, Arthur AE. Nutrition in head and neck cancer care: a roadmap and call for research. Lancet Oncol 2025; 26:e300-e310. [PMID: 40449504 DOI: 10.1016/s1470-2045(25)00087-7] [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: 11/11/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 06/03/2025]
Abstract
Survivors of head and neck cancer have complex nutritional and supportive care needs. These needs result from the tumour's proximity to organs essential for normal eating function and the intensive treatment targeting those organs. Despite the crucial role of nutrition and supportive care in head and neck cancer, research and funding are lacking compared with other cancer types. This Review was compiled and written by a team of multidisciplinary medical professionals. Topics include poor access to medical nutrition therapy (MNT), MNT reimbursement policies, long-term survivorship care needs, percutaneous endoscopic gastrostomy tube placement, nutrition literacy, psychological services, speech-language pathology care, and concomitant physical activity. The goal of this work is to define current issues in research and practice, advocate for the expansion of head and neck cancer funding opportunities, and raise awareness of head and neck cancer supportive care needs and challenges. This work provides a roadmap for health-care professionals, researchers, policy makers, and funding agencies to prioritise nutrition in head and neck cancer care, with the overarching goal of improving treatment outcomes and quality of life.
Collapse
Affiliation(s)
- Xin Chen
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brooke Beilman
- Department of Communication Sciences and Disorders, Maryville University, St Louis, MO, USA; Department of Speech-Language Pathology, Mercy Medical Center, Cedar Rapids, IA, USA; TIMS Medical, Boston, MA, USA
| | - Heather D Gibbs
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jessica L Hamilton
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nathan Parker
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | - Andrés M Bur
- Department of Otolaryngology - Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jimmy J Caudell
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Gregory N Gan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jill M Hamilton-Reeves
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, USA; Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather S L Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | - Kedar Kirtane
- Department of Head and Neck - Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Chris Lominska
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sylvia L Crowder
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | - Anna E Arthur
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
23
|
Wang L, Li Q, Sun Y, Wang S, Fu X, Wang X, Zheng Y, Gao A, Sun Y, Li J. Tumor-derived immunoglobulin-like transcript 3 inhibition reshapes the immunosuppressive tumor microenvironment and potentiates programmed cell death ligand 1 blockade immunotherapy in lung adenocarcinoma. Transl Oncol 2025; 56:102381. [PMID: 40199156 PMCID: PMC12008602 DOI: 10.1016/j.tranon.2025.102381] [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: 08/11/2024] [Revised: 01/13/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
The low response rate of current immune checkpoint inhibitors in cancer has necessitated the development of new immune targets. Survival and public databases analyses were performed to determine the clinical significance of immunoglobulin-like transcript 3 (ILT3). The impact of ILT3 and apolipoprotein E (APOE) on tumor-associated macrophage (TAM) recruitment and polarization were evaluated by transwell assay, flow cytometry (FCM), and real-time PCR, while their impact on T cell survival and cytotoxicity was detected by CFSE, apoptotic assay, FCM and ELISA. These pro-tumoural activity of (an ortholog of ILT3 in mouse) were verified in vivo models. Survival and public databases analyses revealed that high ILT3 expression was significantly associated with worse prognosis in lung adenocarcinoma (LUAD), but not in squamous cell carcinoma. The same association was observed with its ligand, APOE. In vitro assays demonstrated that tumor-derived ILT3/APOE promoted recruitment and M2-like polarization of TAMs in LUAD and directly inhibited T cell proliferation and cytotoxicity. In vivo knockdown of gp49b enhanced anti-tumor immunity and suppressed tumor progression by counteracting TAM- and dysfunctional T cell-induced tumor microenvironment immunosuppression. Furthermore, combined inhibition of gp49b and programmed cell death ligand 1 (PD-L1) showed the most drastic tumor regression in C57BL/6 mice models. Tumor-derived ILT3 overexpression suppresses anti-tumor immunity by recruiting M2-like TAMs and impairing T cell activities, while ILT3 inhibition counteracts this immunosuppression and enhances the efficacy of PD-L1 blockade in LUAD. Thus, ILT3 could be a promising novel immunotherapeutic target for combined immunotherapy.
Collapse
Affiliation(s)
- Leirong Wang
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China; Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qing Li
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong, China
| | - Yanxin Sun
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shuyun Wang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuebing Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiufen Wang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yan Zheng
- Jinan Center Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Aiqin Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuping Sun
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China; Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Juan Li
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China; Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
24
|
Fontana G, Pepa M, Camarda AM, Strikchani M, Meregaglia M, Vai A, Mirandola A, Vischioni B, Pella A, Baroni G, Jereczek-Fossa BA, Scorsetti M, Cianchetti M, D'Angelo E, Bonomo P, Krengli M, Orlandi E. Envisioning an Italian Head and Neck Proton Therapy Model-Based Selection: Challenge and Opportunity. Int J Part Ther 2025; 16:100745. [PMID: 40230401 PMCID: PMC11995119 DOI: 10.1016/j.ijpt.2025.100745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Affiliation(s)
- Giulia Fontana
- Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Matteo Pepa
- Bioengineering Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Anna Maria Camarda
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Mimoza Strikchani
- Administrative Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Michela Meregaglia
- Center for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Milan, Italy
| | - Alessandro Vai
- Medical Physics Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Alfredo Mirandola
- Medical Physics Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Barbara Vischioni
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Andrea Pella
- Bioengineering Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Guido Baroni
- Bioengineering Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano (POLIMI), Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Marta Scorsetti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marco Cianchetti
- Proton Therapy Unit, Azienda Provinciale per i Servizi Sanitari, Trento, Italy
| | - Elisa D'Angelo
- Radiation Oncology Department, Bellaria Hospital, AUSL of Bologna, Bologna, Italy
| | - Pierluigi Bonomo
- Department of Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Marco Krengli
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
- Radiotherapy Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Ester Orlandi
- Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| |
Collapse
|
25
|
Lee KJ, Ahn JH, Kim JH, Lee YS, Lee JS, Lee JH, Kim TJ, Choi JH. Non-coding RNA RMRP governs RAB31-dependent MMP secretion, enhancing ovarian cancer invasion. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167781. [PMID: 40057205 DOI: 10.1016/j.bbadis.2025.167781] [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: 11/08/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Non-coding RNAs (ncRNAs) are frequently dysregulated in various cancers and have been implicated in the etiology and progression of cancer. Ovarian cancer, the most fatal gynecological cancer, has a poor prognosis and a high patient fatality rate due to metastases. In this study, we classified patients with ovarian cancer into three groups based on their ncRNA expression levels. Notably, an ncRNA transcribed by RNA polymerase III, RNA component of mitochondrial RNA processing endoribonuclease (RMRP), is highly expressed in a group with a poor prognosis. Functional assays using SKOV3 and HeyA8 human ovarian cancer cell lines revealed that while RMRP modulation had no significant effect on cell viability, it markedly enhanced cell invasion. Knockdown and ectopic expression experiments demonstrated that RMRP promotes the secretion of matrix metalloproteinase (MMP)-2 and -9, thereby facilitating ovarian cancer cell invasiveness. Transcriptomic analysis further revealed a positive correlation between RMRP expression and genes involved in cellular localization, including RAB31, a member of the Ras-related protein family. Notably, RAB31 knockdown abrogated the pro-invasive effects of RMRP, identifying it as a key downstream effector in SKOV3 and HeyA8 cells. In addition, MechRNA analysis identified RAB31 as a putative RMRP-interacting transcript. These findings establish RMRP as a critical regulator of RAB31-dependent MMP secretion and ovarian cancer cell invasion. Moreover, our results suggest that RMRP could serve as a promising prognostic biomarker for ovarian cancer.
Collapse
Affiliation(s)
- Ki Jun Lee
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, South Korea; College of Pharmacy, Kyung Hee University, South Korea
| | - Ji-Hye Ahn
- Department of Korean Pharmacy, College of Pharmacy, Woosuk University, South Korea
| | - Jin-Hyung Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, South Korea
| | - Yong Sun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, South Korea
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Jae-Hyung Lee
- Department of Oral Microbiology, College of Dentistry, Kyung Hee University, South Korea
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, South Korea
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, South Korea; College of Pharmacy, Kyung Hee University, South Korea.
| |
Collapse
|
26
|
Ahmed YB, Nan Feng AS, Alrawashdeh M, Ellaithy A, Khanduja S, AlBarakat MM, Alshwayyat S, Uchino K, Gusdon AM, Cho SM. Temporal trends and risk factors associated with stroke mortality among cancer patients. J Clin Neurosci 2025; 136:111249. [PMID: 40252475 DOI: 10.1016/j.jocn.2025.111249] [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: 02/22/2025] [Revised: 03/30/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND This study aimed to explore the risk of stroke death (SD) in cancer patients, estimate rates, and identify risk factors associated with SD. METHODS In this retrospective study, we used the 17 National Cancer Institute Surveillance, Epidemiology, and End Results registries (2000-2020). A total of 5,922,533 patients diagnosed with their first primary cancer were included. The primary outcome was the standardized mortality ratio (SMR) of SD in cancer patients. Secondary outcomes included SD incidence rates and risk factors. Rates were calculated per 100,000 persons with the annual percentage change (APC). RESULTS Among included patients, 56,686 (2.0 %) died due to stroke. Compared to the general population, younger patients (≤39 years) (SMR: 2.31) and patients receiving no treatment (SMR: 1.36) had the highest risk. Cancer types with the fastest-declining SD rates were in the male genital (APC: -13.9 %) and breast (APC: -11.8 %). Older age (hazard ratio [HR]: 1.11, p < 0.001), male sex (HR: 1.06, p < 0.001), and non-white race (HR: 1.13, p < 0.001) were associated with increased risk of SD. Cancers of the nervous system (HR: 3.42, p < 0.001), respiratory (HR: 1.38, p < 0.001), and head and neck (HR: 1.37, p < 0.001) had higher risk of SD vs. breast cancer. Patients with primary chemotherapy (HR: 0.69, p < 0.001) and radiotherapy (HR: 0.69, p < 0.001) demonstrated less risk vs. those without treatment. CONCLUSION SD has declined over the years for both sexes and all cancer types. Older age, non-white race, and certain cancers (nervous system, respiratory system, and head and neck) pose significant risks for SD.
Collapse
Affiliation(s)
- Yaman B Ahmed
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan; Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Division of Neuroscience Critical Care, Departments of Neurosurgery, Anesthesiology, Critical Care Medicine, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Amy Shi Nan Feng
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mohammad Alrawashdeh
- Faculty of Nursing, Community and Mental Health Nursing, Jordan University of Science & Technology, Irbid, Jordan
| | - Asmaa Ellaithy
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Shivalika Khanduja
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Majd M AlBarakat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Sakhr Alshwayyat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ken Uchino
- Cerebrovascular Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Aaron M Gusdon
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sung-Min Cho
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Division of Neuroscience Critical Care, Departments of Neurosurgery, Anesthesiology, Critical Care Medicine, The Johns Hopkins Hospital, Baltimore, MD 21287, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Chao L, Aodeng G, Ga L, Ai J. Design and synthesis of a highly polarity-sensitive fluorescent probe and its application in tumor cell imaging. Bioorg Chem 2025; 159:108399. [PMID: 40158240 DOI: 10.1016/j.bioorg.2025.108399] [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: 02/03/2025] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Among cancer patients, those in advanced stages generally face higher mortality rates, highlighting the importance of early diagnosis in improving cure rates and survival outcomes. Compared to normal cells, tumor cells exhibit a lower polarity in their microenvironment, providing a promising avenue for early cancer detection. It is feasible to distinguish tumor cells from normal cells by leveraging the fluorescence response of probes to polarity. In this study, we designed a fluorescence probe, PCC, with high sensitivity to polarity for early cancer diagnosis. The probe demonstrated a remarkable fluorescence intensity increase of 100-fold in a low-polarity solvent (1,4-dioxane) compared to a high-polarity solvent (water). Additionally, PCC exhibited excellent tumor-targeting ability, large Stokes shift, strong anti-interference capability, and high photostability. When applied to tumor cells (HeLa and CT26、SGC-7901) and normal cells (RAW 264.7、HUVEC、L-02), the probe produced a fluorescence intensity difference exceeding fourfold. These findings indicate that PCC, as a polarity-sensitive fluorescence probe, holds significant promise for early cancer diagnosis.
Collapse
Affiliation(s)
- Lumen Chao
- College of Chemistry and Enviromental Science, Institute of Environment and Health, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China
| | - Gerile Aodeng
- College of Chemistry and Enviromental Science, Institute of Environment and Health, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China
| | - Lu Ga
- College of Pharmacy, Inner Mongolia Medical University, Jinchuankaifaqu, Hohhot 010110, China
| | - Jun Ai
- College of Chemistry and Enviromental Science, Institute of Environment and Health, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China.
| |
Collapse
|
28
|
Jamil A, Qureshi Z, Siddique R, Altaf F, Jamil R, Wali N. A Meta-analysis on Effects of Chimeric Antigen Receptor T-cell Therapy in Relapsed or Refractory B-cell Acute Lymphoblastic Leukemia. Am J Clin Oncol 2025; 48:283-289. [PMID: 39956997 DOI: 10.1097/coc.0000000000001176] [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] [Indexed: 02/18/2025]
Abstract
OBJECTIVES This review evaluates the long-term outcomes and adverse events associated with chimeric antigen receptor (CAR) T-cell therapy in patients with relapsed/refractory B-cell acute lymphoblastic leukemia (r/r B-ALL). METHODS We conducted the search in relevant databases up to June 2024. We included clinical trials on CAR T-cell therapy for patients with r/r B-ALL. Meta-analyses were conducted using Comprehensive Meta-Analysis V3 and Review Manager 5.4. RESULTS Out of 2659 identified studies, 10 were included in this review. The pooled analysis demonstrated a high minimal residual disease-negative complete remission, with an overall event rate (ER) of 70% (95% CI: 61%-78%, I2 =8 8.35%). Anti-CD19 CAR T-cell therapy showed the highest efficacy with an ER of 74.75% (95% CI: 61%-80%, I2 = 89.84%). Combination therapies targeting CD19 and CD22 had an ER of 69% (95% CI: 53%-83%, I2 = 82.56%). Significant adverse effects included cytokine release syndrome with a mean incidence of 81.8% (95% CI: 76.7%-86.9%), neurotoxicity at 33.2% (95% CI: 28.1%-38.3%), and hematologic toxicities at 71.9% (95% CI: 66.4%-77.4%). CONCLUSIONS CAR T-cell therapy is a groundbreaking advancement in treating r/r B-ALL, offering high rates of durable remissions.
Collapse
Affiliation(s)
- Abdur Jamil
- Department of Medicine, Samaritan Medical Centre
| | - Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| | | | - Neehal Wali
- Vituity Hospitalist Group, HSHS St. John's Hospital Springfield, IL
| |
Collapse
|
29
|
Teng F, Zhang R, Wang Y, Li Q, Wang B, Chen H, Liu T, Liu Z, Meng J, Wang C, Dong S, Li Y. Machine Learning and Mendelian Randomization Reveal a Tumor Immune Cell Profile for Predicting Bladder Cancer Risk and Immunotherapy Outcomes. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:1141-1157. [PMID: 40122457 DOI: 10.1016/j.ajpath.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 03/25/2025]
Abstract
This study's objective was to develop predictive models for bladder cancer (BLCA) using tumor infiltrated immune cell (TIIC)-related genes. Multiple RNA expression data and scRNA-seq were downloaded from the TCGA and GEO databases. A tissue specificity index was calculated and a computational framework developed to identify TIIC signature scores based on three algorithms. Univariate Cox analysis was performed, and the TIIC-related model was generated by 20 machine learning algorithms. A significant correlation between TIIC signature score and survival status, tumor stage, and TNM staging system was found. Patients in the high-score BLCA group had more favorable survival outcomes and enhanced response to PD-L1 immunotherapy as compared to those in the low-score group. This TIIC model showed better performance in prognosing BLCA. Diverse frequencies of mutations were observed in human chromosomes across groups categorized by TIIC score. No statistically significant correlation was observed between noncancerous bladder conditions and BLCA when examining the single nucleotide polymorphisms (SNPs) associated with the genes in the prognostic model. However, a statistically significant association was found at the SNP sites of rs3763840. There was no significant association between bladder stones and BLCA, but there was a significant association on the SNP sites of rs3763840. A novel TIIC signature score was constructed for the prognosis and immunotherapy for BLCA, which offers direction for predicting overall survival of patients with BLCA.
Collapse
Affiliation(s)
- Fei Teng
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Renjie Zhang
- Department of Pathophysiology, Guangdong Medical University, Zhanjiang, China
| | - Yunyi Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian Li
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Bei Wang
- Department of Gynecology, Affiliated Hospital of Hebei University, Baoding, China
| | - Huijing Chen
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Tongtong Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Zehua Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Jia Meng
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Ce Wang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Shilei Dong
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China.
| | - Yanhong Li
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, China.
| |
Collapse
|
30
|
Komatsu S, Nagamata S, Terashima K, Demizu Y, Suga M, Kido M, Yanagimoto H, Toyama H, Tokumaru S, Okimoto T, Terai Y, Fukumoto T. Combination Treatment with Spacer Placement Surgery Followed by Particle Radiotherapy for Lymph Node Metastasis from Uterine Cancer. Ann Surg Oncol 2025; 32:4313-4321. [PMID: 40000562 PMCID: PMC12049338 DOI: 10.1245/s10434-025-17039-9] [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: 11/12/2024] [Accepted: 02/04/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The effectiveness of local treatment in lymph node metastasis from uterine cancer has been proven; the standard treatment is surgical intervention. Although radiotherapy, including particle radiotherapy (PRT), is an alternative local treatment, its application is often contraindicated owing to its proximity to the gastrointestinal tract. Combination treatment with spacer placement surgery followed by PRT is a potential solution to this problem. This study aimed to evaluate the outcomes of this combination treatment of lymph node metastases from uterine cancer. PATIENTS AND METHODS Between December 2007 and March 2023, ten consecutive patients who underwent combination treatment comprising spacer placement surgery and subsequent PRT were assessed for treatment outcomes. RESULTS The median survival time was 53.5 months; the 3- and 5-year overall survival rates were 76.2% and 38.1%, respectively. The 3- and 5-year local control rates in all patients were both 88.9%. The median volume irradiated at 95% of the treatment planning dose (V95%) of the gross tumor volume, clinical target volume, and planning target volume were 100.0%, 99.8%, and 92.2%, respectively. The median dose intensity covering 95% of the target volume (D95%) of the gross tumor volume/planned dose, clinical target volume/planned dose, and planning target volume/planned dose were 98.9%, 99.0%, and 87.2%, respectively. CONCLUSIONS Spacer placement surgery contributed to the optimized PRT dose distribution and might have contributed to favorable local control and survival rates. This innovative combination treatment might have a significant effect on the treatment of lymph node metastases from uterine cancers.
Collapse
Affiliation(s)
- Shohei Komatsu
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Satoshi Nagamata
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazuki Terashima
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Yusuke Demizu
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
- Department of Radiation Oncology, Hyogo Ion Beam Medical Center Kobe Proton Center, Kobe, Japan
| | - Masaki Suga
- Department of Radiation Physics, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Masahiro Kido
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroaki Yanagimoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hirochika Toyama
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sunao Tokumaru
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Tomoaki Okimoto
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Fukumoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
31
|
Akkus E, Tuncalı T, Beksac M. Familial Multiple Myeloma: Insights From Epidemiology and Underlying Germline Genetic Predisposition to the Clinic. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e411-e416. [PMID: 39947961 DOI: 10.1016/j.clml.2025.01.011] [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: 11/07/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 05/23/2025]
Abstract
Familial multiple myeloma (MM) is a relatively rare entity. Data on epidemiology, underlying germline genetic predisposition, and clinical features of familial MM continue to expand, especially in recent years. The risk of MM has been reported to be increased by 2 to 4 times among first-degree relatives of MM patients along with monoclonal gammopathy of undetermined significance (MGUS), other hematological malignancies, and several solid tumors by multiple groups. The association between MM risk and having a first-degree relative with a history of MM is stronger among African Americans/Blacks (OR = 5.52, 95% CI: 1.87-16.28) than European Americans/Whites (OR = 1.26, 95% CI: 0.75-2.10). Although data on clinical features and prognosis of familial cases are limited, preliminary data suggest a better prognosis among familial cases. Sequencing studies in familial cases have discovered rare disease-causing variants among which CDKN2A, USP45, MYH14, EPOR, HERC1, KLHL18, and KLHDC3 are shared by independent groups. Some of these candidate genes possess functions related to myelomagenesis such as B-cell development, immunoglobulin gene regulation, immune pathways, DNA damage/repair, ubiquitination, and cell adhesion mechanisms. Our group has recently completed an analysis among our Turkish MM families to confirm the findings of the International Consortium. The odds ratio (OR) for single nucleotide polymorphism rs28199 in nonfamilial MM cases compared to healthy control was 1.18, with an OR of 2.86 in familial MM cases compared to nonfamilial cases. Despite the accumulation of data, most questions remain to be answered. Future studies may elucidate underlying biological mechanisms, and develop polygenic risk scores, leading advancements in prognostic evaluation and clinical management.
Collapse
Affiliation(s)
- Erman Akkus
- Faculty of Medicine, Department of Medical Oncology, Ankara University, Ankara, Türkiye
| | - Timur Tuncalı
- Faculty of Medicine, Department of Medical Genetics, Ankara University, Ankara, Türkiye
| | - Meral Beksac
- Department of Hematology, Ankara Liv Hospital, Istinye University, Ankara, Türkiye.
| |
Collapse
|
32
|
Shah N, Patkar S, Varty G, Gundavda K, Chaubal G, Goel M. A Multimodality Treatment Strategy for an Intrahepatic Cholangiocarcinoma and Complex Liver Resection with Vascular Reconstruction-A Video Vignette. Ann Surg Oncol 2025:10.1245/s10434-025-17282-0. [PMID: 40450171 DOI: 10.1245/s10434-025-17282-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/24/2025] [Indexed: 06/03/2025]
Abstract
INTRODUCTION Radical resection remains the only curative option for intrahepatic cholangiocarcinoma (iCCA).1-3 Patients with multifocal tumors, vascular invasion, or lymph node metastasis are considered to have locally advanced disease.4 Neoadjuvant chemotherapy with gemcitabine and cisplatin as first line or with capecitabine and irinotecan as second line have shown oncological benefit.5-8 More recently, Durvalumab has also showed benefit in addition to chemotherapy.9 For tumours with major vascular invasion, stereotactic body radiation therapy (SBRT) is a safe treatment option where upfront surgery may not provide the desired oncological outcomes.10-13 We present a video where combination of modalities was used to achieve downstaging followed by surgery. CASE A 64-year-old male patient was diagnosed with locally advanced iCCA involving left lobe with left portal vein thrombosis, left hepatic vein (LHV) encasement, and middle hepatic vein (MHV) abutment. Cancer antigen (CA) 19-9 was 1890 U/mL. After three cycles of gemcitabine and cisplatin, response computed tomography (CT) scan showed disease progression, and second-line chemotherapy with immunotherapy (capecitabine, irinotecan, durvalumab) was started with stereotactic body radiation therapy. Response CT showed significant size reduction. CA 19-9 dropped to 17 U/mL. Patient was planned for left hepatectomy with portal vein resection (PVR) and segment 8 venous reconstruction. STEPS OF SURGERY 1. Kocherization, inter-aortocaval nodal sampling, and peri-portal dissection. 2. Parenchymal transection with left hepatic duct division, PVR with primary repair 3. Isolation of segment 8 vein, division distal to MHV-LHV confluence, specimen retrieval. 4. Reconstruction of segment 8 vein to MHV-LHV confluence using polytetrafluoroethylene graft (PTFE). Final histopathology report revealed complete pathological response. Patient was planned for two more cycles of same chemotherapy followed by observation. CONCLUSION Resection with periportal lymphadenectomy and negative margins remains the only curative option for iCCA. Multimodality options can be utilized to downstage tumor, and complex liver resection with vascular reconstruction can achieve excellent oncological outcomes.
Collapse
Affiliation(s)
- Niket Shah
- Department of Gastrointestinal and Hepatobiliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Shraddha Patkar
- Department of Gastrointestinal and Hepatobiliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Gurudatt Varty
- Department of Gastrointestinal and Hepatobiliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Kaival Gundavda
- Department of Gastrointestinal and Hepatobiliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | | | - Mahesh Goel
- Department of Gastrointestinal and Hepatobiliary Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| |
Collapse
|
33
|
Cao Z, Pu C, Jiang X, Han G, Shen X, Wang W, Ding W, Huang Z, Huang X, Jia B, Lu VX, Tian L, Wu Z, Xiao L. Novel PAP-targeted CAR-T therapy enhances antitumor efficacy through CoupledCAR approach. J Immunother Cancer 2025; 13:e011238. [PMID: 40449956 DOI: 10.1136/jitc-2024-011238] [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] [Accepted: 05/04/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND The challenges that remain in the treatment of solid tumors with chimeric antigen receptor (CAR)-T cells include limited solid tumor-specific targets and poor CAR-T cell expansion and function due to limited availability of solid tumor antigens outside the tumor microenvironment. Prostate cancer is the second most common cancer among men worldwide. Current CAR-T therapies for prostate cancer lack specific targets, posing safety risks. To overcome these problems, we identified prostatic acid phosphatase (PAP, also known as ACPP or ACP3) as a feasible CAR-T target for prostate cancer and developed CoupledCAR, a novel approach for expanding tumor-targeting CAR-T cells without tumor antigens. METHODS We analyzed the expression of PAP from The Cancer Genome Atlas database and validated its expression in normal and cancer tissues through immunohistochemistry staining. To generate anti-PAP specific antibodies, we screened the human single-chain antibody library using transmembrane PAP-His antigen and selected antibodies based on their binding ability and specificity. We constructed PAP-targeted CAR and evaluated their antitumor efficacy both in vitro and in vivo. We validated the function of PAP CoupledCAR in both in vitro and in vivo experiments, and further analyzed its mechanism using single-cell RNA sequencing (scRNA-Seq). RESULTS PAP was specifically expressed in prostate epithelial and prostate cancer cells, with no expression in other tissues. Seven single-chain variable fragments were screened from the human single-chain antibody library, with S5D1 showing the highest binding ability for the PAP. PAP CAR-T cells demonstrated strong antitumor efficacy both in vitro and in vivo. Furthermore, the CoupledCAR system significantly expanded PAP CAR-T cells, promoting memory-like status, reducing exhaustion, and enhancing their antitumor efficacy. The scRNA-Seq demonstrated that the expansion of PAP CAR-T cells in the CoupledCAR system is mediated by costimulatory signals and cytokine signals, rather than T-cell receptor signals. CONCLUSIONS Our study is the first to demonstrate that PAP is a specific target for CAR-T therapy in prostate cancer, both in vitro and in vivo. We developed the CoupledCAR platform technology for solid tumor CAR-T cell therapy, enabling the expansion of tumor-targeting CAR-T cells without requiring tumor antigens and thereby enhancing their functionality against solid tumors.
Collapse
Affiliation(s)
- Zhiyuan Cao
- Innovative Cellular Therapeutics, Shanghai, China
| | - Chengfei Pu
- Innovative Cellular Therapeutics, Shanghai, China
| | | | - Guiting Han
- Innovative Cellular Therapeutics, Shanghai, China
| | | | | | - Wei Ding
- Innovative Cellular Therapeutics, Shanghai, China
| | | | - Xi Huang
- Innovative Cellular Therapeutics, Shanghai, China
| | - Beibei Jia
- Innovative Cellular Therapeutics, Shanghai, China
| | - Victor X Lu
- Innovative Cellular Therapeutics Holdings Limited, Rockville, Maryland, USA
| | - Le Tian
- Innovative Cellular Therapeutics Holdings Limited, Rockville, Maryland, USA
| | - Zhao Wu
- Innovative Cellular Therapeutics, Shanghai, China
| | - Lei Xiao
- Innovative Cellular Therapeutics Holdings Limited, Rockville, Maryland, USA
| |
Collapse
|
34
|
Mo X, He J, Shen X, Li C, Mo X, Liang K, He L, Li T, Pan X, Cao S, Mao N, Xing S, Chen Z, Luo Z, Yang J. Connexin43 Promotes the Invasion and Metastasis of Lung Squamous Cell Carcinoma via GJIC-Dependent Ca 2+/ERK Signaling Activation. Cancer Sci 2025. [PMID: 40449992 DOI: 10.1111/cas.70076] [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: 11/15/2024] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 06/03/2025] Open
Abstract
Lung squamous cell carcinoma (LUSC) is an extremely metastatic cancer with limited available treatment and poor outcomes. Connexin43 (Cx43) is frequently overactivated and positively correlated with tumorigenesis in many cancers, including breast cancer and lung adenocarcinoma, but its role in LUSC remains elusive. In this study, we demonstrated that Cx43 was highly expressed in LUSC tissues as compared to matching normal lung tissues (n = 103) and negatively related to prognosis. Through the 3D spheroid cell invasion assay, zCDX (zebrafish cell line-derived xenograft), and orthotopic lung cancer xenograft model, we further revealed that Cx43 promotes LUSC invasion and migration via forming GJIC. Knockdown of Cx43 reduced the Ca2+ transmission and ERK phosphorylation, whereas the addition of Ca2+ enhanced ERK phosphorylation and promoted LUSC invasion and migration. Furthermore, verapamil (40 μM and 80 μM), a calcium channel inhibitor, significantly inhibited ERK phosphorylation as well as the invasion and migration of LUSC cells. Mechanistically, Cx43 promoted the invasion and metastasis of LUSC via activating the Ca2+/ERK signaling pathway by gap junctional intracellular communication (GJIC). Our findings provide a novel mechanism insight for LUSC invasion and migration and a proof of concept for a new therapeutic strategy to tackle this disease.
Collapse
Affiliation(s)
- Xiaocheng Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingchuan He
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
- Department of Pharmacy, The Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xiaoju Shen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Changsheng Li
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xiaoxiang Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Kai Liang
- Department of Thoracic Tumor Surgery, The Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Liangjun He
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Tingting Li
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xiaoqin Pan
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Sisi Cao
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Naiquan Mao
- Department of Thoracic Tumor Surgery, The Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Shangping Xing
- Department of Chinese Materia Medica, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Zhiquan Chen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Zhuo Luo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jie Yang
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
35
|
Lu X, Pan C, Yao L, Wan J, Xu X, Wang W, Wang X, Liu X, Jin Z, Wang H, He Y, Yang B. Integrating multimodal data to predict the progression of hormone-sensitive prostate cancer. Clin Proteomics 2025; 22:21. [PMID: 40442579 PMCID: PMC12121097 DOI: 10.1186/s12014-025-09543-7] [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: 11/28/2024] [Accepted: 05/11/2025] [Indexed: 06/02/2025] Open
Abstract
Identifying the population at risk of rapid progression from hormone-sensitive prostate cancer (HSPC) to lethal castration-resistant prostate cancer (CRPC) is a challenge. This work has highlighted important prognostic insights based on proteomics data, magnetic resonance imaging (MRI) and histopathological specimens. We retrospectively developed a multi-omics-based model based on 77 patients with HSPC. In order to identify the features related to survival time under each mode, we used the Boruta algorithm for feature screening. In order to demonstrate the effectiveness of our selected features, we used six machine learning methods to validate the classification of the selected features for each mode. A total of 63 proteome signatures, 60 HE signatures, 56 T2WI signatures, and 54 ADC signatures were identified as features related to the speed of HSPC progression. Ultimately, 30 multi-omics-based features were determined by the least absolute shrinkage and selection operator (LASSO) method and multivariate cox regression. In order to stratify patients with significant disparities in progress, a nomogram model was developed, of which the C-index was 0.906. Accordingly, the developed model could help identify patients who are at a high risk of rapid CRPC progression, and aid clinicians in guiding personalized clinical management and decision-making.
Collapse
Affiliation(s)
- Xiangfu Lu
- Department of Urology, 967 th hospital of PLA Joint Logistics Support Force, No.80 Shengli Road, Dalian, 116014, PR China
| | - Chenxi Pan
- State key laboratory of fine chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, PR China
| | - Luhan Yao
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, Dalian, 116023, PR China
| | - Jiayu Wan
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, Dalian, 116023, PR China
| | - Xiaolong Xu
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Wei Wang
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Xiangying Wang
- State key laboratory of fine chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, PR China
| | - Xiaoyun Liu
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Zhonghua Jin
- Department of chest surgery, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Hongyu Wang
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, Dalian, 116023, PR China.
| | - Yi He
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China.
- The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, Liaoning, China.
| | - Bo Yang
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China.
- The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, Liaoning, China.
| |
Collapse
|
36
|
Ma T, Wang H, Dai W, Shen P, Zhang J, Xie R. Historical trends in histological composition and cause specific mortality of small intestine tumors based on SEER database analysis. Sci Rep 2025; 15:18628. [PMID: 40436948 PMCID: PMC12120026 DOI: 10.1038/s41598-025-03046-z] [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: 07/07/2024] [Accepted: 05/19/2025] [Indexed: 06/01/2025] Open
Abstract
Small intestine tumors, though rare, have shown a concerning increase in incidence and mortality in recent years. This study aimed to investigate the historical trends in histological composition and causes of death among patients with small intestine tumors using the SEER database. A total of 18,234 patients diagnosed with primary small intestine cancer from 1992 to 2018 were included in this study. Demographic details, tumor characteristics, treatment modalities, and outcomes were collected. Cause-specific mortality was analyzed using Fine and Gray regression, with non-small intestine-specific deaths considered as competing risks. Small intestine-specific deaths were the leading cause of mortality, with adenocarcinoma and carcinoid tumors being the most common histological types. Heart disease emerged as a significant cause of death following diagnosis, surpassing small intestine-specific deaths after 5-6 years. The study revealed variations in mortality causes across histological subtypes and identified risk factors for small intestine-specific mortality, including age, tumor site, grade, and treatment modalities. The findings highlight the substantial impact of heart disease on the long-term survival of patients with small intestine tumors. This underscores the potential benefits of adopting comprehensive management strategies that integrate oncological and cardiological care to improve survival rates and quality of life. A multidisciplinary approach in oncocardiology may help address the complex needs of these patients and optimize clinical outcomes.
Collapse
Affiliation(s)
- Tianheng Ma
- Department of Gastroenterology, The Affiliated Huai'an No 1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Honggang Wang
- Department of Gastroenterology, The Affiliated Huai'an No 1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Weijie Dai
- Department of Gastroenterology, The Affiliated Huai'an No 1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Peng Shen
- Department of Gastroenterology, The Affiliated Huai'an No 1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huai'an, 223300, Jiangsu, China
| | - Jialing Zhang
- Department of Gastroenterology, The Affiliated Huai'an No 1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huai'an, 223300, Jiangsu, China.
| | - Rui Xie
- Department of Gastroenterology, The Affiliated Huai'an No 1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huai'an, 223300, Jiangsu, China.
| |
Collapse
|
37
|
Jeong JH, Shin D, Kim SY, Bae DJ, Sung YH, Koh EY, Kim J, Kim CJ, Park JS, Choi JK, Kim SC, Jun E. Spatial distribution and activation changes of T cells in pancreatic tumors according to KRAS mutation subtype. Cancer Lett 2025; 618:217641. [PMID: 40090570 DOI: 10.1016/j.canlet.2025.217641] [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: 12/03/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
To enhance immunotherapy efficacy in pancreatic cancer, it is crucial to characterize its immune landscape and identify key factors driving immune alterations. To achieve this, we quantitatively analyzed the immune microenvironment using multiplex immunohistochemistry, assessing the spatial relationships between immune and tumor cells to correlate with patient survival rates and oncological factors. Additionally, through Whole Exome Sequencing analysis based on public data, we explored genetic mutations that could drive these compositions. Finally, we validated T cell (Tc) migration mechanisms using patient-derived tumor organoids with induced KRAS mutation subtypes. Through this approach, we obtained the following meaningful results. First, immune cells in pancreatic cancer are denser in stromal regions than near tumor cells, with higher Tc distribution linked to increased patient survival rates. Second, the distance between tumor and Tc was within 100 μm, with higher Tc density found within 15-30 μm of the tumor cells. Third, while increasing CAF levels correspond to higher Tc density, higher ECM density tends to decrease Tc presence. Fourth, compared to KRAS G12D, KRAS G12V mutation increases various immune cells, notably Tc, which is closely linked to a dramatic rise in vascular cells. Finally, Tc migration was enhanced in tumor organoids with the G12V mutation, attributed to a reduction in the secretion of immunosuppressive cytokines. Our results indicate that KRAS mutation subtypes influence immune cell composition and function in the pancreatic cancer microenvironment, leading to varied immunotherapy responses. This underscores the need for personalized immune therapeutics and research models specific to KRAS mutations.
Collapse
Affiliation(s)
- Ji Hye Jeong
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Dakyum Shin
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation Surgery, Department of General Surgery, Chosun University Hospital, 365, Pilmun-daero, Dong-gu, Gwangju Metropolitan City, 61453, Republic of Korea
| | - Sang-Yeob Kim
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Dong-Jun Bae
- PrismCDX, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Young Hoon Sung
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Cell and Genetic Engineering, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Eun-Young Koh
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinju Kim
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Chong Jai Kim
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jae Soon Park
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea; SCL-KAIST Institute of Translational Research, Daejeon, 34141, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea; SCL-KAIST Institute of Translational Research, Daejeon, 34141, Republic of Korea.
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Surgery, BK21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Eunsung Jun
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul, 05505, Republic of Korea; Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea.
| |
Collapse
|
38
|
Afghani E, Lau B, Becker LK, Goggins M, Klein AP. Impact of race, sex and age on the probability of pancreatic cancer among patients with newly diagnosed diabetes in a claims-based cohort. BMJ Open 2025; 15:e099488. [PMID: 40425248 PMCID: PMC12107564 DOI: 10.1136/bmjopen-2025-099488] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
OBJECTIVE Pancreatic cancer diagnoses are frequently preceded by a new diabetes diagnosis. Screening individuals newly diagnosed with diabetes could enable the earlier detection of pancreatic cancer. We sought to estimate the risk of pancreatic cancer by age, sex, race and time since diabetes diagnosis. DESIGN Claims-based cohort study. SETTING Johns Hopkins Medicine conducted this deidentified claims-based cohort study using the Optum Labs Data Warehouse. PARTICIPANTS Insurance enrollees from 1/2008-9/2018 were identified as non-diabetic or newly diagnosed diabetic. Our risk set included 4 732 313 individuals (424 129 newly diabetic) in 5 844 934 enrolment periods. PRIMARY OUTCOME MEASURES Time to pancreatic cancer. Diabetes and cancer were defined using International Classification of Diseases (ICD)-9/10 codes. RESULTS Individuals with newly diagnosed diabetes were at an increased HR of pancreatic cancer, but this effect waned over time. The HR of pancreatic cancer following a diabetes diagnosis was higher in younger individuals and varied by race (lower HR in non-White individuals) (p<0.01, main effects and interactions). Thus, the probability of pancreatic cancer following a diabetes diagnosis was dependent on age, race and sex. For example, the 1-year probability of pancreatic cancer in a White male aged 75 was 0.45% (95%CI 0.41% to 0.49%) if they were newly diagnosed with diabetes and 0.090% (95%CI 0.084% to 0.096%) if they were free of diabetes. In contrast, the risk was lower at 0.15% (new-diabetic, 95% CI 0.13% to 0.16%) and 0.022% (diabetes free, 95%CI 0.020% to 0.023%) at age 55. The HR of pancreatic cancer for individuals with newly diagnosed diabetes compared with those free of diabetes was highest in the month following diagnosis (HR=14.7 and 9.6 for a 55 and 75 year old White male, respectively) but decreased in the following months, with a HR of 7.8 and 5.8 at 3 months, 5.6 and 4.1 at 6 months, and 3.9 and 2.8 at 1 year (p<0.01). CONCLUSIONS Consideration of the age-race-sex specific probability of pancreatic cancer and time since diabetes diagnosis is necessary when evaluating the risk of pancreatic cancer following a diabetes diagnosis.
Collapse
Affiliation(s)
- Elham Afghani
- Division of Gastroenterology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Bryan Lau
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | | | - Michael Goggins
- Division of Gastroenterology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alison P Klein
- Division of Gastroenterology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
39
|
Wei X, Zou L, Huang Y, Qiu C, Cheng G, Chen Y, Rao J. LDHA-mediated YAP lactylation promotes the tumor progression of hepatocellular carcinoma by inducing YAP dephosphorylation and activation. Biol Direct 2025; 20:64. [PMID: 40414964 DOI: 10.1186/s13062-025-00655-6] [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: 01/22/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is among the deadliest cancers globally. Yes-Associated Protein (YAP), a Hippo pathway effector, crucially regulates cell proliferation and apoptosis. Recent research has implicated YAP's role in HCC progression, but the mechanisms are unclear. This study aims to clarify YAP's function in HCC, emphasizing its regulation of key pathways and targets. RESULTS Gene knockout and overexpression models were established in nude mice and cell lines of HCC cells to investigate YAP's impact on tumorigenesis. Additionally, functional assays and molecular biology techniques were employed to identify YAP's regulatory networks. The study demonstrates that LDHA-regulated lactate production promotes YAP activation and malignant phenotypes in HCC. Overexpression of LDHA in HepG2 and Huh7 cells increased lactate levels and activated the YAP pathway, enhancing cell proliferation, migration, and invasion. Lactate treatment also promoted these malignant phenotypes by inhibiting YAP phosphorylation at Ser127. In a xenograft model, lactate accelerated tumor growth through YAP activation. YAP lactylation at K102 antagonized its Ser127 phosphorylation, further promoting malignant progression. CONCLUSIONS This study highlights the significance of YAP in HCC pathogenesis, providing insights into potential therapeutic targets for HCC management.
Collapse
Affiliation(s)
- Xiaoyong Wei
- Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, 330029, China
| | - Long Zou
- Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, 330029, China
| | - Yanqing Huang
- The Medical College of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Chuan Qiu
- The Medical College of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Guang Cheng
- Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, 330029, China
| | - Ye Chen
- Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, 330029, China
| | - Jun Rao
- Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, 330029, China.
| |
Collapse
|
40
|
Zhong X, Han J, Li H, Shen X, Yu B, Chen T, Li H, Li J, Pang J, Qian L, Wu W, Tong X, Ding B. Glycosylated protein-related microenvironmental features in breast cancer are associated with patient prognosis. Mamm Genome 2025:10.1007/s00335-025-10137-9. [PMID: 40411577 DOI: 10.1007/s00335-025-10137-9] [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: 03/14/2025] [Accepted: 05/10/2025] [Indexed: 05/26/2025]
Abstract
The tumor microenvironment (TME) and aberrant glycosylation have been suggested to play key roles in cancer. This study integrated differentially expressed genes (DEGs) and weighted gene coexpression network analysis (WGCNA) to identify tumor microenvironment-related genes and construct a TME-risk prognostic signature (TMERS) through LASSO Cox regression. After batch effect removal, 44 TME-prognosis-related genes (TMEPGs) were identified and classified into three molecular subtypes via K-means clustering. The finalized 22-gene TMERS model demonstrated robust prognostic predictive capacity in GEO datasets. The results revealed distinct immune profiles and prognostic stratifications among genetic subtypes and risk groups, confirming that the TMERS is an independent prognostic indicator for breast cancer (BRCA). Glycosyltransferase genes (GTs) have potential therapeutic relevance through immune regulation, with TMEPG member killer cell lectin like receptor B1 (KLRB1) significantly correlated with BRCA prognosis. Cellular experiments demonstrated that KLRB1 overexpression suppressed BRCA cell proliferation and migration. This work establishes a novel prognostic model for BRCA while highlighting KLRB1 as a potential biomarker, providing new insights into TME-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxiao Zhong
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| | - Jiaxuan Han
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiangyu Shen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Bowen Yu
- Department of Gastrointestinal Surgery, Third XiangYa Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Ting Chen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Haobing Li
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, China
| | - Jun Li
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Jin Pang
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Liyuan Qian
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Wei Wu
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiaoliang Tong
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, 410000, China.
| | - Boni Ding
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| |
Collapse
|
41
|
Wang L, Yang Z, Xing S, Zhao S, Ouyang M, Yu H. miR-1246 enhances chemo-resistance of polyploid giant cancer cells in H1299 cells by targeting GSK3β/β-catenin. Discov Oncol 2025; 16:901. [PMID: 40411667 PMCID: PMC12103447 DOI: 10.1007/s12672-025-02756-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 05/19/2025] [Indexed: 05/26/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is characterized by a high mortality rate. Chemotherapy has been observed to potentially increase the prevalence of polyploid giant cancer cells (PGCCs), which may play a role in the development of chemo-resistance in NSCLC. The dysregulated expression of miR-1246 has been implicated in the modulation of gene expression related to drug resistance. Therefore, the objective of this study is to examine the role of miRNA-1246 in PGCCs and to elucidate its regulatory mechanisms. H1299 cells were treated with 100 nM docetaxel (Doc) for 24 h, then allowed to recover for 3 days to form polyploid giant cancer cells (PGCCs). The miRNA profiles of these PGCCs were analyzed, focusing on miR-1246. Transfection with miR-1246 mimics or inhibitors was performed, and various assays were used to assess the effects of miR-1246 inn PGCCs. The study found miR-1246 levels were significantly higher in PGCCs than in the original cells, affecting chemo-resistance, apoptosis, migration, and epithelial-mesenchymal transition. These findings suggested that NSCLC H1299 cells may employ polyploidy formation as a survival mechanism in response to docetaxel-based treatment, mediated by the miR-1246/GSK3β/β-catenin axis, ultimately leading to enhanced chemo-resistance.
Collapse
Affiliation(s)
- Lili Wang
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, Shenyang, 110016, Liaoning, China
| | - Zien Yang
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, Shenyang, 110016, Liaoning, China
| | - Sining Xing
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, Shenyang, 110016, Liaoning, China
| | - Song Zhao
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, Shenyang, 110016, Liaoning, China
| | - Mingyue Ouyang
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, Shenyang, 110016, Liaoning, China
| | - Huiying Yu
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
42
|
Tao X, Ke X, Xu G. *Mechanisms of circular RNA in drug resistance of lung cancer: therapeutic targets, biomarkers, and future research directions. Discov Oncol 2025; 16:896. [PMID: 40410444 PMCID: PMC12102044 DOI: 10.1007/s12672-025-02713-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 05/14/2025] [Indexed: 05/25/2025] Open
Abstract
Lung cancer is one of the most prevalent malignant tumors globally, posing significant challenges to treatment outcomes. Circular RNAs (circRNAs), a novel class of non-coding RNAs, have emerged as crucial regulators in cancer biology, influencing drug resistance, progression, and prognosis. Due to their closed-loop structure, circRNAs demonstrate high stability and resistance to degradation, making them promising diagnostic and therapeutic targets. Here we summarize the mechanisms by which circRNAs mediate drug resistance in lung cancer, focusing on their roles in chemotherapy, targeted therapies, and immunotherapy. We highlight how circRNAs interact with microRNAs (miRNAs) and proteins to regulate signaling pathways and alter drug sensitivity. Additionally, circRNA expression patterns hold potential as biomarkers for predicting treatment response. By synthesizing the latest research, we offer new insights into circRNA functions and suggest future directions for overcoming drug resistance in lung cancer.
Collapse
Affiliation(s)
- Xuanlin Tao
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Zunyi, 563000, Guizhou, China
| | - Xixian Ke
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Zunyi, 563000, Guizhou, China.
| | - Gang Xu
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
43
|
Li W, Shi Y, Chen X, Wang H, Wei D, Yao J, Li X, Lu J, Li X, Chang J, Qiao Y. TCPTP inhibition as a novel therapeutic strategy for esophageal squamous cell carcinoma: discovery and efficacy of COH29. Biochem Pharmacol 2025; 239:116997. [PMID: 40414512 DOI: 10.1016/j.bcp.2025.116997] [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: 08/26/2024] [Revised: 04/17/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a malignant tumor that poses a serious threat to human health and is often associated with poor prognosis. Therefore, it is urgent to explore new therapeutic strategies to improve the survival rate of patients with ESCC. T cell protein tyrosine phosphatase (TCPTP) has been reported as a complicated factor in cancer. In this study, we found that TCPTP was highly expressed in ESCC tissues and suppression of TCPTP can effectively inhibit the proliferation of ESCC cells in vitro and in vivo. To identify potential TCPTP inhibitors, we employed a comprehensive research approach encompassing virtual screening, pull down assay, and cellular thermal shift assay. This led to the discovery of two promising candidates: COH29 and gallocatechin gallate (GCG). Both compounds showed inhibitory effects on ESCC cell proliferation, with COH29 displaying superior efficacy. Further enzyme kinetics assay and molecular dynamics simulations confirmed COH29's unique ability to bind to both the substrate and allosteric sites of TCPTP, making it a promising lead compound for future inhibitor development. Flow cytometry analysis revealed that COH29 treatment caused cell cycle arrest in the G1 phase in ESCC cells. In vivo studies further validated COH29's robust growth suppression of ESCC, highlighting its potential as a therapeutic agent.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yaqian Shi
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Pathology, The Ninth Hospital of Xi'an, Xi'an, Shaanxi 710054, China
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Huizhen Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Donghui Wei
- The College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jing Yao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xin Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junbiao Chang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
44
|
Valenti G, Laise P, Wu F, Takahashi R, Ruan T, Vasciaveo A, Jiang Z, Kobayashi H, Sunagawa M, Middelhoff M, Nienhüser H, Fu N, Malagola E, Companioni O, Hayakawa Y, Iuga AC, Califano A, Wang TC. Regulatory network analysis of Dclk1 gene expression reveals a tuft cell-ILC2 axis that inhibits pancreatic tumor progression. Cell Rep 2025; 44:115734. [PMID: 40408246 DOI: 10.1016/j.celrep.2025.115734] [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: 11/14/2024] [Revised: 01/21/2025] [Accepted: 05/02/2025] [Indexed: 05/25/2025] Open
Abstract
Doublecortin-like kinase 1 (Dclk1) expression identifies cells that are rare in normal pancreas but occur with an increased frequency in pancreatic neoplasia. The identity of these cells has been a matter of debate. We employed Dclk1 reporter mouse models and single-cell RNA sequencing (scRNA-seq) to define Dclk1-expressing cells. In normal pancreas, Dclk1 identifies subsets of ductal, islet, and acinar cells. In pancreatic neoplasia, Dclk1 identifies several cell populations, among which acinar-to-ductal metaplasia (ADM)-like cells and tuft-like cells are predominant. These two populations play opposing roles, with Dclk1+ ADM-like cells sustaining and Dclk1+ tuft-like cells restraining tumor progression. The generation of Dclk1+ tuft-like cells requires the transcription factor SPIB and is sustained by a paracrine loop involving type 2 innate lymphoid cells (ILC2s) and cancer-associated fibroblasts (CAFs) that provide interleukin (IL)-13 and IL-33, respectively. Dclk1+ tuft-like cells release angiotensinogen to restrain tumor progression. Overall, our study defines pancreatic Dclk1+ cells and unveils a protective tuft cell-ILC2 axis against pancreatic neoplasia.
Collapse
Affiliation(s)
- Giovanni Valenti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Pasquale Laise
- Department of Systems Biology, Columbia University, New York, NY, USA; DarwinHealth, Inc., New York, NY, USA
| | - Feijing Wu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Ryota Takahashi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Tuo Ruan
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | | | - Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Hiroki Kobayashi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Masaki Sunagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Moritz Middelhoff
- Klinik und Poliklinik für Innere Medizin II, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Na Fu
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Osmel Companioni
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA
| | - Yoku Hayakawa
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Alina C Iuga
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA; DarwinHealth, Inc., New York, NY, USA; Chan Zuckerberg Biohub New York, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
45
|
Shen W, Ma Y, Yang C, Yan S, Ye K. Role of N6-methyladenosine methyltransferase component RBM15 in cancer progression and its therapeutic potential. Discov Oncol 2025; 16:855. [PMID: 40402374 PMCID: PMC12098234 DOI: 10.1007/s12672-025-02644-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025] Open
Abstract
Cancer ranks as a primary cause of mortality globally, and the study of its molecular markers and regulatory mechanisms holds paramount importance. N6-methyladenosine (m⁶A) represents the predominant modification in messenger RNA (mRNA), influencing key biological processes including RNA stability, splicing, and translation. The dynamic modulation of m⁶A modification is mediated by an array of enzymes comprising methyltransferases ("writers"), demethylases ("erasers"), and m⁶A-binding proteins ("readers").As a pivotal member of the m⁶A "writer" family, RNA binding motif protein 15 (RBM15) facilitates the recruitment of the methyltransferase complex (MTC) to mRNA, thus orchestrating the addition of m⁶A modifications. Although prior research has underscored the critical role of m⁶A in oncogenesis, the precise mechanisms through which RBM15 operates in cancer are yet to be elucidated. This study endeavors to elucidate the structural characteristics and functional roles of RBM15, investigate its potential regulatory mechanisms across diverse tumors, uncover its distinct functions in tumor genesis, progression, and metastasis, and evaluate the therapeutic potential of targeting RBM15 in cancer treatment.
Collapse
Affiliation(s)
- Wenxiang Shen
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yulong Ma
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Chunwang Yang
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Shishun Yan
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Kaishan Ye
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
46
|
Zou J, Kinosada H, Takayanagi SI, Ishii T, Amano T, Nihira K, Kanie S, Adachi M, Tahara H, Sakoda T, Kikushige Y, Akashi K, Satou H. KK2845, a PBD dimer-containing antibody-drug conjugate targeting TIM-3-expressing AML. Leukemia 2025:10.1038/s41375-025-02642-2. [PMID: 40404985 DOI: 10.1038/s41375-025-02642-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025]
Abstract
Acute myeloid leukemia (AML) is a common hematopoietic malignancy with high recurrence rates, and there is an urgent need for new therapeutic agents. T-cell immunoglobulin mucin-3 (TIM-3) is expressed on the surface of both LSCs and blasts in most AML patients, but not in normal hematopoietic stem cells (HSCs). We have developed KK2845, an antibody drug conjugate (ADC) that consists of an anti-TIM-3 fully human IgG1 antibody, a valine-alanine linker and a highly potent DNA cross-linking pyrrolobenzodiazepine (PBD) dimer SG3199. KK2845 exhibited potent cytotoxicity against AML cells both in vitro and in vivo. The cytotoxicity against AML cells was almost comparable between KK2845 and CD33-ADC, an anti-CD33 antibody conjugated with PBD dimer that has shown high remission rates in clinical studies. In addition to the cytotoxicity depending on PBD dimer, KK2845 also showed potent antibody-dependent cell cytotoxicity (ADCC) activity against AML cells. KK2845 showed less cytotoxicity against human normal bone marrow cells than CD33-ADC. The pharmacokinetics of KK2845 in cynomolgus monkey after intravenous infusion demonstrated a favorable profile. Taken together, these data suggest that KK2845 could be a novel ADC therapeutic in AML.
Collapse
Affiliation(s)
- Jian Zou
- Fuji Research Park, Biomedical Science Research Laboratories 2, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan.
| | - Haruka Kinosada
- Fuji Research Park, Biomedical Science Research Laboratories 2, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| | - Shin-Ichiro Takayanagi
- Tokyo Research Park, Biomedical Science Research Laboratories 2, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Machida, Tokyo, 194-8533, Japan
| | - Toshihiko Ishii
- Fuji Research Park, Biomedical Science Research Laboratories 2, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| | - Toru Amano
- Fuji Research Park, Biomedical Science Research Laboratories 2, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| | - Kaito Nihira
- Fuji Research Park, Toxicological Research Laboratories, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| | - Shohei Kanie
- Fuji Research Park, Toxicological Research Laboratories, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| | - Maiko Adachi
- Fuji Research Park, Pharmacokinetic Research Laboratories, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| | - Harunobu Tahara
- Fuji Research Park, Pharmacokinetic Research Laboratories, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| | - Teppei Sakoda
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, 812-8582, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Yoshikane Kikushige
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, 812-8582, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, 812-8582, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Hidetaka Satou
- Fuji Research Park, Biomedical Science Research Laboratories 2, Research Unit, Research Division, Kyowa Kirin Co., Ltd., Sunto-gun, Shizuoka, 411-8731, Japan
| |
Collapse
|
47
|
Wang X, Lu Y, Liu R, Huang L, Xu K, Xiong H, Nan D, Shou Y, Sheng H, Zhang H, Wang X, Chen X. LZTS2 methylation as a potential diagnostic and prognostic marker in LIHC and STAD: Evidence from bioinformatics and in vitro analyses. Sci Rep 2025; 15:17873. [PMID: 40404727 PMCID: PMC12098705 DOI: 10.1038/s41598-025-03153-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: 01/20/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
The rising mortality rate from cancer, driven by the absence of reliable biomarkers, highlights the pressing need for advanced diagnostic and prognostic strategies. This study investigates LZTS2's role as a pan-cancer biomarker, emphasizing its predictive value for immunotherapy and therapeutic targeting. Unlike existing biomarkers such as AFP in hepatocellular carcinoma or HER2 in gastric cancer, which exhibit tissue-specific utility, LZTS2 demonstrates unique cross-cancer applicability, as evidenced by its consistent dysregulation in both liver hepatocellular carcinoma (LIHC) and stomach adenocarcinoma (STAD) alongside emerging associations with other malignancies. Leveraging advanced bioinformatics tools and databases including UALCAN, KM-plotter, and The Cancer Genome Atlas (TCGA), alongside experimental validation in LIHC and STAD cell lines, we analyze LZTS2 expression patterns and their clinical relevance. Notably, LZTS2's dual role-acting as a tumor suppressor in some cancers while promoting oncogenesis in others-distinguishes it from conventional single-function markers, offering novel insights into its regulatory versatility. Our findings reveal that LZTS2 mutations and expression levels are closely associated with cancer progression and patient survival, solidifying its potential as a prognostic biomarker. Notably, LZTS2 expression correlates with various clinicopathological parameters, underscoring its significance in cancer biology. Pathway analysis highlights LZTS2's involvement in critical biological processes, providing actionable insights for therapeutic interventions. Quantitative real-time polymerase chain reaction (qRT-PCR) and quantitative methylation-specific PCR (qMSP) experimental validations confirm these results, further establishing LZTS2's utility as a multi-dimensional biomarker that integrates genetic, epigenetic, and immunological features-a capability rarely observed in existing markers. This comprehensive analysis positions LZTS2 as a pivotal player in cancer progression, opening promising avenues for enhanced clinical management.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Medical Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanwei Lu
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ruiqi Liu
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luanluan Huang
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Keke Xu
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hao Xiong
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ding Nan
- Graduate Department, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yiyi Shou
- Graduate Department, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Hailong Sheng
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Haibo Zhang
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Xiaoyan Chen
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
48
|
Zhang B, Chen Y, Gu X, Zheng Y, Jiang ZH. Fucosyltransferase 11 restrains ferroptosis via upregulation GPX4 expression in gastric cancer. BMC Cancer 2025; 25:923. [PMID: 40405129 PMCID: PMC12100908 DOI: 10.1186/s12885-025-14340-4] [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: 07/12/2024] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Ferroptosis is a novel iron-dependent type of programmed cell death that is characterized by the oxidation of lipids by divalent iron ions to produce lipid peroxides, which leads to cell death. Fucosyltransferase 11 (FUT11) is highly expressed in most tumors and is involved in tumorigenesis. However, there have been few studies regarding the relationship between FUT11 and ferroptosis. In this study, we found that FUT11 expression was abnormally high in gastric cancer (GC) cells and that the prognosis of patients with GC and high expression of FUT11 was poor. FUT11 expression was significantly correlated with the TNM stage of GC.Specific knockdown of FUT11 significantly inhibited the proliferation of GC cells, reduced the abundance of the key anti-ferroptotic protein glutathione peroxidase 4(GPX4), induced lipid peroxidation and ferroptosis in GC cells, and inhibited the proliferation of these cells. The overexpression of GPX4 reduced the inhibitory effect of FUT11 on GC cells. In addition, the knockdown of FUT11 significantly inhibited GC tumor growth in mice, and this inhibitory effect was reduced by the overexpression of GPX4. In conclusion, we have shown that FUT11 promotes GC progression by targeting GPX4, thereby inhibiting ferroptosis in GC cells. These findings suggest that FUT11 is a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Bingbing Zhang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China
| | - Yali Chen
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China
| | - Xuezhou Gu
- Department of General Surgery, Sheyang People's Hospital, Yancheng, Jiangsu, 224006, China
| | - Yu Zheng
- Department of Laboratory Medicine, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China
| | - Zhong Hua Jiang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, The Yancheng Clinical Medical College of Jiangsu University, Yancheng, Jiangsu, 224006, China.
| |
Collapse
|
49
|
Irestorm E, Bakker A, Tissing WJE, Maurice-Stam H, Dors N, Mavinkurve-Groothuis A, Plasschaert SLA, van Bindsbergen KLA, Grootenhuis MA, van Litsenburg RRL. Benchmarking the pediatric quality of life (PedsQL) cancer module in a large Dutch national cohort of childhood cancer patients. BMC Cancer 2025; 25:915. [PMID: 40399814 PMCID: PMC12096553 DOI: 10.1186/s12885-025-14322-6] [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: 02/27/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Pediatric cancer diagnosis and treatment reduce Health-Related Quality of Life (HRQoL). The Pediatric Quality of Life (PedsQL) Cancer Module is the most commonly used cancer specific HRQoL instrument, but most studies included relatively small cohorts and no reference scores are available. The development of a PedsQL Cancer Module benchmark, based on a large population of pediatric cancer patients, may benefit future research by providing insight into issues children encounter during treatment or clinical trials. A benchmark would also allow for earlier identification of patients who may benefit from targeted interventions in HRQoL monitoring programs. The primary aim of this study was therefore to provide benchmark scores from a large national cohort for use in pediatric oncology care and research. Secondary aims were to compare scores between subgroups (age, sex, and diagnosis group) and to describe the prevalence of impaired HRQoL. METHODS Data was collected from a national HRQoL monitoring program and extracted from medical records. HRQoL was measured with the PedsQL Cancer Module during treatment for childhood cancer, approximately 3 months after diagnosis. Comparisons were made for sex, age and diagnosis groups: hemato-oncology (HO), central nervous system-tumors (CNS), and solid tumors (ST) using the Mann-Whitney U test for the different subscales. Effect sizes were calculated. Impairment was defined on item level using descriptive statistics. RESULTS Proxy-reports of 492 children (age 2-7 years) and self-reports of 500 children (age 8-18 years) were available. HRQoL differences between age groups, sex and diagnosis groups were small, with the exception of nausea. On the nausea subscale there was a medium effect size difference with children with ST reporting more problems than children with CNS. Impaired HRQoL was most often reported on items reflecting nausea and procedural anxiety. CONCLUSIONS Early in the cancer trajectory, there are only minor differences between subgroups based on age, sex, and diagnosis group. These results from a large national cohort can be used as benchmark data in future clinical trials, studies and clinical assessments, and offer and adapt support targeted for improving HRQoL related to treatment for childhood cancer.
Collapse
Affiliation(s)
- Elin Irestorm
- Faculty of Medicine, Department of Paediatrics, Lund University, Lund, Sweden
- Princess Máxima Center for pediatric oncology, Utrecht, The Netherlands
| | - Anne Bakker
- Princess Máxima Center for pediatric oncology, Utrecht, The Netherlands
- Faculty of Medicine, Utrecht University, Utrecht, The Netherlands
| | - Wim J E Tissing
- Princess Máxima Center for pediatric oncology, Utrecht, The Netherlands
| | | | - Natasja Dors
- Princess Máxima Center for pediatric oncology, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
50
|
Wu Y, Liu J, Yin T, Li X, Liu X, Peng X, Zhan X. SELP can affect the immune microenvironment of gastric cancer and is associated with poor prognosis. Discov Oncol 2025; 16:846. [PMID: 40397261 PMCID: PMC12095770 DOI: 10.1007/s12672-025-02629-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in the occurrence and progression of gastric cancer. Yet, we still don't understand how immune and stromal components of TMEs are modulated. In this study, we applied the ESTIMATE algorithm to calculate the number of immune and stromal components in 410 STAD cases in the Cancer Genome Atlas (TCGA) database. COX regression analysis and protein-protein interaction (PPI) network construction were used to analyze differentially expressed genes (DEGs). Then, P-selectin (SELP) was identified as a predictor by cross-analysis of univariate COX and PPI. After verifying the clinical significance of SELP for study, we performed an immune infiltration analysis and identified 54 immunomodulators associated with SELP through public data. Immunomodulation associated with gastric cancer prognosis was then confirmed by LASSO regression, and the previous results were further validated with single-cell data. Finally, we verified that SELP can promote EMT on gastric cancer cells. In conclusion, we validated that SELP may affect the biological phenotype of gastric cancer with the immune microenvironment alteration of gastric cancer.
Collapse
Affiliation(s)
- Yue Wu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Jingyu Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Tong Yin
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaoxiao Li
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xian Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| |
Collapse
|