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] [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
|
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
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
Abd El-Kaream SA, Hassan NAM, Saleh HSA, Albahloul MAS, Khedr AM, El-Kholey SM. Microwave assisted drug delivery of titanium dioxide/rose Bengal conjugated chitosan nanoparticles for micro-photodynamic skin cancer treatment in vitro and in vivo. BMC Cancer 2025; 25:896. [PMID: 40389852 DOI: 10.1186/s12885-025-14285-8] [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: 07/28/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Micro-photodynamic therapy (MWPDT) combines photo-dynamic (PDT) and microwave-dynamic (MWDT) therapies with sensitizers, offers new avenues for cancer treatment. Despite the fact that novel sensitizers for MWPDT have been successfully synthesized, only a few are being employed effectively. The low tumor-targeting specificity, inability to transport sensitizer's deeper intratumorally, and deteriorating tumor microenvironment all restrict their anti-tumor efficacy. The current work was done aiming at microwave assisted drug delivery of titanium dioxide / rose Bengal conjugated chitosan nanoparticles (TiO2/RB@CSNP) for micro- photo-dynamic skin cancer (SKCA) treatment in vitro and in vivo as activated cancer treatment up-to-date modality. MATERIALS AND METHODS The study was conducted in vitro on human SKCA cells (A-375) and the study protocol application groups in vivo on Swiss albino mice treated with 7,12-dimethylbenz[a]anthracene (DMBA)/croton oil only and were not received any treatment for inducing SKCA, and only after SKCA induction the study treatment protocol began, treatment was daily with TiO2/RB@CSNP as MWPDT sensitizer with or without exposure to laser (IRL) or microwave (MW) or a combination of them for 3 min for two weeks. RESULTS Revealed that CSNP can be employed as effective TiO2/RB delivery system that directly targets SKCA cells. Additionally TiO2/RB@CSNP is a promising MWPS for and when combined with MWPDT can be very effective in treatment of SKCA-A-375 in vitro (cell viability decreased in a dose-dependent basis, the cell cycle progression in G0/G1 was slowed down, and cell death was induced as evidenced by an increase in the population of Pre-G cells, an increase in early and late apoptosis and necrosis, and an increase in autophagic cell death) and DMBA/croton oil SKCA-induce mice in vivo (induced antiproliferative genes (caspase 3,9, p53, Bax, TNFalpha), suppressed antiapoptotic and antiangiogenic genes (Bcl2,VEGF respectively) effectively reducing the tumors growth and leading to cancer cell death as well as decreased oxidative stress (MDA), and ameliorated enzymatic and non-enzymatic antioxidants (SOD, GR, GPx, GST, CAT, GSH, TAC) as well as renal (urea, creatinine) and hepatic (ALT, AST) functions. This process could be attributed to MWPDT; microwave and/or photo-chemical TiO2/RB activation mechanism and antioxidant potential of non activated TiO2/RB as well. CONCLUSION The results indicate that TiO2/RB@CSNP has great promise as an innovative, effective delivery system for selective localized treatment of skin cancer that is activated by MWPDT.
Collapse
Affiliation(s)
- Samir Ali Abd El-Kaream
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | | | | | | | - Sohier Mahmoud El-Kholey
- Medical Biophysics Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
21
|
Wang Y, Liu N, Xu C, Wang J, Dong L, Yang S, Jiang J. Nebulized inhalation of extracellular vesicles containing SPOCK2 suppresses lung adenocarcinoma progression via MAPK inhibition. Discov Oncol 2025; 16:797. [PMID: 40382517 PMCID: PMC12085455 DOI: 10.1007/s12672-025-02626-9] [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: 11/15/2024] [Accepted: 05/08/2025] [Indexed: 05/20/2025] Open
Abstract
Aberrant expression of SPARC/osteonectin, cwcv and kazal-like domains proteoglycan 2 (SPOCK2) plays a role in the development and progression of several human cancers. However, the importance of its expression and function in lung adenocarcinoma (LUAD) remains unclear. The present study aimed to elucidate the role of SPOCK2 in the growth of LUAD and propose a novel therapeutic insight for LUAD through SPOCK2. SPOCK2 protein expression was significantly reduced in LUAD tissues and cells by Immunohistochemical assay and Western blot. CCK-8, colony formation, and Transwell assays were used to demonstrate that SPOCK2 overexpression inhibited both proliferation and migration of LUAD cells in vitro. This inhibition of tumor growth was further confirmed by a LUAD xenograft mouse model in vivo. To explore downstream target signal of SPOCK2 in LUAD, RNA transcriptome sequencing was performed and enrichment analysis showed an association between SPOCK2 expression and the MAPK pathway. Furthermore, HEK293T cells were modified with SPOCK2, and extracellular vesicles (EVs) containing SPOCK2 (SPOCK2-EVs) were collected through ultra-high-speed centrifugation. Interestingly, co-culture with SPOCK2-EVs significantly increased SPOCK2 levels within LUAD cells. Furthermore, SPOCK2-EVs effectively inhibited LUAD growth in vitro and in vivo studies. Because directly injecting SPOCK2-EVs into tumors presents challenges for internal organs, we investigated the efficacy of nebulized SPOCK2-EVs for LUAD treatment. Consistent with our findings from intratumoral injection, nebulized inhalation of SPOCK2-EVs resulted in significant inhibition of LUAD growth. These results strongly suggest that SPOCK2 released by HEK293T-EVs can effectively inhibit LUAD tumor growth and hold promise for future clinical translation in cancer therapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, No. 9 Chongwen Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Ningning Liu
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Chuanqin Xu
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Jing Wang
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Liyang Dong
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu, People's Republic of China.
| | - Shuang Yang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, No. 9 Chongwen Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China.
- Center for Clinical Mass Spectrometry, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
| | - Junhong Jiang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, No. 9 Chongwen Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Hu C, Nong S, Ke Q, Wu Z, Jiang Y, Wang Y, Chen Y, Wu Z, Zhang Q, Liao C, Wu M. Simultaneous co-delivery of Ginsenoside Rg3 and imiquimod from PLGA nanoparticles for effective breast cancer immunotherapy. iScience 2025; 28:112274. [PMID: 40256328 PMCID: PMC12008673 DOI: 10.1016/j.isci.2025.112274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/11/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Breast cancer is a fatal malignancy facing human health, with most patients experiencing recurrence and resistance to chemotherapy. The immunosuppressive tumor microenvironment (TME) greatly limits the actual outcome of immunotherapy. This study aimed to develop a modality of theranostics nanoparticles for breast cancer based on a near-infrared light-triggered nanoparticle for the targeted delivery of ginsenoside Rg3 and immune adjuvants imiquimod (R837) for effective breast cancer immunotherapy. Folate-receptor (FA) targeting IR780-R837/ginsenoside Rg3-perfluorohexane (PFH) @ polyethylene glycol (PEG)-poly (lactide-co-glycolic acid) (PLGA) nanoparticles (FA-NPs) can be activated by near-infrared laser irradiation in tumors, which leads to rapid release of ginsenoside Rg3 and R837 in the regions with high expression of folate receptors and glucose transporter 1 (GLUT1). Meanwhile, the nanoparticles can be used as dual-mode contrast agents for photoacoustic and ultrasound imaging. This strategy provides a strong immune memory effect, which can prevent tumor recurrence after eliminating the initial tumor.
Collapse
Affiliation(s)
- Cong Hu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Shuxiong Nong
- Department of Cardiology, Baise People’s Hospital. Affiliated Southwest Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Qianqian Ke
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Ziming Wu
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuancheng Jiang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Ying Wang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Yixin Chen
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Ziling Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Qi Zhang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Chilin Liao
- Department of Cardiology, Baise People’s Hospital. Affiliated Southwest Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
23
|
Firouzi-Farsani K, Dehghani-Samani M, Gerami R, Sadat Moosavi R, Gerami M, Mahdevar M. Exploring non-coding RNA expression profiles of AKR1B10P1, RP11-465B22.3, WASH8P, and NPM1P25 as a predictive model for hepatocellular carcinoma patient survival. Discov Oncol 2025; 16:771. [PMID: 40372570 PMCID: PMC12081793 DOI: 10.1007/s12672-025-02475-6] [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: 10/31/2024] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
The primary aim of the study was to analyze novel long non-coding RNAs (lncRNAs) in hepatocellular carcinoma (HCC) to assess their roles as potential oncogenes and tumor suppressors and to develop a survival prediction model based on their expression levels. Data from The Cancer Genome Atlas, GSE135631, and GSE214846, were utilized to evaluate changes in lncRNA expression in HCC and their associations with patient prognosis. A risk model was created based on lncRNA expression to predict patient mortality. The co-expression network was employed to identify associated pathways, and the results were subsequently validated using the RT-qPCR method. The findings indicated that 14 lncRNAs were down-regulated in HCC, and their increased expression was associated with a favorable prognosis. Additionally, eight lncRNAs were overexpressed and correlated with poorer patient outcomes. The multivariate Cox regression analysis demonstrated that overexpression of AKR1B10P1, RP11-465B22.3, WASH8P, and the downregulation of NPM1P25 could independently predict patient survival, irrespective of clinical variables. The risk score model based on these lncRNAs effectively stratified patients by their mortality risk. Furthermore, the co-expression network analysis revealed that the identified lncRNAs might be involved in various pathways, including fatty acid metabolism, mTOR signaling, glycolysis, angiogenesis, Wnt-β-catenin pathway, and DNA repair. RT-qPCR results validated the significant increase in the expression level of WASH8P in cancer specimens compared to normal tissues. Our results unveiled that changes in the expression levels of AKR1B10P1, RP11-465B22.3, WASH8P, and NPM1P25 were significantly and independently associated with prognosis. Moreover, the patient mortality risk model constructed using these four lncRNAs exhibited a robust capacity to accurately predict patients' survival rates.
Collapse
Affiliation(s)
| | - Mina Dehghani-Samani
- Department of Bioinformatics, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Razieh Gerami
- Independent Researcher, Graduated of PhD Pharmacology from Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Reihaneh Sadat Moosavi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Marzieh Gerami
- Department of Computer Engineering, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohammad Mahdevar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Genius Gene, Genetics and Biotechnology Company, Isfahan, Iran
| |
Collapse
|
24
|
Dennehy C, Conroy MR, Forde PM. Immunotherapy for resectable lung cancer. Cancer 2025; 131:e35849. [PMID: 40334018 PMCID: PMC12057804 DOI: 10.1002/cncr.35849] [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: 09/16/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 05/09/2025]
Abstract
Lung cancer remains a significant global health challenge, demanding innovative treatment strategies. Immune checkpoint blockade has revolutionized cancer care, leading to improved survival across advanced malignancies and has now become a standard therapy for earlier stage, resectable lung cancer. This review article consolidates the current landscape and future prospects of neoadjuvant and perioperative immunotherapy in lung cancer. The authors outline key findings from clinical trials in resectable lung cancer, including early efficacy, safety profiles, and emerging impact on disease recurrence, and overall survival. Additionally, this review elucidates the challenges encountered, including patient selection criteria, optimal treatment schedules, immune-related adverse events, and impact on surgery. This comprehensive analysis amalgamates current evidence with future directions, providing a roadmap for clinicians, researchers, and stakeholders to navigate the dynamic realm of immunotherapy for surgically resectable lung cancer.
Collapse
Affiliation(s)
- Colum Dennehy
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Michael R. Conroy
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Patrick M. Forde
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
25
|
Wang M, Guo Z, Zhao S, Liu L, Shi Y, Li H, Su J, Zhang N, Li J, Wu Y. CD49d promotes T-cell senescence in chronic lymphocytic leukaemia. Br J Haematol 2025. [PMID: 40375447 DOI: 10.1111/bjh.20135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/25/2025] [Indexed: 05/18/2025]
Abstract
While CD49d (α4 integrin) is an established prognostic marker in chronic lymphocytic leukaemia (CLL) and is associated with aggressive disease, its impact on T-cell biology remains poorly understood. Compared to healthy donors, CLL patients exhibited significantly elevated CD49d expression in both CD4+ and CD8+ T cells (p < 0.001) as detected by flow cytometry, which was also confirmed by the single-cell RNA sequencing (scRNA-seq) (p < 0.001). Differentially expressed genes in CD49d+ T (both CD8+ and CD4+ T cells) versus CD49d- T cells identified in CLL patients were enriched in cellular senescence pathways, while this phenomenon is absent in healthy individuals. Functional validation demonstrated that CD49d+ T cells displayed elevated senescence-associated markers (e.g. interferon-gamma, granzyme B) and a shift towards memory phenotypes, correlating with immunosuppressive signatures. This discovery suggests that targeting CD49d-dependent senescence pathways may reverse T-cell dysfunction in CLL immunotherapy.
Collapse
Affiliation(s)
- Min Wang
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Zhen Guo
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Sishu Zhao
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Lu Liu
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Yu Shi
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Hui Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Su
- Department of Hematology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Ninghan Zhang
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Jianyong Li
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Yujie Wu
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Deng S, Wang J, Zou F, Cheng D, Chen M, Gu J, Shi J, Yang J, Xue Y, Jiang Z, Qin L, Mao F, Chang X, Nie X, Liu L, Cao Y, Cai K. Palmitic Acid Accumulation Activates Fibroblasts and Promotes Matrix Stiffness in Colorectal Cancer. Cancer Res 2025; 85:1784-1802. [PMID: 39992719 PMCID: PMC12079102 DOI: 10.1158/0008-5472.can-24-2892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/27/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Obstructions can occur during any stage of colorectal cancer and correspond with poor prognosis. Obstructive colorectal cancer (OCRC) is harder and exhibits increased tumor budding and proliferation of myofibroblasts compared with nonobstructive colorectal cancer, suggesting that the occurrence of obstruction may be related to extracellular matrix (ECM) remodeling. In this study, we found that colorectal cancer and OCRC samples differed substantially in ECM composition, specifically in collagen (newly formed and mature) and proteoglycans (including glycosaminoglycan, hyaluronic acid, and chondroitin sulfate). OCRC also exhibited considerable changes in ECM biomechanics and collagen arrangement. Interestingly, OCRC samples presented a notable increase in matrix cancer-associated fibroblasts (mCAF). The abundance of mCAFs correlated with the accumulation of palmitic acid (PA), and high concentrations of PA increased the secretion of ECM-related proteins by mCAFs. Additionally, PA did not directly affect normal fibroblasts but rather activated the NF-κB pathway in tumor cells to stimulate secretion of CSF1, TGFβ1, and CXCL8, which promoted the activation of normal fibroblasts into mCAFs and exacerbated ECM stiffening. Drug screening with a natural compound library identified vanillylacetone as a potential inhibitor of PA-induced cytokine secretion and ECM stiffening. These findings highlight intratumoral PA accumulation as a key mechanism driving ECM alterations and OCRC progression and suggest that targeting this axis may be useful for treating patients with colorectal cancer with risk of obstruction. Significance: Palmitic acid accumulation activates the NF-κB pathway in colorectal cancer cells to promote cytokine secretion that facilitates the generation of matrix cancer-associated fibroblasts, driving extracellular matrix remodeling and development of obstructions.
Collapse
Affiliation(s)
- Shenghe Deng
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Falong Zou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Denglong Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mian Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junnan Gu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianguo Shi
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Yang
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Xue
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenxin Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Le Qin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuwei Mao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaona Chang
- Department of Pathology, Union Hospital, Tongji Medical, Huazhong University of Science and Technology, Wuhan, China
| | - Xiu Nie
- Department of Pathology, Union Hospital, Tongji Medical, Huazhong University of Science and Technology, Wuhan, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinghao Cao
- Department of Digestive Surgical Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Kim YS, Kim SJ. Diagnostic Performances of Radiolabeled FAPI PET/CT for Lymph Node Staging in Head and Neck Cancer Patients: Comparison With 18F-FDG PET/CT. Clin Nucl Med 2025:00003072-990000000-01726. [PMID: 40367495 DOI: 10.1097/rlu.0000000000005973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
OBJECTIVE The purpose of the current study was to compare the diagnostic performances of radiolabeled FAPI and 18F-FDG PET/CT for the detection of lymph node (LN) metastasis in head and neck cancer (HNC) patients. METHODS The PubMed, Cochrane database, and EMBASE database, from the earliest available date of indexing through December 31, 2024, were searched for studies comparing diagnostic performances of radiolabeled FAPI and 18F-FDG PET/CT for the detection of metastatic LN in HNC patients. We estimated pooled sensitivities and specificities across studies. RESULTS Across 8 studies (14 results), the pooled sensitivity of FAPI PET/CT was 0.89 and the pooled specificity was 0.93. The pooled sensitivity of 18F-FDG PET/CT was 0.91 and the pooled specificity was 0.50. On patient-based analysis, the estimated sensitivity and specificity of FAPI were 0.96 and 0.96, and those of 18F-FDG were 0.95 and 0.34, respectively. On lesion-based analysis, the estimated sensitivity and specificity of FAPI were 0.84 and 0.94, and those of 18F-FDG were 0.86 and 0.78, respectively. On neck side-based analysis, the estimated sensitivity and specificity of FAPI were 0.88 and 0.79, and those of 18F-FDG were 0.91 and 0.29, respectively. CONCLUSIONS Radiolabeled FAPI showed a good diagnostic performance for the detection of metastatic LN in HNC patients. Also, 18F-FDG PET/CT revealed low specificity for LN staging in HNC patients. Future large multicenter research with more patients would be necessary to provide a more comprehensive overview of the usefulness of radiolabeled FAPI for LN staging in HNC patients.
Collapse
Affiliation(s)
- Yun Seong Kim
- Department of Internal Medicine
- BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital
| | - Seong-Jang Kim
- BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital
- Department of Nuclear Medicine, Pusan National University School of Medicine
- Department of Nuclear Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
28
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
29
|
Xiong Y, Tong Y. Development of macrophage M2 relate signature for predicting prognosis and immunotherapy response in ovarian cancer. Discov Oncol 2025; 16:750. [PMID: 40358821 PMCID: PMC12075037 DOI: 10.1007/s12672-025-02559-3] [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: 11/18/2024] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Ovarian cancer ranks as the fifth most common cause of cancer-related deaths in women worldwide. Macrophages M2 is believed to support tumor growth by suppressing immune responses and promoting angiogenesis. METHODS A macrophage M2-related signature (MRS) was developed by applying machine learning methods that included 10 algorithms and utilized data from the TCGA, GSE14764 and GSE140082 datasets. The predictive capacity of the MRS for immunotherapy response was evaluated through various methods, including immunophenoscore, TIDE score, TMB score, immune escape score, as well as two immunotherapy cohorts (IMvigor210 and GSE91061). RESULTS The optimal MRS, developed using the lasso algorithm, served as an independent prognostic factor and demonstrated stable performance in predicting overall survival rates in ovarian cancer. In the TCGA dataset, the AUC values for the 1-, 3-, and 5-year ROC curves were 0.874, 0.808, and 0.813, respectively. The C-index of our MRS was superior to that of clinical stage, tumor grade, and several other established prognostic signatures. Ovarian cancer patients with low risk score exhibited higher ESTIMATE score, increased levels of immune cells, elevated PDI&CTLA4 immunophenoscore, higher TMB score, reduced TIDE score, and lower immune escape score. Additionally, the survival prediction nomogram displayed significant potential for clinical application in estimating the 1-, 3-, and 5-year overall survival rates of ovarian cancer patients. CONCLUSION Our study developed a novel MRS for ovarian cancer, which could act as an indicator for predicting the prognosis and immunotherapy response in ovarian cancer.
Collapse
Affiliation(s)
- Yifei Xiong
- Reproductive Medicine Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Yan Tong
- Reproductive Medicine Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.
| |
Collapse
|
30
|
Koprulu M, Wheeler E, Kerrison ND, Denaxas S, Carrasco-Zanini J, Orkin CM, Hemingway H, Wareham NJ, Pietzner M, Langenberg C. Sex differences in the genetic regulation of the human plasma proteome. Nat Commun 2025; 16:4001. [PMID: 40360480 PMCID: PMC12075630 DOI: 10.1038/s41467-025-59034-4] [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: 03/07/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Mechanisms underlying sex differences in the development and prognosis of many diseases remain largely elusive. Here, we systematically investigated sex differences in the genetic regulation of plasma proteome (>5800 protein targets) across two cohorts (30,307 females; 26,058 males). Plasma levels of two-thirds of protein targets differ significantly by sex. In contrast, genetic effects on protein targets are remarkably similar across sexes, with only 103 sex-differential protein quantitative loci (sd-pQTLs; for 2.9% and 0.3% of protein targets from antibody- and aptamer-based platforms, respectively). A third of those show evidence of sexual discordance, i.e., effects observed in one sex only (n = 30) or opposite effect directions (n = 1 for CDH15). Phenome-wide analyses of 365 outcomes in UK Biobank did not provide evidence that the identified sd-pQTLs accounted for sex-differential disease risk. Our results demonstrate similarities in the genetic regulation of protein levels by sex with important implications for genetically-guided drug target discovery and validation.
Collapse
Grants
- MC_UU_00006/1 RCUK | Medical Research Council (MRC)
- MC_PC_13046 RCUK | Medical Research Council (MRC)
- MC_UU_00006/1 RCUK | Medical Research Council (MRC)
- SP/19/3/34678 British Heart Foundation (BHF)
- The Fenland Study (DOI 10.22025/2017.10.101.00001) is funded by the Medical Research Council (MC_UU_12015/1). We further acknowledge support for genomics from the Medical Research Council (MC_PC_13046). This work is supported by the Medical Research Council (MC_UU_00006/1 - Etiology and Mechanisms) (C.L., E.W., M.P., N.K., and N.J.W.). M.K. is supported by Gates Cambridge Trust. H.H. is supported by Health Data Research UK and the NIHR University College London Hospitals Biomedical Research Centre. S.D. is supported by a) the BHF Data Science Centre led by HDR UK (grant SP/19/3/34678), b) BigData@Heart Consortium, funded by the Innovative Medicines Initiative-2 Joint Undertaking under grant agreement 116074, c) the NIHR Biomedical Research Centre at University College London Hospital NHS Trust (UCLH BRC), d) a BHF Accelerator Award (AA/18/6/24223), e) the CVD-COVID-UK/COVID-IMPACT consortium and f) the Multimorbidity Mechanism and Therapeutic Research Collaborative (MMTRC, grant number MR/V033867/1). J.C.Z. was supported by a 4-year Wellcome Trust PhD Studentship and the Cambridge Trust.
Collapse
Affiliation(s)
- Mine Koprulu
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Nicola D Kerrison
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Spiros Denaxas
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
- British Heart Foundation Data Science Centre, London, UK
- National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Julia Carrasco-Zanini
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Chloe M Orkin
- Blizard Institute and SHARE Collaborative, Queen Mary University of London, London, UK
- Department of Infection and Immunity, Barts Health NHS Trust, London, UK
| | - Harry Hemingway
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
- National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Maik Pietzner
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
31
|
Kong L, Yiu CH, Lu CY. Effectiveness and Safety of Immune Checkpoint Inhibitors in Colorectal Cancer: A Systematic Review of Real-World Studies. Curr Oncol Rep 2025:10.1007/s11912-025-01676-0. [PMID: 40358904 DOI: 10.1007/s11912-025-01676-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICIs) have demonstrated significant efficacy in the treatment of colorectal cancer (CRC). However, most evidence has come from clinical trials with strict eligibility criteria. Understanding real-world effectiveness and safety of ICIs in CRC is important to guide routine clinical practice across diverse populations. RECENT FINDINGS A systematic review following PRISMA guidelines was conducted to identify observational studies evaluating ICI-based regimens compared to conventional or combination therapies in patients with CRC. Three databases (MEDLINE, Embase, and Scopus) were searched from inception through March 15, 2025. Eligible studies reported at least one efficacy outcome (e.g., progression-free survival [PFS], overall survival [OS], etc.) and/or safety outcome (e.g., adverse events) among real-world populations with CRC treated with ICIs. Study quality was assessed using the Newcastle-Ottawa Scale, and a narrative synthesis was performed to summarize the key findings. Eleven real-world studies met the inclusion criteria, encompassing data from 2,049 patients. In MSI-H/dMMR metastatic CRC, real-world findings aligned with the survival benefits observed in clinical trials, demonstrating improved PFS and OS compared to conventional therapies. For MSS/pMMR metastatic CRC, combining ICIs with other agents (e.g., tyrosine kinase inhibitors or chemotherapy) showed improvements but yielded conflicting results. Overall, the safety profiles were comparable to conventional therapies, with treatment-related adverse events occurring at similar rates. Real-world evidence supports the efficacy of ICI monotherapy in MSI-H/dMMR metastatic CRC and suggests potential benefits of ICI-based combination therapies in MSS/pMMR metastatic CRC. However, most of the data are derived from small, single-center cohorts, which limit their generalizability. Further multi-center studies are needed, especially to assess the efficacy of ICI-based combination therapies in the broader CRC population.
Collapse
Affiliation(s)
- Leping Kong
- The University of Sydney School of Pharmacy, Camperdown, NSW, Australia
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Chin Hang Yiu
- The University of Sydney School of Pharmacy, Camperdown, NSW, Australia
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Christine Y Lu
- The University of Sydney School of Pharmacy, Camperdown, NSW, Australia.
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia.
- Department of Pharmacy, Royal North Shore Hospital, St Leonards, New South Wales, Australia.
| |
Collapse
|
32
|
Hailu BA. Trends in cancer incidence and mortality in the United States (1999-2021): A retrospective cohort study of health disparities and regional variations. J Natl Med Assoc 2025:S0027-9684(25)00030-6. [PMID: 40350349 DOI: 10.1016/j.jnma.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/18/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
OBJECTIVE Analyze cancer incidence and mortality in the U.S. 1999-2021, focusing on variations by sex, race, cancer type, and state. The study aims to identify disparities in cancer rates and highlight regions and populations most affected, to inform public health policies and interventions. METHODS Age-adjusted rates were analyzed by race, cancer type, sex, and state. Bayesian statistical techniques were employed for accurate trend estimation, and cancer hotspots were identified using the Getis-Ord Gi* statistic. Joinpoint regression was used to estimate the Average Annual Percent Change (AAPC) in cancer rates. The analysis was conducted using R. RESULTS The overall trend was noted reduction in aggregate cancer rate (AAPC =-0.007) and mortality (AAPC =-0.004). Regional and racial disparities are still prevalent with outcomes showing that Kentucky, West Virginia, and Mississippi have the highest prevalence of both. Cancer incidence was reduced most significantly among Black or African American individuals (AAPC=-0.008) and mortality in (AAPC=-0.021) but their rates are still higher. Hotspot analyses identified high cancer incidence and mortality clusters, particularly in the Midwestern and Southern regions of the United States. DISCUSSION The implications arising from these results suggest that focused public health campaigns need to be directed toward cancer, smoking, and access to care services for effective health promotion. These findings therefore call for innovative public health measures to reduce the burden of cancer and disparity in the future. More sophisticated studies of the social, environmental, and genetic causes of these trends are required to improve the efficacy of cancer control.
Collapse
Affiliation(s)
- Bayuh Asmamaw Hailu
- Planning, Monitoring, and Evaluation Unit Leader, Wollo University, Dessie, Ethiopia.
| |
Collapse
|
33
|
Zhao N, Xiong L, Bai X, Pan W, Hai P, Ye H, Zhao T, Cui K, Ma R, Wang Y. Risk factors for radiation pneumonitis in NSCLC patients treated with third-generation EGFR TKIs and chest radiotherapy. Radiat Oncol 2025; 20:72. [PMID: 40346592 PMCID: PMC12065359 DOI: 10.1186/s13014-025-02649-0] [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: 03/12/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) patients receiving third-generation EGFR TKIs with thoracic radiotherapy (TRT) significantly prolong survival and also increase the incidence of radiation pneumonitis (RP). The aim of our study was to investigate the incidence and risk factors of RP in NSCLC patients receiving third-generation EGFR TKIs and TRT. PATIENTS AND METHODS We retrospectively evaluated NSCLC patients who received both third-generation EGFR TKIs and TRT at the General Hospital of Ningxia Medical University from January 2023 to September 2024. RP was diagnosed by clinical symptoms on computed tomography (CT) scans and graded according to the Common Terminology Criteria for Adverse Events 5.0. Risk factors for RP were determined by univariate and multivariate logistic regression analysis. RESULTS Of the 42 patients included, 26 (61.9%) developed RP and 14 (33.3%) developed grade ≥ 2 RP. Grade ≥ 2 RP all occurred within 6 months of receiving TRT, and the median time from TRT to RP was 3.69 months (2-10 months). GTV ≥ 39 ml and total lung V20 ≥ 14.95% were found to be independent risk factors for RP development. CONCLUSION The strategy of combining a third-generation TKI with TRT significantly increases the incidence of RP, and the risk of RP in these patients can be reduced by adjusting lung radiation dosimetry parameters. In NSCLC patients taking triple-generation TKIs with primary tumour progression, the timing and dose of TRT addition must be strictly controlled to optimise the therapeutic strategy and reduce the incidence of RP. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Nan Zhao
- Master Training Station, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Liang Xiong
- Graduate School of Ningxia Medical University, Yinchuan, China
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xuehong Bai
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wenyan Pan
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ping Hai
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hongqiang Ye
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ting Zhao
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Kai Cui
- Master Training Station, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Rong Ma
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yanyang Wang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, China.
- Cancer Institute, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
34
|
Hemo O, Chepeliuk O, Zilberman DE, Shvero A, Kleinmann N, Dotan ZA, Rosenzweig B. Impact of prior endoscopic management of upper tract urothelial carcinoma on nephroureterectomy complexity. World J Urol 2025; 43:289. [PMID: 40343505 DOI: 10.1007/s00345-025-05634-0] [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: 01/28/2025] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
PURPOSE Kidney-sparing endoscopic surgery is considered the preferred approach for the treatment of low-risk upper tract urothelial cancer (UTUC), yet its impact on subsequent radical nephroureterectomy's (RNU) surgical complexity remains uncertain. This study aims to evaluate the effect of prior endoscopic procedures on peri-operative outcomes of UTUC patients undergoing RNU. METHODS A retrospective analysis of 176 consecutive UTUC patients who underwent RNU from July 2008 to July 2023 at a single tertiary center. Demographic, clinical, and pathological data were recorded. The cohort was stratified based on the intention of endoscopic intervention: patients who did not undergo ureteroscopy and those who underwent a single diagnostic ureteroscopy only (non-mURS group) were compared to those who underwent multiple endoscopic treatments with laser ablation as part of a kidney-sparing strategy before RNU referral (mURS group). Statistical analyses addressed the correlations between ureteroscopy frequency and surgical complexity. Multivariate regression analysis was conducted to assess surgical complexity. RESULTS Among 176 patients who underwent RNU, 63 (35.7%) were included in the mURS group, and 113 (64.3%) in the non-mURS group. The mURS patients had longer RNU duration (306 ± 114 vs. 269 ± 114 min, p = 0.005), greater blood loss (hemoglobin decrease of 3.5 ± 1.1 vs. 1.02 ± 0.49 g/dL, p < 0.001), increased peri-ureteral adhesions (88.9% vs. 25.7%, p < 0.001), higher postoperative opioid use (87% vs. 53%, p < 0.001), and higher prevalence of fibrosis in pathology reports (88.9% vs. 30.9%, p < 0.001). No significant differences were found in post-operative complications. CONCLUSION Pre-RNU endoscopic treatment for UTUC is associated with increased surgical complexity of RNU without adversely affecting postoperative complication rates.
Collapse
Affiliation(s)
- Orel Hemo
- Department of Urology, Sheba Medical Center at Tel Hashomer, Ramat-Gan, Israel.
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Oleksander Chepeliuk
- Department of Urology, Sheba Medical Center at Tel Hashomer, Ramat-Gan, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit E Zilberman
- Department of Urology, Sheba Medical Center at Tel Hashomer, Ramat-Gan, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Asaf Shvero
- Department of Urology, Sheba Medical Center at Tel Hashomer, Ramat-Gan, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Kleinmann
- Department of Urology, Sheba Medical Center at Tel Hashomer, Ramat-Gan, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zohar A Dotan
- Department of Urology, Sheba Medical Center at Tel Hashomer, Ramat-Gan, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Barak Rosenzweig
- Department of Urology, Sheba Medical Center at Tel Hashomer, Ramat-Gan, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
35
|
Fang A, Frigo DE, Hahn A, Razouki Z, Hwang J, Koutroumpakis E, Lawen T, Smith M, Hamilton-Reeves J, DiGiovanni J, Higgason N, Garcia RS, Chapin BF, Pettaway C, Chery L, Troncoso P, Logothetis C, Daniel CR, Wei P, Gregg JR. GLP-1 Agonist Use Among Men with Localized Prostate Cancer: A Narrative Review and Rationale for Prospective Clinical Trials. Urology 2025:S0090-4295(25)00426-1. [PMID: 40348027 DOI: 10.1016/j.urology.2025.04.056] [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: 03/11/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
GLP-1 receptor agonists are a revolutionary treatment for obesity and investigations into other uses of these agents are ongoing. Epidemiologic data suggest that prostate cancer risk may decrease following GLP-1 receptor agonist initiation. To better define the potential role of GLP-1 receptor agonist use among men with prostate cancer, we will define changes that occur in men with prostate cancer following lifestyle intervention; illuminate the current relationship between these interventions and prostate cancer risk; and explore the potential mechanism of action on tumor biology, providing rationale for evaluating its effects on prostate cancer in a planned clinical trial.
Collapse
Affiliation(s)
- Andrew Fang
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Daniel E Frigo
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Andrew Hahn
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Zayd Razouki
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jessica Hwang
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | | | - Tarek Lawen
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Matthew Smith
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | | | | | - Noel Higgason
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Rebekka S Garcia
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Brian F Chapin
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Curtis Pettaway
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Lisly Chery
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | | | | | - Carrie R Daniel
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Peng Wei
- The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Justin R Gregg
- The University of Texas MD Anderson Cancer Center, Houston, TX USA.
| |
Collapse
|
36
|
Chiaverini L, Ferraro G, Di Leo R, Barresi E, La Mendola D, Bartoli F, Famlonga L, Satriano C, Faviana P, Zucchi A, Pacini M, Gailer J, Giacomelli C, Marzo T. From conventional therapy to novel nano-based approaches. A focus on prostate cancer. Nanomedicine (Lond) 2025:1-18. [PMID: 40329819 DOI: 10.1080/17435889.2025.2501513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
The currently available clinical anticancer approaches pertaining to the treatment of prostate cancer are summarized here. After providing an overview of the main features of this highly impactful global disease, the currently available clinical treatments are briefly reviewed. Then, alternative and innovative nano-based therapeutic options that have been proposed or are currently being explored to significantly improve prostate cancer management (i.e. anti-prostate cancer polymeric nanoparticles loaded with drugs to promote their release and biological activity, including non-targeted and functionalized PLGA-PEG NPs and AuNPs), are introduced. Furthermore, the problem of gathering insights into the mechanistic aspects related to the fate of the nanoformulation in complex matrices, such as blood plasma, is addressed.
Collapse
Affiliation(s)
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples 'Federico II', Napoli, Italy
| | - Riccardo Di Leo
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Institute of Clinical Physiology, Nationale Research Council (CNR), Pisa, Italy
| | | | | | - Francesco Bartoli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Luca Famlonga
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Cristina Satriano
- NanoHybrid BioInterfaces Laboratory (NHBIL), Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Pinuccia Faviana
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Alessandro Zucchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Matteo Pacini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Jürgen Gailer
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB, Canada
| | | | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Pisa, Italy
| |
Collapse
|
37
|
Cheng Y, Zhao L, Wen Y, Ren Z, Zeng J, Shi R, Cai X, Dong Q, Chen L, Lin C, Chen Z. In vivo spatiotemporal acquisition of metabolic vibrational signatures for unraveling gastric ulcer genesis. Biomaterials 2025; 322:123383. [PMID: 40367813 DOI: 10.1016/j.biomaterials.2025.123383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
Metabolic abnormalities in gastric juice usually directly reflect pathological changes of the gastric mucosa. Accurate in-situ gastric metabolic dynamics acquisition is crucial for understanding the occurrence and development of gastric diseases but is challenging. Here, an integrated magnetoplasmonic system (MPS) for long-term spatiotemporal metabolic information profiling and ulcer assessment in vivo is presented. Porous calcium alginate-silver plasmonic hydrogel shell and FeCo@Graphene magnetic core were fabricated into the durable magnetoplasmonic system via a coaxial electrospinning technique. MPS pumped gastric juice through enriching, filtering and magnetic actuation, which had synergistic effect on improving efficient capture of free-metabolites with promotion of 9.76 times. Multiplexed metabolites vibration fingerprint profiles were concurrently determined both in harsh simulated gastric fluid (SGF) and isolated stomach models. We also successfully achieved acquisition of in-situ metabolites changes within rat stomach. Marginal histograms, derived from time-resolved surface-enhanced Raman spectroscopy (SERS) investigations of free small molecules adenine, tyrosine, and phenylalanine, suggested a positive correlation in metabolite levels across different stages. Moreover, ulcers revelation was accomplished with high precision through leveraging spectral dimensionality reduction and random forest classification of SERS profiles. Metabolites correlation analysis indicated that Raman signal appearing at 1602 cm-1 and 2112 cm-1 corresponding to phenylalanine and amine exhibited strong positive correlations following ulcer onset. This research represents the first endeavor to profile in-situ metabolic information within stomach and explore their correlations during the genesis of disease, demonstrating its potential in facilitating clinical diagnosis.
Collapse
Affiliation(s)
- Yuqi Cheng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Lingjin Zhao
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yijing Wen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China; College of Biology, Hunan University, Changsha, 410082, China
| | - Zhen Ren
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Jiayu Zeng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Rui Shi
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Xinqi Cai
- Xiamen Institute of Standardization, Xiamen, 361012, China.
| | - Qian Dong
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China.
| | - Long Chen
- Faculty of Science and Technology, University of Macau, Taipa, 999078, Macau
| | - Changwei Lin
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China.
| |
Collapse
|
38
|
Chehayeb RJ, Odzer N, Albany RA, Ferrucci L, Sarpong D, Perez-Escamilla R, Lewis JB, Phipps AI, Meisner A, Pusztai L. Breastfeeding attributable fraction of triple negative breast cancer in the US. NPJ Breast Cancer 2025; 11:40. [PMID: 40328734 PMCID: PMC12055980 DOI: 10.1038/s41523-025-00755-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Rates of triple negative breast cancer (TNBC) are higher in Black women than in non-Hispanic White women. Breastfeeding duration and younger age at first birth are known risk factors for TNBC and vary by race. To quantify the contribution of these risk factors to disparities in TNBC, we calculated the population-attributable fraction (PAF). A PubMed search was performed to identify relevant studies and pooled odds ratios for breastfeeding for < 6 months and age at first birth < 25 years were calculated. PAF was calculated using the Levin formula. PAF of breastfeeding for < 6 months was 12% (95% confidence interval (CI) 5-20%) among White women and 15% (95%CI 3-26%) among Black women. We estimate that up to 15% of annual new TNBC in Black women and 12% in White women might be avoided by supporting breastfeeding. Policies supporting breastfeeding could hence reduce TNBC incidence and lessen racial disparities.
Collapse
Affiliation(s)
| | - Nicole Odzer
- Yale University School of Medicine, New Haven, CT, USA
| | - Roberta A Albany
- Cancer-in-the-Know, Mt Penn, PA, USA
- SWOG Clinical Trial Network, Seattle, WA, USA
| | | | - Daniel Sarpong
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Jessica B Lewis
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Amanda I Phipps
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Allison Meisner
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lajos Pusztai
- Yale University School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
39
|
Wang Z, Guo W, Zhang X, Wei Y, Zhang W, Du N, Li C, Wu X, Yi F, Zhou T, Dong X, Guo Q, Xu H, Wang E, Li N, Cheng R, Li Z, Song X, Sun Y, Sun X, Cao L. Tumor microenvironment-associated oxidative stress impairs SIRT1 secretion to suppress anti-tumor immune response. Cell Rep 2025; 44:115679. [PMID: 40343797 DOI: 10.1016/j.celrep.2025.115679] [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: 05/15/2024] [Revised: 11/10/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Sirtuin-1 (SIRT1) is a classical histone deacetylase well known for its roles in intracellular pathways such as energy metabolism, DNA damage response, and genome stability maintenance. We report that SIRT1 can be secreted into the tumor microenvironment (TME) through an unconventional protein secretion pathway, effectively inhibiting tumor growth. However, under the stressful conditions of the TME, SIRT1 undergoes increased methylation, which impedes its secretion. Consequently, tumor-infiltrating M2 macrophages are unable to acquire sufficient SIRT1 from the TME, resulting in a significant decrease in SIRT1 levels within these cells. This SIRT1 decline leads to elevated expression of programmed cell death ligand 1 (PD-L1) on M2 macrophages, which in turn contributes to CD8+ T cell exhaustion through the programmed cell death protein 1/PD-L1 interaction pathway. These findings unveil the multifaceted roles and regulatory mechanisms of SIRT1 within the complex TME, providing deeper insights that significantly enhance our understanding of tumor immune-evasion strategies.
Collapse
Affiliation(s)
- Zhuo Wang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Wendong Guo
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaowen Zhang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Yufei Wei
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China
| | - Wanying Zhang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Ning Du
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Chunlu Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xuan Wu
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Fei Yi
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Tingting Zhou
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiang Dong
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China
| | - Qiqiang Guo
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Hongde Xu
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Erli Wang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Na Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Rong Cheng
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Ziwei Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaoyu Song
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China.
| | - Yingxian Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning 110122, China.
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China.
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
40
|
Pinto-Marín Á, Trilla-Fuertes L, Miranda Poma J, Vasudev NS, García-Fernández E, López-Vacas R, Miranda N, Wilson M, López-Camacho E, Pertejo A, Dittmann A, Kunz L, Brown J, Pedroche-Just Y, Zapater-Moros A, de Velasco G, Castellano D, González-Peramato P, Espinosa E, Banks RE, Fresno Vara JÁ, Gámez-Pozo A. A prognostic microRNA-based signature for localized clear cell renal cell carcinoma: the Bio-miR study. Br J Cancer 2025:10.1038/s41416-025-03008-2. [PMID: 40335662 DOI: 10.1038/s41416-025-03008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Two thirds of renal cell carcinoma (RCC) patients have localized disease at diagnosis. A significant proportion of these patients will relapse. There is a need for prognostic biomarkers to improve risk-stratification and specific treatments for patients that relapse. The objective of this study is to determine the clinical utility of microRNA signatures as prognostic biomarkers in localized clear cell RCC (ccRCC) and propose new therapeutic targets in patients with a high-risk of relapse. PATIENTS AND METHODS The microRNA profiles from a discovery cohort of 71 T1-T2 ccRCC patients (n = 88) were analyzed using microarrays. MicroRNAs prognostic value was established, and a microRNAs signature predicting relapse for T1b-T3 disease was defined. Independent validation was carried out by qPCR in cohorts from UK (n = 75) and Spain (n = 180), and the TCGA cohort (n = 175). In the Spanish validation cohort, proteomics experiments were done. Proteins were extracted from FFPE tissue and analyzed using by data-independent acquisition mass spectrometry. Additionally, ccRCC TCGA RNA-seq data was also analyzed. Both protein and RNA-seq data was analyzed using Significance Analysis of Micorarrays (SAM) and probabilistic graphical models, which allow the identification of relevant biological processes between low and high-risk tumors. RESULTS A 9-microRNAs signature, Bio-miR, classified patients into low- and high-risk with disease-free survival (DFS) at 5 years of 87.12 vs. 54.17% respectively (p = 0.0086, HR = 3.58, 95%CI: 1.37-8.3). Results were confirmed in the validation cohorts with 5-year DFS rates of 94% vs. 62% in the UK cohort (HR = 7.14, p = 0.001), 82.9% vs. 58.7% in the Spanish cohort (HR = 2.46, p = 0.0013), and 5-year overall survival rates of 72.7% vs. 44.5% in the TCGA cohort (HR = 2.43, p = 0.0012). Among low-risk patients according to adjuvant immunotherapy clinical trial criteria, Bio-miR identified a high-risk group. Maybe those patients ought to be considered to receive adjuvant therapy. Proteins overexpressed in the high-risk group were mainly related to focal adhesion, serine and inositol metabolism, and angiogenesis. Probabilistic graphical models defined eight functional nodes related to specific biological processes. Differences between low- and high-risk tumors were detected in complement activation and translation functional nodes. In ccRCC TCGA cohort, 676 genes were differentially expressed between low and high-risk patients, mainly related to complement activation, adhesion, and chemokine and cytokine cascades. In this case, probabilistic graphical models defined ten functional nodes. Calcium binding, membrane, adhesion, extracellular matrix, blood microparticle, inflammatory response and immune response had higher functional node activity, and metabolism node, containing genes related to retinol and xenobiotic and CYP450 metabolism, had lower activity in the high-risk group. CONCLUSIONS Bio-miR dichotomizes ccRCC patients with non-metastatic disease into those with low- and high-risk of relapse. This has implications for treatment and follow-up, identifying patients most likely to benefit from adjuvant treatment in clinical trials, preventing unnecessary exposure to side-effects, and providing health economics benefits. Additionally, promising therapeutic targets, as angiogenesis, immune response, metabolism, or complement activation, were found deregulated in high-risk ccRCC patients defined by Bio-miR. These findings may be useful to select patients for tailored, molecularly-driven clinical trials. Identifying which patients with kidney cancer are most at risk of their cancer coming back after surgery is critical, so that they can be prioritized for early treatment. We have identified a combination of biomarkers present in the cancer tissue (called BiomiR) which can help to do this.
Collapse
Affiliation(s)
- Álvaro Pinto-Marín
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain.
| | | | - Jesús Miranda Poma
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
| | - Naveen S Vasudev
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | | | - Rocío López-Vacas
- Molecular Oncology Lab, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
| | - Natalia Miranda
- Urology Service, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Michelle Wilson
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | | | - Ana Pertejo
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
| | - Antje Dittmann
- Proteomics Group, Functional Genomics Center Zurich, Zurich, Switzerland
| | - Laura Kunz
- Proteomics Group, Functional Genomics Center Zurich, Zurich, Switzerland
| | - Joanne Brown
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | | | | | | | - Daniel Castellano
- Medical Oncology Service, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Enrique Espinosa
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Rosamonde E Banks
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | - Juan Ángel Fresno Vara
- Molecular Oncology Lab, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Angelo Gámez-Pozo
- Molecular Oncology Lab, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain.
- Biomedica Molecular Medicine SL, Madrid, Spain.
| |
Collapse
|
41
|
Liu R, Wang Y, Wu C, Wang W, Li X. A self-catabolic smart DNAzyme nanocapsule for amplified chemo-photodynamic therapy. J Mater Chem B 2025; 13:5334-5342. [PMID: 40223582 DOI: 10.1039/d4tb02608j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Violent degradation strategies of traditional nucleic acid hydrogels may bring adverse toxicity to complex biological systems when administered systemically due to uncontrolled digestion. Herein, an Mn2+-driven self-catabolic smart deoxyribozyme (DNAzyme) nanocapsule is developed for precise on-demand drug release to amplify cancer chemo-photodynamic therapy. Loaded manganese dioxide (MnO2) can generate oxygen (O2) to overcome tumor hypoxia and enhance photodynamic therapy, and a microRNA-21 (miR-21) antisense sequence can adsorb and clear intracellular miR-21 to amplify chemotherapy. The encoded DNAzymes and substrate sequences enable the programmable digestion of nucleic acid hydrogel carriers with Mn2+ ions as cofactors, so as to accurately deliver various therapeutic drugs. The results show that the smart nanocapsules can amplify chemo-photodynamic therapy by improving hypoxia in the tumor microenvironment and functional genes to kill tumor cells, which is expected to play an important role in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Rencun Liu
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| | - Yuanyuan Wang
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| | - Chen Wu
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| | - Weicai Wang
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| | - Xuemei Li
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| |
Collapse
|
42
|
Hamed M, Zayed BA, Mansour FR. A novel model for accurate and fast prediction of cancer incidence. BMC Public Health 2025; 25:1671. [PMID: 40329246 PMCID: PMC12053845 DOI: 10.1186/s12889-025-22624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Predicting cancer incidence has long been a challenge for clinicians and researchers. Accurate predictions are essential for health planning to ensure adequate resources for diagnosis, treatment, and rehabilitation. Current prediction methods rely on historical data, assuming persistent patterns of cancer incidence. METHOD In this study, the Google Trends tool was used to obtain the relative search volume index (RSVI) for the topic "cancer" each year from 2017 to 2023 in the United States and worldwide. The proposed model incorporated actual cancer incidence rates and yearly changes in RSVI. RESULTS The model was applied to predict the rates of new cancer cases in fifty American states over four consecutive years (2017, 2018, 2019, 2020). The selection of years was restricted with data availability. In most states, the percentage error did not exceed 6%. The high degree of similarity between the actual and predicted cancer incidence rates was notable. Similar results were obtained when predicting cancer incidence rates in the countries studied. CONCLUSION The model has successfully provided accurate short-term predictions of cancer incidence rates across all 50 American states and 54 countries since 2017.
Collapse
Affiliation(s)
- Mahmoud Hamed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Misr International University, Km 28 Ismailia Road, Cairo, 44971, Egypt
- MIU Chemistry Society (MIU-CS), Faculty of Pharmacy, Misr International University, Km 28 Ismailia Road, Cairo, 44971, Egypt
| | - Berlanty A Zayed
- Tanta Student Research Academy, Faculty of Medicine, Tanta University, Tanta, 31111, Egypt
| | - Fotouh R Mansour
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31111, Egypt.
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Egypt.
| |
Collapse
|
43
|
Ma H, Wang H, Sun R, Gan WJ, Ge JF. Facile Way to Differentiate Normal and Cancerous Tissues via Polarity-Sensitive Fluorescent Probes Based on 1,6-Naphthyridine Derivatives. Anal Chem 2025; 97:9447-9453. [PMID: 40267369 DOI: 10.1021/acs.analchem.5c00787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Cancer is a genetic disorder caused by the long-term interaction of many factors, which has become the most important factor to take away human health; therefore, it is essential to develop a more efficient and sensitive cancer detection technology. This study developed two polarity sensitive probes 1a and 1b based on a 1,6-naphthyridine moiety linked to different targeting groups by vinyl as the π bridge. As the solvent polarity decreased, the emission wavelength of probes 1a and 1b experienced a blue shift, resulting in a significant enhance in fluorescence intensity by 135-fold and 53-fold, respectively, and a good linear relationship between Fmax of probes 1a and 1b and Δf was established with high correlation coefficients. Furthermore, probes 1a and 1b exhibited large Stokes shifts, high photostability, and low cytotoxicity, successfully targeting intracellular lipid droplets and mitochondria. Fluctuation in polarity was detected by real-time changes in fluorescence intensity of probes in lipid droplets and mitochondria. Moreover, probe 1b was capable of real-time monitoring mitochondrial polarity during starvation or rapamycin-induced autophagy. It was worth standing out 1a and 1b could distinguish normal cells from cancer cells, and then the probes also were successfully applied for imaging to differentiate between human normal tissues and cancerous tissues, with the fluorescence intensity of malignant tumor tissues being 15.4-19.9 folds higher than that of normal tissues and 5.3-7.2 times higher than that of benign tumor tissues. Therefore, this research offers potential applications for cancer diagnosis.
Collapse
Affiliation(s)
- Huan Ma
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, No. 199 Ren Ai Road, Suzhou 215123, China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, No. 199 Ren Ai Road, Suzhou 215123, China
| | - Ru Sun
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, No. 199 Ren Ai Road, Suzhou 215123, China
| | - Wen-Juan Gan
- The Fourth Affiliated Hospital of Soochow University, Suzhou 215123, China
| | - Jian-Feng Ge
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, No. 199 Ren Ai Road, Suzhou 215123, China
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou 215163, China
| |
Collapse
|
44
|
Kim S, Jin HO, Jang SK, Ahn SH, Kim G, Kim H, Lee TG, Kim CH, Park IC. Iron overload enhances the susceptibility to cysteine deprivation-induced ferroptosis in non-small cell lung cancer cells. Med Oncol 2025; 42:201. [PMID: 40327117 DOI: 10.1007/s12032-025-02757-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Ferroptosis is an iron-dependent regulated cell death characterized by lipid peroxidation accumulation. Due to the high iron demand of cancer cells, targeting ferroptosis is considered a promising approach for cancer therapy. This study aimed to elucidate the mechanisms underlying the differences in ferroptosis sensitivity in non-small cell lung cancer (NSCLC) cells and identify strategies to overcome ferroptosis resistance. H1299 cells were more sensitive to cysteine deprivation-induced ferroptosis and exhibited higher transferrin receptor (TfR) expression than H460 cells. Transferrin enhanced ferroptosis in cysteine-deprived H1299 cells, while TfR knockdown reduced ferroptosis, suggesting the involvement of TfR/transferrin system in this process. In H460 cells with low TfR expression, transferrin treatment did not induce ferroptosis under cysteine deprivation, indicating that the TfR/transferrin system was not involved. However, treatment with cell-permeable ferric ammonium citrate increased the sensitivity of ferroptosis to cysteine deprivation or RSL3 treatment. In conclusion, iron overload could be a potential strategy to overcome ferroptosis resistance in NSCLC.
Collapse
Affiliation(s)
- Selim Kim
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea
- Department of Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hyeon-Ok Jin
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Se-Kyeong Jang
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Se Hee Ahn
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Gyeongmi Kim
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea
- Department of Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hyunggee Kim
- Department of Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Tae-Gul Lee
- Division of Pulmonology, Department of Internal Medicine, Korea Cancer Center Hospital, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea
| | - Cheol Hyeon Kim
- Division of Pulmonology, Department of Internal Medicine, Korea Cancer Center Hospital, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea.
| | - In-Chul Park
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Republic of Korea.
| |
Collapse
|
45
|
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] [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
|
46
|
Facilissimo I, Natoli G, Gaspari F, Comandone T, Bongiovanni D, Gollini P, Provenza C, Comandone A. The role of bone radiotherapy during immune checkpoint inhibitors treatment of non-small-cell lung cancer: a single-institution experience. Ther Adv Med Oncol 2025; 17:17588359251332451. [PMID: 40336632 PMCID: PMC12056327 DOI: 10.1177/17588359251332451] [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: 08/25/2023] [Accepted: 03/18/2025] [Indexed: 05/09/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) represent a keystone of cancer treatment, including non-small-cell lung cancer (NSCLC). Unfortunately, the efficacy of ICIs remains poor in patients with bone metastases from NSCLC. Recently, several case reports have suggested the clinical benefit of radiotherapy in advanced NSCLC patients. However, whether this positive effect is applicable during ICI treatment of NSCLC involving bones remains to be established. Methods We retrospectively reviewed the records of patients with bone metastases who received ICIs as monotherapy (anti-PD1 or anti-programmed death-ligand 1) as well as in combination with platinum-based-chemotherapy (carboplatin or cisplatin). We next analyzed the presence or the absence of radiotherapy targeting bone metastases (RT) among these patients during immunotherapy. Results A total of 40 patients were included in this study; among them, 10 (25%) received palliative RT for symptomatic bone metastases during cancer immunotherapy treatment with ICIs (RT group); the remaining 30 (75%) patients did not receive bone irradiation (Non-RT group). We observed that the RT group had a significantly longer overall survival (OS) than the Non-RT group, with a median survival of 16 months in the RT group versus 3 months in the Non-RT group (log-rank test p < 0.048; hazard ratio (HR) for OS = 0.44; 95% confidence interval (CI): 0.18-1.00). Similar results were observed with respect to progression-free survival (PFS; log-rank test p < 0.016; HR for PFS = 0.34; 95% CI: 0.15-1.00). Conclusion Our results suggest that radiotherapy to bone metastases may improve ICIs efficacy in patients with bone metastatic NSCLC.
Collapse
Affiliation(s)
- Ivan Facilissimo
- Division of Oncology, San Giovanni Bosco Hospital, Torino, Italy
| | - Guido Natoli
- Division of Oncology, San Giovanni Bosco Hospital, Piazza del Donatore di Sangue 3, Torino 10154, Italy
| | - Fabio Gaspari
- Division of Oncology, San Giovanni Bosco Hospital, Torino, Italy
| | | | - Diego Bongiovanni
- Division of Radiation Oncology, San Giovanni Antica Sede Hospital, Torino, Italy
| | - Paola Gollini
- Division of Radiology, San Giovanni Bosco Hospital, Torino, Italy
| | - Claudia Provenza
- Division of Pathology, San Giovanni Bosco Hospital, Torino, Italy
| | | |
Collapse
|
47
|
Pang X, Zhao Y, Chen X, Wang M, Chen X, Yuan H, Sun Y, Han J, Zhao X. Preclinical Evaluation and First-in-Human Study of [ 68Ga]Ga-αvβ6-2: A Novel Dimeric Integrin αvβ6-Targeted PET Probe for Pancreatic Cancer Imaging. Mol Pharm 2025; 22:2650-2659. [PMID: 40193102 DOI: 10.1021/acs.molpharmaceut.5c00051] [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: 04/24/2025]
Abstract
Integrin αvβ6 is highly expressed in pancreatic cancer, making it an ideal target for molecular imaging diagnosis. Multimerization is considered an effective strategy to increase the accumulation of molecular probes in tumors. Here, we synthesized monomeric and dimeric αvβ6-targeting molecular probes, labeled with 68Ga, and designated them [68Ga]Ga-αvβ6-1 and [68Ga]Ga-αvβ6-2, respectively. Both in vitro and in vivo studies were conducted using human pancreatic cancer BxPC-3 cells and BxPC-3 tumor-bearing mice. Additionally, positron emission tomography/computed tomography (PET/CT) imaging with [68Ga]Ga-αvβ6-2 was performed in three patients with pancreatic cancer. In vitro studies demonstrated that [68Ga]Ga-αvβ6-2 exhibited greater binding affinity, cellular uptake, and internalization than did [68Ga]Ga-αvβ6-1. Micro-PET/CT imaging and biodistribution studies revealed the superior imaging performance of [68Ga]Ga-αvβ6-2. Furthermore, the first-in-human evaluation highlighted the favorable in vivo distribution and diagnostic efficacy of [68Ga]Ga-αvβ6-2 in pancreatic cancer. These results underscore the effectiveness of the multimerization strategy in the application of αvβ6-targeted molecular probes, suggesting that [68Ga]Ga-αvβ6-2 may possess favorable clinical translation potential.
Collapse
Affiliation(s)
- Xiao Pang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Medical Imaging College, North Sichuan Medical College; Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yan Zhao
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Mengjiao Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaoshan Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Huiqing Yuan
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Yuhan Sun
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei 050011, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei 050011, China
| |
Collapse
|
48
|
Maratta MG, Sparagna I, Occhipinti D, Roca L, Sgambato M, Raia S, Bianchi A, Chiloiro S, Rossi E, Rindi G, Tortora G, Schinzari G. Upfront Oxaliplatin-Fluoropyrimidine Chemotherapy and Somatostatin Analogues in Advanced Well-Differentiated Gastro-Entero-Pancreatic Neuroendocrine Tumors. Cancers (Basel) 2025; 17:1561. [PMID: 40361487 PMCID: PMC12071586 DOI: 10.3390/cancers17091561] [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: 04/01/2025] [Revised: 04/24/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
(1) Background: GEP-NETs are frequently diagnosed at advanced stage. For well-differentiated somatostatin receptor-positive (SSTR+) NETs, SSA are the preferred first-line therapy. However, in newly diagnosed patients with G2/G3 and a high tumor burden, SSA alone might not be enough; (2) Methods: We conducted a retrospective analysis to assess the effectiveness of combining oxaliplatin-fluoropyrimidine chemotherapy with SSA as an upfront strategy in newly diagnosed metastatic G2/G3 GEP-NET patients treated with oxaliplatin-fluoropyrimidine-based chemotherapy; (3) Results: Between March 2017 and October 2023, 32 pts (19 males, 13 females; M:F = 1.5:1; median age 54 years, range 31-82) were deemed eligible to receive oxaliplatin-fluoropyrimidine chemotherapy in addition to SSA; 14 received XELOX and 18 FOLFOX. After a median follow-up of 26 mo., each patient had completed at least two cycles of chemotherapy. The ORR was 25%, with a median DoR of 21.3 mo. The DCR was 87.5%. Notably, 28.1% of patients experienced tumor shrinkage sufficient for radical surgery on residual tumor lesions, encompassing both primary tumors and metastases; (4) Conclusions: Upfront treatment with the combination of oxaliplatin-fluoropyrimidine and SSA demonstrated effectiveness and safety. This approach may be considered to facilitate conversion surgery in eligible patients.
Collapse
Affiliation(s)
- Maria Grazia Maratta
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ileana Sparagna
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Denis Occhipinti
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Luigi Roca
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Margherita Sgambato
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Salvatore Raia
- Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
| | - Antonio Bianchi
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
| | - Sabrina Chiloiro
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
| | - Ernesto Rossi
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
| | - Guido Rindi
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli—IRCCS, 00168 Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giovanni Schinzari
- Medical Oncology Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
49
|
Aguayo F, Tapia JC, Calaf GM, Muñoz JP, Osorio JC, Guzmán-Venegas M, Moreno-León C, Levican J, Andrade-Madrigal C. The Role of Xenobiotics and Anelloviruses in Colorectal Cancer: Mechanisms and Perspectives. Int J Mol Sci 2025; 26:4354. [PMID: 40362591 PMCID: PMC12072659 DOI: 10.3390/ijms26094354] [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: 02/24/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Xenobiotics are non-natural chemical compounds to which the human population is exposed. Chronic exposure to certain xenobiotics is associated with various diseases, including cancer development. Anelloviruses (AVs), including Torque Teno Virus (TTV), Torque Teno Mini Virus (TTMV), and Torque Teno Midi Virus (TTMDV), are ubiquitous viruses found in the general population. As no disease has been definitively associated with AVs, they are sometimes referred to as "viruses awaiting a disease". This review explores the potential roles of xenobiotics and AVs in colorectal cancer (CRC) development and suggests a potential interplay between them. Evidence suggests an association between certain xenobiotics (like pesticides, cigarette smoke components, and dietary factors) and CRC, while such an association is less clear for AVs. The high prevalence of AVs suggests these infections alone may be insufficient to disrupt homeostasis; thus, additional factors might be required to promote disease, potentially including cancer.
Collapse
Affiliation(s)
- Francisco Aguayo
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Julio C. Tapia
- Laboratorio de Transformación Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Santiago 8380453, Chile
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile;
| | - Julio C. Osorio
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Matías Guzmán-Venegas
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Carolina Moreno-León
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Jorge Levican
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Cristian Andrade-Madrigal
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| |
Collapse
|
50
|
Tersigni C, Onori M, Buzzonetti A, Ferrante A, Fattorossi A, Battaglia A, Corrado G, Neri C, Beneduce G, Sannino F, Petrecca A, Scambia G, Fagotti A. Diagnostic performance of circulating EpCAM+ and CD45+ extracellular vesicles in platinum-sensitivity in high-grade serous ovarian cancer: a pilot study. J Ovarian Res 2025; 18:93. [PMID: 40317028 PMCID: PMC12046641 DOI: 10.1186/s13048-025-01656-9] [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: 10/17/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Platinum-resistant high-grade serous ovarian cancer (HGSOC) is a fatal disease. The main goal of current study is to develop new strategies to predict platinum resistance, moving towards personalized therapy. Currently, there are no validated biomarkers able to predict at diagnosis platinum resistance. Circulating tumor-derived extracellular vesicles (EVs) represent a liquid biopsy of the tumor of origin and reflect its biological profile. EpCAM-expressing EVs are largely used biomarkers of epithelial cancers in translational research. Aim of this study was to quantify, by nano-flow cytometry, circulating EpCAM+ EVs in patients with histologically confirmed diagnosis of HGSOC FIGO stage III/IV, before and after intravenous administration of the first dose of chemotherapy with paclitaxel and carboplatin, and correlate EVs levels to platinum-sensitivity. As comparison, leukocyte-derived EVs were also assessed using the pan-leukocyte marker CD45. RESULTS Patients with platinum-sensitive HGSOC showed significantly lower pre-chemotherapy circulating levels of both EpCAM+ and CD45+ EVs compared to platinum-resistant cases (p<0.01). Platinum-sensitive patients displayed significantly higher levels of circulating EpCAM+ and CD45+ EVs 21 days post administration of the first dose of chemotherapy compared to pre-treatment levels (p<0.05 and p<0.01, respectively). Platinum-resistant and platinum-refractory patients showed significantly higher EpCAM+CD45+ EVs levels than platinum-sensitive patients (p<0.01, p<0.05, respectively). CONCLUSIONS Tumor-derived EVs are valuable candidate biomarkers for early prediction of platinum resistance and need to be investigated in larger prospective clinical studies.
Collapse
Affiliation(s)
- Chiara Tersigni
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy.
| | - Marianna Onori
- Università Cattolica del Sacro Cuore, L.go F.Vito 1, Rome, 00168, Italy
| | - Alexia Buzzonetti
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy
| | - Arianna Ferrante
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy
| | - Andrea Fattorossi
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy
| | | | - Giacomo Corrado
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy
| | - Caterina Neri
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy
| | - Giuliana Beneduce
- Università Cattolica del Sacro Cuore, L.go F.Vito 1, Rome, 00168, Italy
| | - Fabio Sannino
- Università Cattolica del Sacro Cuore, L.go F.Vito 1, Rome, 00168, Italy
| | | | - Giovanni Scambia
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy
- Università Cattolica del Sacro Cuore, L.go F.Vito 1, Rome, 00168, Italy
| | - Anna Fagotti
- Fondazione Policlinico Universitario A.Gemelli IRCCS, L.go A.Gemelli 8, Rome, 00168, Italy
- Università Cattolica del Sacro Cuore, L.go F.Vito 1, Rome, 00168, Italy
| |
Collapse
|