1
|
Deng B, Wang K, He H, Xu M, Li J, He P, Liu Y, Ma J, Zhang J, Dong W. Biochanin A mitigates colitis by inhibiting ferroptosis-mediated intestinal barrier dysfunction, oxidative stress, and inflammation via the JAK2/STAT3 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156699. [PMID: 40215818 DOI: 10.1016/j.phymed.2025.156699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease marked by intestinal barrier dysfunction, oxidative stress, and inflammation. Biochanin A (BCA) is a natural flavonoid found in various plants, known for its anti-inflammatory, antioxidant, and anticancer properties, while its therapeutic role in UC and potential mechanism remains unexplored. PURPOSE To explores the therapeutic potential of BCA in alleviating UC, focusing on its effects on ferroptosis and the JAK2/STAT3 signaling pathway. METHODS The BCA's therapeutic effects on Dextran sulfate sodium (DSS)-induced colitis model in C57BL/6J mice was investigated. Subsequently, a comprehensive range of techniques was performed to investigate the impact of BCA on intestinal barrier integrity, oxidative stress, inflammation. Besides, the RNA sequencing was performed to explore the potential mechanism. The role of ferroptosis inhibition in BCA's effects in vitro and in vivo was explored by co-treating with the ferroptosis activator Erastin. RESULTS Treatment of colitis mice with BCA significantly improved DAI scores and histopathological damage scores, reduced oxidative stress, enhanced intestinal barrier function, and suppress inflammatory responses. RNA sequencing result found that BCA could lead to the inhibition of ferroptosis in mice colon tissue. Moreover, erastin co-treatment negated BCA's effects in vitro and vivo. Mechanistically, BCA exerts these effects by suppressing the JAK2/STAT3 pathway, which plays a pivotal role in ferroptosis and inflammation. Molecular docking studies further confirm the direct binding of BCA to both GPX4 and STAT3. CONCLUSION These results establish BCA as a promising natural compound for UC treatment, offering a novel therapeutic strategy by specifically targeting ferroptosis and its associated molecular pathways, thereby addressing key gaps in current UC management and advancing potential clinical applications.
Collapse
Affiliation(s)
- Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kunpeng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Laboratory of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Haodong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Miao Xu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Jiao Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pengzhan He
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yinghui Liu
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Ma
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jixiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Ren M, Sun X, Lin J, Kan Y, Lu S, Zhang X, Liang R, Wang B, Chen H, Wu Y, Luan X. Hypoxia-responsive oncolytic conjugate triggers type-II immunogenic cell death for enhanced photodynamic immunotherapy. J Control Release 2025; 382:113717. [PMID: 40258475 DOI: 10.1016/j.jconrel.2025.113717] [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: 02/08/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Immunogenic cell death (ICD) induced by photodynamic therapy (PDT) holds great promise for enhancing anti-tumor immunotherapy; however, its clinical efficacy is often hampered by suboptimal ICD induction and the exacerbation of an immunosuppressive tumor microenvironment (TME) following PDT. Herein, we present a tumor-targeted and hypoxia-responsive peptide-photosensitizer conjugate, A6-dMP-VP, which integrates an oncolytic peptide (dMP) with a CD44-targeting motif (A6), hypoxia-responsive groups, and the photosensitizer verteporfin (VP). Following systemic administration, A6-dMP-VP preferentially accumulates in 4T1 tumors, where the hypoxic TME triggers its response. Remarkably, the combined oncolytic activity and PDT effect of A6-dMP-VP effectively induce type-II ICD via mitochondrial disruption and endoplasmic reticulum stress, leading to robust antigen release. This process significantly enhances dendritic cell maturation and cytotoxic T cell priming, ultimately achieving potent suppression of both primary and metastatic tumors. Our findings establish A6-dMP-VP as a highly effective type-II ICD inducer, offering a novel strategy to overcome the limitations of PDT and advance photodynamic-oncolytic immunotherapy.
Collapse
Affiliation(s)
- Maomao Ren
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin Sun
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiayi Lin
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yajie Kan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangdong 510006, China
| | - Shengxin Lu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaokun Zhang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rui Liang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bei Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongzhuan Chen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ye Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
3
|
Yang L, He S, Tang C, Shi L, Liu Z, Tian Z, Li H, He X, Liao J, Liu Y. Synthesis of porphyrin-formononetin derivatives and their anti-tumor activity studies. Mol Divers 2025:10.1007/s11030-025-11183-w. [PMID: 40202554 DOI: 10.1007/s11030-025-11183-w] [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: 11/20/2024] [Accepted: 03/30/2025] [Indexed: 04/10/2025]
Abstract
Photodynamic therapy (PDT) has received much attention in cancer treatment because of its low toxicity and side effects. In this study, we successfully synthesized 14 novel porphyrin-formononetin derivatives. In reactive oxygen species detection experiments, the target compounds 4a-6d caused a significant decrease in the fluorescence intensity of DPBF compared with the porphyrin parent and formononetin feedstock after illumination, and it was found that the target compound had a higher ROS quantum yield, among which the quantum yield of compound 6c was higher. In the in vitro anti-tumor activity assay, the target compounds 4a-6d exhibited a certain degree of growth inhibition against six cancer cells (A549, MDA-MB-231, HCT-116, HGC-27, DU145, and TCCSUP) under light conditions, whereas the cytotoxicity of the target compounds against the normal cells H9c2 was less. The results of the scratch assay showed that 6c could inhibit the growth of tumor cells by inhibiting the migration of DU145 cells. The experimental results indicate that the target compounds achieve the synergistic effect of PDT and chemotherapy.
Collapse
Affiliation(s)
- Lingyan Yang
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, 421001, Hunan, China
| | - Shan He
- College of Mathematics and Physics, University of South China, Hengyang, 421001, Hunan, China
| | - Chen Tang
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, 421001, Hunan, China
| | - Lei Shi
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, 421001, Hunan, China
| | - Zhenhua Liu
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, 421001, Hunan, China
| | - Zejie Tian
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Li
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, 421001, Hunan, China
| | - Xufeng He
- Health School of Nuclear Industry, Nanhua Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Jiding Liao
- College of Mathematics and Physics, University of South China, Hengyang, 421001, Hunan, China.
| | - Yunmei Liu
- Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
4
|
Qiao W, Li S, Luo L, Chen M, Zheng X, Ye J, Liang Z, Wang Q, Hu T, Zhou L, Wang J, Ge X, Feng G, Hu F, Liu R, Li J, Yang J. Ce6-GFFY is a novel photosensitizer for colorectal cancer therapy. Genes Dis 2025; 12:101441. [PMID: 39759121 PMCID: PMC11697048 DOI: 10.1016/j.gendis.2024.101441] [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: 10/17/2023] [Revised: 07/10/2024] [Accepted: 10/21/2024] [Indexed: 01/07/2025] Open
Abstract
Photodynamic therapy is an "old" strategy for cancer therapy featuring clinical safety and rapid working, but suitable photosensitizers for colorectal cancer therapy remain lacking. This study synthesized a novel photosensitizer termed Ce6-GFFY based on a self-assembling peptide GFFY and a photo-responsive molecule chlorin e6 (Ce6). Ce6-GFFY forms macroparticles with a diameter of ∼160 nm and possesses a half-life of 10 h, as well as an ideal tumor-targeting ability in mouse models. Ce6-GFFY effectively penetrates cells and generates numerous reactive oxygen species upon 660 nm laser irradiation. The reactive oxygen species promotes the accumulation of cytotoxic T cells and decrease of myeloid-derived suppressor cells in the tumor microenvironment through immunogenic cell death, thus prohibiting the growth of both primary and metastatic tumors after once treatment. This study not only provides a strategy for photosensitizer development but also confirms a promising application of Ce6-GFFY for colorectal cancer therapy.
Collapse
Affiliation(s)
- Wei Qiao
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Shuxin Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Linna Luo
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Meiling Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Xiaobin Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jiacong Ye
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Zhaohui Liang
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Qiaoli Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Ting Hu
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Ling Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jing Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Xiaosong Ge
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Guokai Feng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Fang Hu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Rongbin Liu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Jianjun Li
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jie Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| |
Collapse
|
5
|
Lee JH, Yang SB, Park SJ, Kweon S, Ma G, Seo M, Kim HR, Kang TB, Lim JH, Park J. Cell-Penetrating Peptide Like Anti-Programmed Cell Death-Ligand 1 Peptide Conjugate-Based Self-Assembled Nanoparticles for Immunogenic Photodynamic Therapy. ACS NANO 2025; 19:2870-2889. [PMID: 39761412 DOI: 10.1021/acsnano.4c16128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The tumor-specific efficacy of the most current anticancer therapeutic agents, including antibody-drug conjugates (ADCs), oligonucleotides, and photosensitizers, is constrained by limitations such as poor cell penetration and low drug delivery. In this study, we addressed these challenges by developing, a positively charged, amphiphilic Chlorin e6 (Ce6)-conjugated, cell-penetrating anti-PD-L1 peptide nanomedicine (CPPD1) with enhanced cell and tissue permeability. The CPPD1 molecule, a bioconjugate of a hydrophobic photosensitizer and strongly positively charged programmed cell death-ligand 1 (PD-L1) binding cell-penetrating peptide (CPP), is capable of self-assembling into nanoparticles with an average size of 199 nm in aqueous solution without the need for any carriers. These carrier-free nanoparticles possess the ability to penetrate the cell membrane of cancer cells and target tumors expressing PD-L1 on their surface. Notably, CPPD1 nanoparticles effectively blocked programmed cell death-1 (PD-1)/PD-L1 interactions and reduced PD-L1 expression via lysosomal degradation. They also demonstrated the responsiveness of CPPD1 nanoparticles in photodynamic therapy (PDT) to a 635 nm laser, leading to the generation of ROS, and induction of various immunogenic cell deaths (ICD). Highly penetrating CPPD1 nanoparticles could immunogenically modulate the microenvironment of CT26 cancer and were also effective in treating abscopal metastatic tumors, addressing major limitations of traditional PDT.
Collapse
Affiliation(s)
- Jun-Hyuck Lee
- BK21 Program, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Seong-Bin Yang
- BK21 Program, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Seong Jin Park
- Department of Research, Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Gaeun Ma
- BK21 Program, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Minho Seo
- BK21 Program, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Ha Rin Kim
- School of Medicine, Stanford University, Stanford, California 94305, United States
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Tae-Bong Kang
- BK21 Program, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Ji-Hong Lim
- BK21 Program, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Jooho Park
- BK21 Program, Department of Applied Life Science, Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
6
|
Fiegler-Rudol J, Zięba N, Turski R, Misiołek M, Wiench R. Hypericin-Mediated Photodynamic Therapy for Head and Neck Cancers: A Systematic Review. Biomedicines 2025; 13:181. [PMID: 39857765 PMCID: PMC11759840 DOI: 10.3390/biomedicines13010181] [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: 12/22/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Conventional treatments for cancers of the head and neck region are often associated with high recurrence rates and impaired quality of life. Photodynamic therapy (PDT) has emerged as a promising alternative, leveraging photosensitizers such as hypericin to selectively target tumour cells with minimal damage to surrounding healthy tissues. Objectives: We aimed to evaluate the efficacy and underlying mechanisms of hypericin-mediated PDT (HY-PDT) in treating head and neck cancers. Methods: Adhering to PRISMA 2020 guidelines, a systematic search was conducted across PubMed/Medline, Embase, Scopus, and the Cochrane Library for studies published between January 2000 and December 2024. Inclusion criteria encompassed preclinical in vitro and in vivo studies and clinical trials focusing on HY-PDT for head and neck malignancies and its subtypes. Results: A total of 13 studies met the inclusion criteria, comprising both in vitro and in vivo investigations. HY-PDT consistently demonstrated significant cytotoxicity against squamous cell carcinoma cells through apoptotic and necrotic pathways, primarily mediated by ROS generation. Hypericin exhibited selective uptake in cancer cells over normal keratinocytes. Additionally, HY-PDT modulated the tumour microenvironment by altering cytokine profiles, such as by increasing IL-20 and sIL-6R levels, which may enhance antitumor immunity and reduce metastasis. Conclusions: HY-PDT emerges as a highly promising and minimally toxic treatment modality for head and neck cancers, demonstrating efficacy in inducing selective tumour cell death and modulating the immune microenvironment. Despite the encouraging preclinical evidence, significant methodological variability and limited clinical data necessitate further large-scale, standardized and randomized controlled trials.
Collapse
Affiliation(s)
- Jakub Fiegler-Rudol
- Department of Periodontal Diseases and Oral Mucosa Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Natalia Zięba
- Department of Otorhinolaryngology and Laryngological Oncology in Zabrze, Medical University of Silesia, 41-800 Zabrze, Poland (M.M.)
| | - Radosław Turski
- Individual Dental Practice, Ul. Żarecka 128, 42-208 Częstochowa, Poland;
| | - Maciej Misiołek
- Department of Otorhinolaryngology and Laryngological Oncology in Zabrze, Medical University of Silesia, 41-800 Zabrze, Poland (M.M.)
| | - Rafał Wiench
- Department of Periodontal Diseases and Oral Mucosa Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| |
Collapse
|
7
|
Xu W, Jian D, Yang H, Wang W, Ding Y. Aggregation-induced emission: Application in diagnosis and therapy of hepatocellular carcinoma. Biosens Bioelectron 2024; 266:116722. [PMID: 39232431 DOI: 10.1016/j.bios.2024.116722] [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: 04/30/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Hepatocellular carcinoma (HCC) is a serious health issue due to its low early diagnosis rate, resistance to chemotherapy, and poor five-year survival rate. Therefore, it is crucial to explore novel diagnostic and therapeutic approaches tailored to the characteristics of HCC. Aggregation-induced emission (AIE) is a phenomenon where the luminescence of certain molecules, typically non-luminescent or weakly luminescent in solution, is significantly enhanced upon aggregation. AIE has been extensively applied in bioimaging, biosensors, and therapy. Fluorophore materials based on AIE (AIEgens) have a wide range of application scenarios and potential for clinical translation. This review focuses on recent advances in AIE-based strategies for diagnosing and treating HCC. First, the specific functional mechanism of AIE is described. Next, we summarize recent progress in the application of AIE for multimodal imaging, biosensor detection, and phototherapy. Finally, prospects and challenges for the AIE-based application in the diagnosis and therapy of HCC are discussed.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Danfeng Jian
- MOE Key Laboratory of Advanced Textile Materials & Manufacturing Technology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Weili Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
8
|
Machuca A, Peñalver GA, Garcia RAF, Martinez-Lopez A, Castillo-Lluva S, Garcia-Calvo E, Luque-Garcia JL. Advancing rhodium nanoparticle-based photodynamic cancer therapy: quantitative proteomics and in vivo assessment reveal mechanisms targeting tumor metabolism, progression and drug resistance. J Mater Chem B 2024; 12:12073-12086. [PMID: 39453320 DOI: 10.1039/d4tb01631a] [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: 10/26/2024]
Abstract
Rhodium nanoparticles have been recently discovered as good photosensitizers with great potential in cancer photodynamic therapy by effectively inducing cytotoxicity in cancer cells under near-infrared laser. This study evaluates the molecular mechanisms underlying such antitumoral effect through quantitative proteomics. The results revealed that rhodium nanoparticle-based photodynamic therapy disrupts tumor metabolism by downregulating key proteins involved in ATP synthesis and mitochondrial function, leading to compromised energy production. The treatment also induces oxidative stress and apoptosis while targeting the invasion capacity of cancer cells. Additionally, key proteins involved in drug resistance are also affected, demonstrating the efficacy of the treatment in a multi-drug resistant cell line. In vivo evaluation using a chicken embryo model also confirmed the effectiveness of the proposed therapy in reducing tumor growth without affecting embryo viability.
Collapse
Affiliation(s)
- Andres Machuca
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Gabriel A Peñalver
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | | | - Angelica Martinez-Lopez
- Department Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Sonia Castillo-Lluva
- Department Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Estefania Garcia-Calvo
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Jose L Luque-Garcia
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
9
|
Canti G, Rapozzi V. PDT and antitumor immunity: the beginnings of the story. Photochem Photobiol Sci 2024:10.1007/s43630-024-00627-1. [PMID: 39235681 DOI: 10.1007/s43630-024-00627-1] [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: 07/09/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024]
Abstract
This mini-review reports a brief description of the first experiments conducted by Canti's group on the role of photodynamic therapy in generating immunity against cancer. It highlights for the first time the effective role of PDT in the induction of anti-tumor T lymphocytes and shows that this effect is tumor-specific. It has also been reported how this adoptive immunity can improve the efficacy of chemotherapy. These studies have helped to open an important new field of scientific research on the role of PDT-generated immunity and to stimulate today's important new pre-clinical approaches.
Collapse
Affiliation(s)
- Gianfranco Canti
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20129, Milan, Italy.
| | | |
Collapse
|
10
|
Mahdizade Ari M, Amirmozafari N, Atieh Darbandi, Afifirad R, Asadollahi P, Irajian G. Effectiveness of photodynamic therapy on the treatment of chronic periodontitis: a systematic review during 2008-2023. Front Chem 2024; 12:1384344. [PMID: 38817441 PMCID: PMC11138352 DOI: 10.3389/fchem.2024.1384344] [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: 02/09/2024] [Accepted: 04/10/2024] [Indexed: 06/01/2024] Open
Abstract
Objective This study investigated the effect of photodynamic therapy on chronic periodontitis patients and then evaluated the microbial, immunological, periodontal, and clinical outcomes. The significant effects of photodynamic therapy obtained by in vitro and in vivo studies have made it a popular treatment for periodontal diseases in recent years. Photodynamic therapy is a novel bactericidal strategy that is stronger, faster, and less expensive than scaling and root planing. Method This study registered on PROSPERO (CRD42021267008) and retrieved fifty-three randomized controlled trials by searching nine databases (Medline, Embase, Scopus, Open Gray, Google Scholar, ProQuest, the Cochrane Library, Web of Science, and ClinicalTrials.gov) from 2008 to 2023. Of 721 records identified through database searches following title and full-text analysis, and excluding duplicate and irrelevant publications, 53 articles were included in this systematic review. Fifty of the 53 eligible studies fulfilled all the criteria in the Joanna Briggs Institute's (JBI's) Checklist for RCTs; the remaining articles met 9-12 criteria and were considered high quality. Results The present study showed that photodynamic therapy in adjunct to scaling and root planing has the potential to improve periodontal parameters such as clinical attachment loss or gain, decrease in bleeding on probing, and probing pocket depth. In addition, photodynamic therapy decreases the rate of periodontal pathogens and inflammation markers, which, in turn, reduces the progression of periodontitis. Conclusion Photodynamic therapy is considered a promising, adjunctive, and low-cost therapeutic method that is effective in tissue repair, reducing chronic periodontitis, reducing inflammation, and well-tolerated by patients.
Collapse
Affiliation(s)
- Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, University of Medical Sciences, Tehran, Iran
| | - Nour Amirmozafari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atieh Darbandi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, University of Medical Sciences, Tehran, Iran
| | - Roghayeh Afifirad
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Asadollahi
- Department of Microbiology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Naganawa R, Zhao H, Takano Y, Maeki M, Tokeshi M, Harashima H, Yamada Y. Investigation of the Nanoparticulation Method and Cell-Killing Effect following the Mitochondrial Delivery of Hydrophobic Porphyrin-Based Photosensitizers. Int J Mol Sci 2024; 25:4294. [PMID: 38673875 PMCID: PMC11050504 DOI: 10.3390/ijms25084294] [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: 02/28/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Photodynamic therapy is expected to be a less invasive treatment, and strategies for targeting mitochondria, the main sources of singlet oxygen, are attracting attention to increase the efficacy of photodynamic therapy and reduce its side effects. To date, we have succeeded in encapsulating the photosensitizer rTPA into MITO-Porter (MP), a mitochondria-targeted Drug Delivery System (DDS), aimed at mitochondrial delivery of the photosensitizer while maintaining its activity. In this study, we report the results of our studies to alleviate rTPA aggregation in an effort to improve drug efficacy and assess the usefulness of modifying the rTPA side chain to improve the mitochondrial retention of MITO-Porter, which exhibits high therapeutic efficacy. Conventional rTPA with anionic side chains and two rTPA analogs with side chains that were converted to neutral or cationic side chains were encapsulated into MITO-Porter. Low-MP (MITO-Porter with Low Drug/Lipid) exhibited high drug efficacy for all three types of rTPA, and in Low-MP, charged rTPA-encapsulated MP exhibited high drug efficacy. The cellular uptake and mitochondrial translocation capacities were similar for all particles, suggesting that differences in aggregation rates during the incorporation of rTPA into MITO-Porter resulted in differences in drug efficacy.
Collapse
Grants
- 23H00541 Ministry of Education, Culture, Sports, Science and Technology
- 21H01753 Ministry of Education, Culture, Sports, Science and Technology
- 21K19928 Ministry of Education, Culture, Sports, Science and Technology
- Special Education and Research Expenses Ministry of Education, Culture, Sports, Science and Technology
- JPMJFR203X Japan Science and Technology Agency
Collapse
Affiliation(s)
- Rina Naganawa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hanjun Zhao
- Graduate School of Environmental Science, Hokkaido University, Sapporo 060-0810, Japan (Y.T.)
| | - Yuta Takano
- Graduate School of Environmental Science, Hokkaido University, Sapporo 060-0810, Japan (Y.T.)
- Research Institute for Electronic Science, Hokkaido University, Sapporo 010-0020, Japan
| | - Masatoshi Maeki
- Graduate School of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Manabu Tokeshi
- Graduate School of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Fusion Oriented Research for Disruptive Science and Technology (FOREST) Program, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| |
Collapse
|
12
|
Fan W, Tang J, Tang S, Lin Z, Li M, Zhang Z, Wu D. Bibliometric analysis of photodynamic therapy and immune response from 1989-2023. Front Pharmacol 2024; 15:1299253. [PMID: 38288443 PMCID: PMC10822948 DOI: 10.3389/fphar.2024.1299253] [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: 09/22/2023] [Accepted: 01/03/2024] [Indexed: 01/31/2024] Open
Abstract
Objective: Photodynamic therapy (PDT) is a minimally invasive treatment approach for precancerous and cancerous lesions, known for its ability to activate the host immune response. This study conducted a bibliometric analysis to identify the research trends and hotspots related to the immune response in PDT. Methods: We analyzed articles and reviews published from 1989 to 2023, retrieved from the Web of Science database. Using Citespace and VOSviewer, we visualized the distribution patterns of these studies in time and space. Results: The analysis revealed a substantial increase in the number of publications on PDT-related immune response since 1989. A total of 1,688 articles from 1,701 institutions were included in this analysis. Among thei nstitutions, the Chinese Academy of Sciences demonstrated exceptional productivity and a willingness to collaborate with others. Additionally, 8,567 authors contributed to the field, with Mladen Korbelik, Michael R. Hamblin, and Wei R. Chen being the most prolific contributors. The current research focus revolves around novel strategies to enhance antitumor immunity in PDT, including PDT-based dendritic cell vaccines, combination therapies with immune checkpoint inhibitors (ICIs), and the use of nanoparticles for photosensitizer delivery. Furthermore, genes such as CD8A, TNF, CD4, IFNG, CD274, IL6, IL10, CALR, HMGB1, and CTLA4 have been evaluated in the context of PDT-related immunity. Conclusion: PDT not only achieves tumor ablation but also stimulates the immune response, bolstering antitumor immunity. This study highlights the emerging hotspots in PDT-related immune response research and provides valuable insights for future investigations aimed at further enhancing antitumor immunity.
Collapse
Affiliation(s)
- Wanting Fan
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Jianming Tang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Su Tang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Zhengshen Lin
- Department of Stomatology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Mohan Li
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Zheng Zhang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Donglei Wu
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| |
Collapse
|
13
|
Xu H, Wu C, Wang D, Wang H. Alleviating effect of Nexrutine on mucosal inflammation in mice with ulcerative colitis: Involvement of the RELA suppression. Immun Inflamm Dis 2024; 12:e1147. [PMID: 38270298 PMCID: PMC10797652 DOI: 10.1002/iid3.1147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Nexrutine is an herbal extract derived from Phellodendron amurense, known for its anti-inflammatory, antidiarrheal, and hemostatic properties. However, its effect on ulcerative colitis (UC) remains unclear. METHODS A mouse model of UC was induced by 3% dextran sulfate sodium, while human colonic epithelial cells NCM-460 were exposed to lipopolysaccharide. Both models were treated with Nexrutine at 300 or 600 mg/kg, with Mesalazine applied as a positive control regimen. The disease activity index (DAI) of mice was calculated, and the pathological injury scores were assessed through hematoxylin and eosin staining. The viability of NCM-460 cells was determined using the CCK-8 method. Inflammatory cytokines were detected using ELISA kits. Expression of mucin 3 (MUC3), Claudin-1, and tight junction protein (ZO-1) was detected to analyze mucosal barrier integrity. Target genes of Nexrutine were predicted using bioinformatics tools. Expression of RELA proto-oncogene (RELA) was analyzed using qPCR and western blot assays. RESULTS The Nexrutine treatments significantly alleviated DAI of mice, mitigated pathological changes in their colon tissues, decreased the production of pro-inflammatory cytokines, enhanced the barrier integrity-related proteins, and increased NCM-460 cell viability in vitro. RELA, identified as a target gene of Nexrutine, showed elevated levels in UC models but was substantially suppressed by Nexrutine treatment. Adenovirus-mediated RELA upregulation in mice or the overexpression plasmid of RELA in cells counteracted the effects of Nexrutine treatments, exacerbating UC-related symptoms. CONCLUSION This study demonstrates that Nexrutine alleviates inflammatory mucosal barrier damage in UC by suppressing RELA transcription.
Collapse
Affiliation(s)
- Hongyun Xu
- Graduate SchoolHeilongjiang University of Chinese MedicineHarbinHeilongjiangChina
| | - Chunyu Wu
- Department of Continuing EducationFirst Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinHeilongjiangChina
| | - Danning Wang
- Graduate SchoolHeilongjiang University of Chinese MedicineHarbinHeilongjiangChina
| | - Haiqiang Wang
- Department of Liver, Spleen and StomachFirst Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinHeilongjiangChina
| |
Collapse
|