1
|
Zhu J, Zhu X, Su T, Zhou H, Wang S, Shi W. The Development and Assessment of a Unique Disulfidptosis-Associated lncRNA Profile for Immune Microenvironment Prediction and Personalized Therapy in Gastric Adenocarcinoma. Biomedicines 2025; 13:1224. [PMID: 40427051 PMCID: PMC12109475 DOI: 10.3390/biomedicines13051224] [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: 03/13/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) are crucial factors affecting the occurrence, progression, and prognosis of gastric carcinoma (GC). The accumulation of disulfide bonds to excessive levels in cells expressing high SLC7A11 triggers disulfidptosis, which functions as a regulated form of cellular death. Research has demonstrated that upregulated SLC7A11 is common in human cancers, but the effect of disulfidptosis on GC remains unclear. Identifying lncRNAs associated with disulfidptosis (drlncRNAs) and establishing a prognostic risk profile holds considerable importance for advancing GC research and treatment. Methods: Clinical records and transcriptomic datasets from individuals with GC were acquired from The Cancer Genome Atlas (TCGA) repository. A three-drlncRNA risk model was built using three common regression analysis methods. Then we used receiver operating characteristic (ROC) curves, independent prognostic analysis, and additional statistical approaches to assess the precision of the model. This investigation additionally encompassed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, immune cell infiltration evaluation, and pharmacological sensitivity predictions. To further investigate immunotherapy response disparities between patient cohorts with elevated- and reduced-risk scores, analyses of tumor mutational burden (TMB), tumor immune dysfunction and exclusion (TIDE), and microsatellite instability (MSI) were implemented. Results: We constructed a unique model composed of three drlncRNAs (AC107021.2, AC016394.2, and AC129507.1). Its independent prognostic capability for GC patients was validated through both single-variable and multivariable Cox regression analyses. GO and KEGG pathway assessments revealed predominant enrichment within the elevated-risk cohort, particularly in pathways involving sulfur compound interactions, traditional Wnt signaling mechanisms, cell-substrate adherens junctions, and cAMP signaling cascades, among others. Tumor microenvironment (TME) evaluation demonstrated elevated ImmuneScores, StromalScores, and ESTIMATEScores within the high-risk patient population. Concurrently, this elevated-risk cohort exhibited enhanced immune cell infiltration patterns, whereas the reduced-risk group displayed superior expression of immune checkpoints (ICPs). Additional investigations revealed that patients categorized into the reduced-risk classification possessed greater tumor mutational burden, increased MSI-high proportions, and diminished tumor immune dysfunction and exclusion scores compared to their high-risk counterparts. Pharmacological sensitivity assessments confirmed the superior efficacy of several therapeutic agents, including gemcitabine and veliparib (ABT.888), in patients with lower risk classifications. Conclusions: Our established risk stratification system demonstrates independent prognostic predictive capacity while offering personalized clinical intervention guidance for individuals diagnosed with GC.
Collapse
Affiliation(s)
- Jiyue Zhu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Xiang Zhu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Tingting Su
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
| | - Huiqing Zhou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China;
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Weibin Shi
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| |
Collapse
|
2
|
Deng C, Zhang J, Yang Y, Ding Y, An F, Wang F. Chemodynamic Therapy Enhanced 131I-Radiotherapy for Efficient Inhibition on Cancer Growth and Metastasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503117. [PMID: 40376987 DOI: 10.1002/smll.202503117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/22/2025] [Indexed: 05/18/2025]
Abstract
Iodine-131 (131I), a cornerstone of thyroid cancer therapy, suffers from limited efficacy in other cancers due to poor tumor accumulation and hypoxia-driven radiotherapy resistance. To overcome these challenges, 131I-M@HI, a theranostic nanoparticle was engineered that synergizes radiotherapy with chemodynamic therapy (CDT). This platform integrated Mn(III) porphyrin and indocyanine green self-assembled on albumin, enabling dual-mode fluorescence/MRI-guided imaging, tumor/sentinel lymph node-targeted accumulation, and hypoxia modulation. The Mn(III) porphyrin catalyzes intratumoral hydrogen peroxide into cytotoxic hydroxyl radicals for CDT while alleviating hypoxia to amplify 131I radiotherapy. In subcutaneous tumors, 131I-M@HI achieved >85% tumor inhibition by inducing immunogenic cell death, marked by calreticulin exposure and high mobility group box 1 release, and triggered systemic anti-tumor immunity. Strikingly, in a breast cancer metastasis model, 131I-M@HI selectively eradicated sentinel lymph node metastases, reducing lung metastatic nodules by >90%, representing a critical advancement for preventing metastatic spread. This work pioneers a multifunctional nanoplatform that not only enhances radiotherapy but also redefines precision metastasis inhibition, offering a transformative strategy for advanced cancer therapy.
Collapse
Affiliation(s)
- Caiting Deng
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Jingjing Zhang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yuchen Yang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yuhan Ding
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Feifei An
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Fu Wang
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
3
|
Zhang Q, Liang Q, Xu C. Exploring the association between chemotherapy and prognosis among patients less than 50 years old with hepatocellular carcinoma: a retrospective cohort study based on the SEER database. Discov Oncol 2025; 16:682. [PMID: 40332634 PMCID: PMC12058605 DOI: 10.1007/s12672-025-02490-7] [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: 02/10/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND/AIM Hepatic carcinoma, including hepatocellular carcinoma (HCC), is one of the most common malignant tumors globally, with an increasing incidence among younger populations. While chemotherapy is effective for advanced HCC, its impact on the prognosis of younger patients, who typically have better physiological conditions, remains unclear. Younger patients may have different tumor biology and chemotherapy responses than older patients. This study aims to evaluate the impact of chemotherapy on the prognosis and survival rates of younger HCC patients. METHODS A retrospective analysis was conducted using the Surveillance, Epidemiology, and End Results Program (SEER) database, which provides information on cancer statistics among the US population. We selected patients diagnosed with primary HCC between 2010 and 2015. The patients were divided into two groups based on whether they received chemotherapy or not. Kaplan-Meier analyses were utilised to evaluate the impact of chemotherapy on prognosis by comparing the overall survival (OS) and cancer-specific survival (CSS) between the two groups. After performing 1:1 propensity score matching (PSM), the differences in OS and CSS were reassessed. RESULTS Before PSM, there were 1662 participants with primary HCC. After PSM, the sample was reduced to 1154 participants, with 577 individuals in each chemotherapy and non-chemotherapy group. Before PSM, there was no statistically significant difference in OS and CSS between the chemotherapy and non-chemotherapy groups (P = 0.25 and P = 0.06). After PSM, although the survival time in the chemotherapy group was slightly extended, the difference remained statistically insignificant (P = 0.09 and P = 0.38). Kaplan-Meier curves indicated no significant difference between the chemotherapy and non-chemotherapy groups, both before and after PSM, further supporting the conclusion that chemotherapy did not significantly improve survival in young patients with HCC. CONCLUSION Chemotherapy did not significantly improve survival for young patients with HCC. Treatment decisions should be approached cautiously, especially in cases with complex tumor characteristics. Future studies should explore the mechanisms of chemotherapy in younger patients and develop personalized treatment strategies to improve long-term outcomes.
Collapse
Affiliation(s)
- Qiyu Zhang
- Department of Interventional Treatment, Beijing NO.6 Hospital, No. 36, North Ertiao, Jiaodaokou, Dongcheng District, Beijing, 100009, China
| | - Qiongyu Liang
- Department of Interventional Treatment, Beijing NO.6 Hospital, No. 36, North Ertiao, Jiaodaokou, Dongcheng District, Beijing, 100009, China
| | - Chi Xu
- Department of Interventional Treatment, Beijing NO.6 Hospital, No. 36, North Ertiao, Jiaodaokou, Dongcheng District, Beijing, 100009, China.
| |
Collapse
|
4
|
Ning L, Li X, Xu Y, Si Y, Zhao H, Ren Q. Immunogenic cell death genes in single-cell and transcriptome analyses perspectives from a prognostic model of cervical cancer. Front Genet 2025; 16:1532523. [PMID: 40259929 PMCID: PMC12009919 DOI: 10.3389/fgene.2025.1532523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
Background The role of immunogenic cell death (ICD) in cervical cancer (CESC) is not well understood. This study sought to investigate the significance of ICD in CESC and to establish an ICDRs prognostic model to improve immunotherapy efficacy for patients with cervical cancer. Methods ICD-associated genes were screened at the single-cell and transcriptome levels based on AddModuleScore, single-sample gene set enrichment analysis (ssGSEA) and weighted gene co-expression network (WGCNA) analysis. Immunogenic cell death-related features (ICDRs) were constructed using multiple machine algorithms, and ICDRs were evaluated in training and validation sets to provide quantitative tools for predicting prognosis in clinical practice. Predictive models were used to risk subgroups for response to immunotherapy, as well as drug sensitivity. Finally, the expression of ICD-related genes was verified by RT-qPCR. Results Through an integrated analysis of single-cell data, transcriptomic profiling, and computational modeling, seven ICD-related genes were identified as highly prognostic for CESC patients. Multivariate analysis demonstrated that low-risk patients had significantly better overall survival compared to high-risk patients, confirming the model as an independent prognostic tool. Assessments of the tumor microenvironment (TME), mutation characteristics, and drug sensitivity within ICDRs risk subgroups indicated a stronger immunotherapy response in the low-risk group.
Collapse
Affiliation(s)
- Li Ning
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, China
| | - Xiu Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, China
| | - Yating Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, China
| | - Yu Si
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, China
| | - Hongting Zhao
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, China
| | - Qinling Ren
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Fang K, Yuan S, Zhang X, Zhang J, Sun SL, Li X. Regulation of immunogenic cell death and potential applications in cancer therapy. Front Immunol 2025; 16:1571212. [PMID: 40207233 PMCID: PMC11979251 DOI: 10.3389/fimmu.2025.1571212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Immunogenic cell death (ICD), a type of regulatory cell death, plays an important role in activating the adaptive immune response. Activation of the tumor-specific immune response is accompanied by the cell surface exposure of calreticulin and heat-shock proteins, the secretion of adenosine triphosphate, and the release of high mobility group box-1. In this review, we summarize and classify the latest types of ICD inducers and their molecular mechanisms, and discuss the effects and potential applications of inducing ICD by chemotherapy drugs, targeted drugs, and oncolytic viruses in clinical research. We also explore the potential role of epigenetic modifiers in the induction of ICD, and clarify the synergistic anti-tumor effects of nano-pulse stimulation, radiosensitizers for radiotherapy, photosensitizers for photodynamic therapy, photothermal therapy, and other physical stimulation, combined with radiotherapy and chemotherapy induced-ICD, in multimodal immunotherapy. In addition, we elucidate the molecular mechanism of ICD in detail, including the calcium imbalance, mitochondrial stress, and the interactions in the tumor microenvironment. Ultimately, this review aims to offer deeper insight into the factors and mechanisms of ICD induction and provide a theoretical basis for the future development of ICD-based immunotherapy.
Collapse
Affiliation(s)
- Kun Fang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xue Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Jingdong Zhang
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
- Department of Medical Oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
| | - Shu-lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Xiaoxi Li
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Zhuang W, Pan K, Wu J, Liu L, Lv S, Hu J, Shi F, Zhao W, Yu D. Harnessing the power of traceable system C-GAP: homologous-targeting to fire up T-cell immune responses with low-dose irradiation. J Nanobiotechnology 2025; 23:207. [PMID: 40075499 PMCID: PMC11905511 DOI: 10.1186/s12951-025-03281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
While radiotherapy-induced immunogenic cell death (ICD) holds potential for enhancing cancer immunotherapy, the conventional high-dose irradiation often leads to an immunosuppressive microenvironment and systemic toxicity. Therefore, a biomimetic nanoplatform cell membrane coated-nitrogen-doped graphene quantum dots combined with Au nanoparticles (C-GAP) was developed in this study. Firstly, homologous and traceable targeting features of C-GAP enables tumor-selective accumulation, providing reference for the selection of the timing of radiotherapy. Secondly, radiosensitization by C-GAP with Low-dose irradiation (LDI) amplifies reactive oxygen species (ROS) generation to trigger potent ICD. Thirdly, remarkable immune remodeling induced by C-GAP enhances CD8+ T cell infiltration and effector function. Single-cell RNA sequencing revealed that C-GAP-LDI combination upregulates TNF and CCL signaling pathway expression in tumor-infiltrating CD8+ T cells which potentiates tumor eradication. Our findings present a novel approach for safe and effective radioimmunotherapy, where C-GAP sensitized LDI achieves therapeutic enhancement through precise ICD induction and systemic immune activation.
Collapse
Affiliation(s)
- Weijie Zhuang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Kuangwu Pan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
- Department of Stomatology, The Third People's Hospital of Chengdu, Sichuan, China
| | - Jie Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Leyi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Shiyu Lv
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Jiajun Hu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Fangyang Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Wei Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China.
| | - Dongsheng Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China.
| |
Collapse
|
7
|
Hong S, Park J, Oh Y, Cho H, Kim K. Nanotechnology-Based Strategies for Safe and Effective Immunotherapy. Molecules 2024; 29:5855. [PMID: 39769944 PMCID: PMC11676242 DOI: 10.3390/molecules29245855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer immunotherapy using immune checkpoint blockades has emerged as a promising therapeutic approach. However, immunotherapy faces challenges such as low response rates in solid tumors, necessitating strategies to remodel the immune-suppressive tumor microenvironment (TME) into an immune-activated state. One of the primary approaches to achieve this transformation is through the induction of immunogenic cell death (ICD). Herein, we discussed strategies to maximize ICD induction using nanoparticles. In particular, this review highlighted various studies integrating chemotherapy, radiation therapy (RT), photodynamic therapy (PDT), and photothermal therapy (PTT) with nanoparticle-based immunotherapy. The research covered in this review aims to provide valuable insights for future studies on nanoparticle-assisted immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (S.H.); (J.P.); (Y.O.); (H.C.)
| |
Collapse
|
8
|
Xu S, Wu X, Zhu J, Wu Q, Gao L, Yang F, Zhang Z. Research Progress of Endoplasmic Reticulum Targeting Metal Complexes in Cancer Therapy. Drug Dev Res 2024; 85:e70027. [PMID: 39676587 DOI: 10.1002/ddr.70027] [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: 08/03/2024] [Revised: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
The development of anticancer drugs that target different organelles has received extensive attention due to the characteristics of cancer recurrence, metastasis, and drug resistance. The endoplasmic reticulum (ER) is an important structure within the cell that is primarily responsible for protein synthesis, folding, modification, and transport and plays a crucial role in cell function and health. ER stress activation induces cancer cell apoptosis. New anticancer drugs with different anticancer mechanisms and selectivity can be designed because of redox activity, composition diversity, and metal complexes structure regulation. Over the past few decades, dozens of metal complexes have killed cancer cells through ER stress, showing powerful tumor-suppressive effects. This review summarizes the progress of research on anticancer metallic drugs that induce ER stress over the past few years, which is expected to bring more breakthroughs in the field of medicine and life science.
Collapse
Affiliation(s)
- Shihang Xu
- School Hospital, Guangxi Normal University, Guilin, Guangxi, P.R. China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Xiaoling Wu
- School Hospital, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Jia Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Qiuming Wu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Lijuan Gao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Zhenlei Zhang
- School Hospital, Guangxi Normal University, Guilin, Guangxi, P.R. China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| |
Collapse
|