1
|
Feng X, Qin Y, Feng Y, Zhuo Y. Research on the functions and potential mechanisms of STAT3 in chronic myelogenous leukemia. Discov Oncol 2025; 16:739. [PMID: 40353923 PMCID: PMC12069186 DOI: 10.1007/s12672-025-02492-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVE To explore the bioinformatics characteristics and potential mechanisms of signal transducer and activator of transcription (STAT3) in chronic myelogenous leukemia (CML). METHODS Through the cancerSEA and CCLE databases, the expression of STAT3 in CML was verified and analyzed. Subsequently, K562 cells were treated with the STAT3 inhibitor Stattic. Western blotting, cell counting, and flow cytometry were utilized to observe its impact on the functions of K562 cells. Then, Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) were applied to deeply explore the regulatory mechanism of STAT3. The "LIMMA" software package was used to calculate STAT3-related differentially expressed genes (DEGs). Machine-earning methods were utilized to screen the STAT3-related hub genes. The "pROC" software package was employed to perform Receiver Operating Characteristic (ROC) curve analysis on the hub genes. The "corrplot" software package was used to conduct a correlation analysis of the hub genes. The "RMS" software package was applied to construct a nomogram of the hub genes. Based on the DisGENET database, a disease network of the hub genes was constructed, and the DGIdb database was used to construct a drug network of the hub genes. RESULTS In CML, the expression of STAT3 is upregulated compared to housekeeping genes. Among the 14 cell lines related to CML, STAT3 has the highest expression level in K562 cells. Stattic at a concentration of 5 μM can inhibit the proliferation of K562 cells, promote their apoptosis, and block the cell cycle at the S phase (P < 0.05). GSEA and GSVA indicates that amino acid metabolism, NOD-like receptor of STAT3. LASSO and SVM-RFE show that NCF4, PLAS1, IL7R, and TAGLN2 are hub differentially expressed genes (DEGs) related to STAT3. ROC and Nomogram indicate that the hub DEGs have high clinical diagnostic value. Correlation analysis shows that PLAS1 and NCF4 are negatively correlated, while PLAS1 and TAGLN2 are positively correlated. The construction of gene-disease networks reveals that these genes not only participate in the occurrence and development of CML but also jointly participate in multiple disease processes. The gene-drug network obtained 38 drugs targeting genes. CONCLUSION STAT3 might serve as a potential target for the treatment of CML. In CML, NCF4, PLAS1, IL7R, and TAGLN2 are hub genes associated with STAT3. These findings offer a fundamental theory for comprehending the pathogenesis of CML.
Collapse
Affiliation(s)
- Xiaoyun Feng
- Shizhen College of Guizhou University of Traditional Chinese Medicine, Guiyang, 550200, Guizhou, China.
| | - Yufeng Qin
- Department of Prosthodontics, Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Yulong Feng
- Shizhen College of Guizhou University of Traditional Chinese Medicine, Guiyang, 550200, Guizhou, China
| | - Yingquan Zhuo
- Department of Pediatric Surgery, The Afliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
2
|
Chen JH, Zhao CL, Zhang J, Cheng JW, Hu JP, Yu P, Yang MH, Xia YZ, Yin Y, Zhang ZZ, Luo JG, Kong LY, Zhang C. Enhancing immunogenicity and release of in situ-generated tumor vesicles for autologous vaccines. J Control Release 2025; 381:113614. [PMID: 40068738 DOI: 10.1016/j.jconrel.2025.113614] [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: 10/23/2024] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/21/2025]
Abstract
In situ vaccination (ISV) strategies offer an innovative approach to cancer immunotherapy by utilizing drug combinations directly at tumor sites to elicit personalized immune responses. Tumor cell-derived extracellular vesicles (TEVs) in ISV have great potential but face challenges such as low release rates and immunosuppressive proteins like programmed death ligand 1 (PD-L1) and CD47. This study develops a nanoparticle-based ISV strategy (Combo-NPs@shGNE) that enhances TEV release and modulates cargo composition. This approach combines Andrographolide, Icariside II, and shRNA targeting UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE), which accumulates in the tumor region, resulting in the regulation of immunosuppressive pathways and the reduction of sialic acid production. Decreasing the level of sialylation on the membrane through necroptosis and inhibition of sialic acid synthesis decreased the loading of PD-L1 and CD47 on vesicles, while increasing the loading of heat shock protein 70 and high mobility group box 1 on vesicles, and induced the release of highly immunogenic TEVs from the cancer cells, with a 56.44 % release, 9.57 times higher than that of blank nanoparticle-treated cells. In vivo studies demonstrate that Combo-NPs@shGNE enhances TEV yield, tumor growth, reduces metastases, and improves survival in an osteosarcoma mouse model. It promotes dendritic cell maturation, increases CD4+ and CD8+ T cell infiltration, and alters the microenvironment by reducing myeloid-derived suppressor cells and enhancing immunostimulatory factors. Additionally, it transitions tumor-associated macrophages from M2 to an M1 phenotype, thereby augmenting tumor immunity. Overall, Combo-NPs@shGNE offers a promising method for transforming tumors into personalized autologous vaccines, potentially advancing cancer treatment strategies.
Collapse
Affiliation(s)
- Jin-Hu Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Cai-Li Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jing Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jia-Wen Cheng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jian-Ping Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Pei Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ming-Hua Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yuan-Zheng Xia
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yong Yin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhen-Zhen Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jian-Guang Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Ling-Yi Kong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Chao Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
3
|
Jin W, Zhang Y, Wang B, Kang Z, Li H, Song J, Chen Y, Xiong H, Chen J. Structural optimization and characterization of highly potent and selective STAT3 inhibitors for the treatment of triple negative breast cancer. Eur J Med Chem 2025; 287:117332. [PMID: 39938409 DOI: 10.1016/j.ejmech.2025.117332] [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: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/14/2025]
Abstract
Effective targeted treatments for triple-negative breast cancer (TNBC), which has the worst prognosis among various types of breast cancer, are lacking owing to its clinical heterogeneity and malignant nature. STAT3, a key transcription factor, regulates multiple physiological functions. Aberrant activation of STAT3 plays a pivotal role in the initiation and progression of TNBC and is closely associated with a poor prognosis. Therefore, targeting STAT3 is a promising potential therapeutic approach for TNBC. In this study, we further optimized the core structure of 6f, which our research group previously identified as a STAT3 inhibitor and treatment for osteosarcoma, to identify additional potential STAT3 inhibitors for TNBC treatment. We identified WR-S-462 as a high-binding affinity inhibitor of STAT3 that effectively suppresses its phosphorylation and biological functions in vitro. Notably, WR-S-462 significantly inhibits TNBC growth and metastasis in a dose-dependent manner, providing robust evidence for its potential as a clinical intervention for TNBC.
Collapse
Affiliation(s)
- Wangrui Jin
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China; Institute for Advanced Study, Shenzhen University, 518060, Shenzhen, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Yuzhu Zhang
- Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, China
| | - Baozhen Wang
- School of Clinical Medicine, Ningxia Medical University, Ningxia, 750004, China; Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Ningxia, 750004, China
| | - Zhaoyong Kang
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Huachao Li
- Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, China
| | - Jingfeng Song
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China
| | - Yihua Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University, 518060, Shenzhen, China.
| | - Jing Chen
- School of Basic Medical Sciences, Ningxia Medical University, Ningxia, 750004, China; Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Ningxia, 750004, China.
| |
Collapse
|
4
|
Wu F, An X, Li S, Qiu C, Zhu Y, Ye Z, Song S, Wang Y, Shen D, Di X, Yao Y, Zhu W, Jiang X, Shi X, Chen R, Kou L. Enhancing chemoimmunotherapy for colorectal cancer with paclitaxel and alantolactone via CD44-Targeted nanoparticles: A STAT3 signaling pathway modulation approach. Asian J Pharm Sci 2025; 20:100993. [PMID: 39917727 PMCID: PMC11795048 DOI: 10.1016/j.ajps.2024.100993] [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: 05/24/2024] [Revised: 09/28/2024] [Accepted: 10/22/2024] [Indexed: 02/09/2025] Open
Abstract
Chemoimmunotherapy has the potential to enhance chemotherapy and modulate the immunosuppressive tumor microenvironment by activating immunogenic cell death (ICD), making it a promising strategy for clinical application. Alantolactone (A) was found to augment the anticancer efficacy of paclitaxel (P) at a molar ratio of 1:0.5 (P:A) through induction of more potent ICD via modulation of STAT3 signaling pathways. Nano drug delivery systems can synergistically combine natural drugs with conventional chemotherapeutic agents, thereby enhancing multi-drug chemoimmunotherapy. To improve tumor targeting ability and bioavailability of hydrophobic drugs, an amphiphilic prodrug conjugate (HA-PTX) was chemically modified with paclitaxel (PTX) and hyaluronic acid (HA) as a backbone. Based on this concept, CD44-targeted nanodrugs (A@HAP NPs) were developed for co-delivery of A and P in colorectal cancer treatment, aiming to achieve synergistic toxicity-based chemo-immunotherapy. The uniform size and high drug loading capacity of A@HAP NPs facilitated their accumulation within tumors through enhanced permeability and retention effect as well as HA-mediated targeting, providing a solid foundation for subsequent synergistic therapy and immunoregulation. In vitro and in vivo studies demonstrated that A@HAP NPs exhibited potent cytotoxicity against tumor cells while also remodeling the immune-suppressive tumor microenvironment by promoting antigen presentation and inducing dendritic cell maturation, thus offering a novel approach for colorectal cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Fugen Wu
- Department of Pediatrics, Wenling Hospital of Wenzhou Medical University, Wenling 317500, China
| | - Xingsi An
- Department of Pediatrics, Wenling Hospital of Wenzhou Medical University, Wenling 317500, China
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shize Li
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenyu Qiu
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yixuan Zhu
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhanzheng Ye
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shengnan Song
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yunzhi Wang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Dingchao Shen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyu Di
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinsha Yao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Wanling Zhu
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Xinyu Jiang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
5
|
Amiri M, Jafari S, Lavasanifar A, Molavi O, Montazersaheb S. Nano-delivery of Silibinin Potentiate the Induction of Immunogenic Cell Death (ICD) in Melanoma Cells. Curr Pharm Biotechnol 2025; 26:392-401. [PMID: 38482616 DOI: 10.2174/0113892010280336240227062954] [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: 10/25/2023] [Revised: 01/17/2024] [Accepted: 02/01/2024] [Indexed: 03/25/2025]
Abstract
BACKGROUND Induction of immunogenic cell death (ICD) in tumors can enhance antitumor immunity and modulate immunosuppression in the tumor microenvironment (TME). OBJECTIVE In the current study, we investigated the effect of silibinin, a natural compound with anticancer activity, and its polymer-based nanoformulations on the induction of apoptosis and ICD in cancer cells. METHODS Free and nanoparticulate silibinin were evaluated for their growth-inhibitory effects using an MTT assay. Annexin V/PI staining was used to analyze apoptosis. Calreticulin (CRT) expression was measured by flow cytometry. Western blotting was conducted to examine the levels of elf2α, which plays a role in the ICD pathway. The HSP90 and ATP levels were determined using specific detection kits. RESULTS Compared to the free drug, silibinin-loaded nanocarriers significantly increased the induction of apoptosis and ICD in B16F10 cells. ICD induction was characterized by significantly increased levels of ICD biomarkers, including CRT, HSP90, and ATP. We also observed an increased expression of p-elf-2α/ elf-2α in B16F10 cells treated with silibinin-loaded micelles compared to cells that received free silibinin. CONCLUSION Our findings showed that the encapsulation of silibinin in polymeric nanocarriers can potentiate the effects of this drug on the induction of apoptosis and ICD in B16F10 melanoma cells.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Jafari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Jafari S, Ardakan AK, Aghdam EM, Mesbahi A, Montazersaheb S, Molavi O. Induction of immunogenic cell death and enhancement of the radiation-induced immunogenicity by chrysin in melanoma cancer cells. Sci Rep 2024; 14:23231. [PMID: 39369019 PMCID: PMC11455848 DOI: 10.1038/s41598-024-72697-1] [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: 11/08/2023] [Accepted: 09/10/2024] [Indexed: 10/07/2024] Open
Abstract
Chrysin is a natural flavonoid with anti-cancer effects. Despite its beneficial effects, little information is available regarding its immunogenic cell death (ICD) properties. In this work, we hypothesized that chrysin can potentiate radiotherapy(RT)-induced immunogenicity in melanoma cell line (B16-F10). We examined the effects of chrysin alone and in combination with radiation on ICD induction in B16-F10 cells. Cell viability was assessed using an MTT assay. Cell apoptosis and calreticulin (CRT) exposure were determined using flow cytometry. Western blotting and ELISA assay were employed to examine changes in protein expression. Combination therapy exhibited a synergistic effect, with an optimum combination index of 0.66. The synergistic anti-cancer effect correlated with increased cell apoptosis in cancer cells. Compared to the untreated control, chrysin alone and in combination with RT induced higher levels of DAMPs, such as CRT, HSP70, HMGB1, and ATP. The protein expression of p-STAT3/STAT3 and PD-L1 was reduced in B16-F10 cells exposed to chrysin alone and in combination with RT. Conditioned media from B16-F10 cells exposed to mono-and combination treatments elicited IL-12 secretion in dendritic cells (DCs), inducing a Th1 response. Our findings revealed that chrysin could induce ICD and intensify the RT-induced immunogenicity.
Collapse
Affiliation(s)
- Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Khodaei Ardakan
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Elnaz Mehdizadeh Aghdam
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran
| | - Asghar Mesbahi
- Medical Radiation Research Team, 84 Gorge Road, South Morang, Melbourne, Australia
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran.
| | - Ommoleila Molavi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran.
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran.
| |
Collapse
|
7
|
Li M, Xie Y, Zhang J, Zhou X, Gao L, He M, Liu X, Miao X, Liu Y, Cao R, Jia Y, Zeng Z, Liu L. Intratumoral injection of mRNA encoding survivin in combination with STAT3 inhibitor stattic enhances antitumor effects. Cancer Lett 2024; 598:217111. [PMID: 38972347 DOI: 10.1016/j.canlet.2024.217111] [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: 06/14/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Intratumoral delivery of mRNA encoding immunostimulatory molecules can initiate a robust, global antitumor response with little side effects by enhancing local antigen presentation in the tumor and the tumor draining lymph node. Neoantigen-based mRNA nanovaccine can inhibit melanoma growth in mice by intratumoral injection. Myeloid-derived suppressor cells (MDSCs) suppress antitumor immune responses by secreting immunosuppressive agents, such as reactive oxygen species (ROS). Suppression of STAT3 activity by stattic may reduce MDSC-mediated immunosuppression in the TME and promote the antitumor immune responses. In this study, in vitro transcribed mRNA encoding tumor antigen survivin was prepared and injected intratumorally in BALB/c mice bearing subcutaneous colon cancer tumors. In vivo studies demonstrated that intratumoral survivin mRNA therapy could induce antitumor T cell response and inhibit tumor growth of colon cancer. Depletion of CD8+ T cells could significantly inhibit survivin mRNA-induced antitumor effects. RT-qPCR and ELISA analysis indicated that survivin mRNA treatment led to increased expression of receptor activator nuclear factor-κB ligand (RANKL). In vitro experiment showed that MDSCs could be induced from mouse bone marrow cells by RANKL and RANKL-induced MDSCs could produce high level of ROS. STAT3 inhibitor stattic suppressed activation of STAT3 and NF-κB signals, thereby inhibiting expansion of RANKL-induced MDSCs. Combination therapy of survivin mRNA and stattic could significantly enhance antitumor T cell response, improve long-term survival and reduce immunosuppressive tumor microenvironment compared to each monotherapy. In addition, combined therapy resulted in a significantly reduced level of tumor cell proliferation and an obviously increased level of tumor cell apoptosis in CT26 colon cancer-bearing mice, which could be conducive to inhibit the tumor growth and lead to immune responses to released tumor-associated antigens. These studies explored intratumoral mRNA therapy and mRNA-based combined therapy to treat colon cancer and provide a new idea for cancer therapy.
Collapse
Affiliation(s)
- Min Li
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Xie
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Jincheng Zhang
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Xue Zhou
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Lei Gao
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Mengmeng He
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Xianmei Liu
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Xinyi Miao
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Yu Liu
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Rong Cao
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China
| | - Yi Jia
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Biological and Medical Engineering/Immune Cells and Antibody Engineering Research Center of Guizhou Province/Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Zhu Zeng
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Biological and Medical Engineering/Immune Cells and Antibody Engineering Research Center of Guizhou Province/Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Lina Liu
- Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, School of Basic Medical Science/School of Biology and Engineering (School of Health Medicine Modern Industry), Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China; Key Laboratory of Biological and Medical Engineering/Immune Cells and Antibody Engineering Research Center of Guizhou Province/Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
8
|
Zhong H, Wang L, Zhu X, Li S, Li X, Ding C, Wang K, Wang X. STAT3 inhibitor Stattic Exhibits the Synergistic Effect with FGFRs Inhibitor Erdafitinib in FGFR1-positive Lung Squamous Cell Carcinoma. J Cancer 2024; 15:5415-5424. [PMID: 39247610 PMCID: PMC11375536 DOI: 10.7150/jca.97477] [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: 04/18/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Lung squamous cell carcinoma (LUSC), a subset of non-small cell lung cancer (NSCLC), accounts for about 30% of all lung cancers (LC) and exhibits a dismal response to current therapeutic protocols. Existed studies have indicated that aberrations in fibroblast growth factor receptors (FGFRs) play a pivotal role in the progression of LUSC, rendering them as attractive targets for therapeutic intervention in this cancer type. This study found that Erdafitinib (Erda), a novel pan-FGF receptor tyrosine kinase inhibitor (TKI), exerted a cytotoxic effect on LUSC cells. However, STAT3, the downstream target of FGFRs, remained still activated despite Erdafitinib treatment. Then, a STAT3 inhibitor, Stattic (Sta), was concurrently used with Erdafitinib, and the combined treatment demonstrated a synergistic efficacy in both in vitro and in vivo models of LUSC when compared to that of the treatment of the Erdafitinib or Stattic alone. Further molecular studies showed that such an effect of Erdafitinib and Stattic was associated with their concurrently inhibitory effect on FGFR1 and STAT3 signaling in LUSC cells. Therefore, the findings of this study indicated that the concurrent use of Erdafitinib and Stattic is a promising therapeutic approach for the treatment of FGFR1-positive LUSC.
Collapse
Affiliation(s)
- Hongqin Zhong
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
- Nantong University Medical School, Nantong 226007, Jiangsu Province, China
| | - Ling Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Shu Li
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi 214126, Jiangsu Province, China
| | - Xiyue Li
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
| | - Chen Ding
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu Province, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Xun Wang
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University (Wuxi No.2 People's Hospital) Wuxi 214126, Jiangsu Province, China
- Nantong University Medical School, Nantong 226007, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi 214126, Jiangsu Province, China
| |
Collapse
|
9
|
Ning J, Lu X, Dong J, Xue C, Ou C, Zhang Y, Zhang X, Gao F. Advanced Strategies for Strengthening the Immune Activation Effect of Traditional Antitumor Therapies. ACS Biomater Sci Eng 2024; 10:4701-4715. [PMID: 38959418 DOI: 10.1021/acsbiomaterials.4c00560] [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: 07/05/2024]
Abstract
The utilization of traditional therapies (TTS), such as chemotherapy, reactive oxygen species-based therapy, and thermotherapy, to induce immunogenic cell death (ICD) in tumor cells has emerged as a promising strategy for the activation of the antitumor immune response. However, the limited effectiveness of most TTS in inducing the ICD effect of tumors hinders their applications in combination with immunotherapy. To address this challenge, various intelligent strategies have been proposed to strengthen the immune activation effect of these TTS, and then achieve synergistic antitumor efficacy with immunotherapy. These strategies primarily focus on augmenting the tumor ICD effect or facilitating the antigen (released by the ICD tumor cells) presentation process during TTS, and they are systematically summarized in this review. Finally, the existing bottlenecks and prospects of TTS in the application of tumor immune regulation are also discussed.
Collapse
Affiliation(s)
- Jingyi Ning
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Xinxin Lu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Jianhui Dong
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Chun Xue
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Changjin Ou
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Yizhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Xianzheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| |
Collapse
|
10
|
Yang LJ, Han T, Liu RN, Shi SM, Luan SY, Meng SN. Plant-derived natural compounds: A new frontier in inducing immunogenic cell death for cancer treatment. Biomed Pharmacother 2024; 177:117099. [PMID: 38981240 DOI: 10.1016/j.biopha.2024.117099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
Immunogenic cell death (ICD) can activate adaptive immune response in the host with normal immune system. Some synthetic chemotherapeutic drugs and natural compounds have shown promising results in cancer treatment by triggering the release of damage-associated molecules (DAMPs) to trigger ICD. However, most chemotherapeutic drugs exhibit non-selective cytotoxicity and may also induce and promote metastasis, thereby significantly reducing their clinical efficacy. Among the natural compounds that can induce ICD, plant-derived compounds account for the largest proportion, which are of increasing value in the treatment of cancer. Understanding which plant-derived natural compounds can induce ICD and how they induce ICD is crucial for developing strategies to improve chemotherapy outcomes. In this review, we focus on the recent findings regarding plant-derived natural compounds that induce ICD according to the classification of flavonoids, alkaloids, glycosides, terpenoids and discuss the potential mechanisms including endoplasmic reticulum (ER) stress, DNA damage, apoptosis, necroptosis autophagy, ferroptosis. In addition, plant-derived natural compounds that can enhance the ICD induction ability of conventional therapies for cancer treatment is also elaborated. The rational use of plant-derived natural compounds to induce ICD is helpful for the development of new cancer treatment methods.
Collapse
Affiliation(s)
- Li-Juan Yang
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Ting Han
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Ruo-Nan Liu
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Shu-Ming Shi
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Shi-Yun Luan
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Sheng-Nan Meng
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
11
|
Dabiri S, Jafari S, Molavi O. Advances in nanocarrier-mediated delivery of chrysin: Enhancing solubility, bioavailability, and anticancer efficacy. BIOIMPACTS : BI 2024; 15:30269. [PMID: 40161948 PMCID: PMC11954748 DOI: 10.34172/bi.30269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 04/02/2025]
Abstract
Chrysin, a natural phytochemical compound found in various plant sources, possesses diverse pharmacological benefits, including anticancer, antioxidant, antidiabetic, neuroprotective, cardioprotective, hepatoprotective, immunoregulatory, and anti-inflammatory properties. Despite its well-documented biological activities, chrysin's low water solubility and bioavailability hinder its clinical development. This review explores the application of nanocarriers as a strategic approach to overcome these challenges and enhance the delivery of chrysin. Nanocarriers, including polymer-based nanoparticles (NPs), lipid-based NPs, and inorganic nanocarriers, have shown promise in improving the solubility, bioavailability, and tumor-targeted delivery of chrysin. The paper discusses chrysin's anticancer effects on different types of human cancers, elucidating its impact on crucial signaling pathways involved in tumorigenesis. The review categorizes and analyzes various nanocarriers, providing insights into their structural properties and drug release profiles. Among the nanocarriers, polymer-based NPs, especially those utilizing PLGA, emerge as promising strategies for chrysin encapsulation, demonstrating improvements in drug release, stability, and bioavailability. Lipid-based NPs and inorganic nanocarriers also exhibit potential in enhancing chrysin delivery. The comprehensive insights provided contribute to a deeper understanding of chrysin's pharmacological properties and its potential clinical applications, offering valuable perspectives for future research and translation into clinical settings. The review underscores the importance of selecting suitable structures for chrysin encapsulation to enhance its physicochemical properties and anticancer effects, paving the way for innovative nanomedicine approaches in cancer therapy.
Collapse
Affiliation(s)
- Sheida Dabiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
12
|
Aebisher D, Przygórzewska A, Bartusik-Aebisher D. The Latest Look at PDT and Immune Checkpoints. Curr Issues Mol Biol 2024; 46:7239-7257. [PMID: 39057071 PMCID: PMC11275601 DOI: 10.3390/cimb46070430] [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: 05/23/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Photodynamic therapy (PDT) can not only directly eliminate cancer cells, but can also stimulate antitumor immune responses. It also affects the expression of immune checkpoints. The purpose of this review is to collect, analyze, and summarize recent news about PDT and immune checkpoints, along with their inhibitors, and to identify future research directions that may enhance the effectiveness of this approach. A search for research articles published between January 2023 and March 2024 was conducted in PubMed/MEDLINE. Eligibility criteria were as follows: (1) papers describing PDT and immune checkpoints, (2) only original research papers, (3) only papers describing new reports in the field of PDT and immune checkpoints, and (4) both in vitro and in vivo papers. Exclusion criteria included (1) papers written in a language other than Polish or English, (2) review papers, and (3) papers published before January 2023. 24 papers describing new data on PDT and immune checkpoints have been published since January 2023. These included information on the effects of PDT on immune checkpoints, and attempts to associate PDT with ICI and with other molecules to modulate immune checkpoints, improve the immunosuppressive environment of the tumor, and resolve PDT-related problems. They also focused on the development of new nanoparticles that can improve the delivery of photosensitizers and drugs selectively to the tumor. The effect of PDT on the level of immune checkpoints and the associated activity of the immune system has not been fully elucidated further, and reports in this area are divergent, indicating the complexity of the interaction between PDT and the immune system. PDT-based strategies have been shown to have a beneficial effect on the delivery of ICI to the tumor. The utility of PDT in enhancing the induction of the antitumor response by participating in the triggering of immunogenic cell death, the exposure of tumor antigens, and the release of various alarm signals that together promote the activation of dendritic cells and other components of the immune system has also been demonstrated, with the result that PDT can enhance the antitumor immune response induced by ICI therapy. PDT also enables multifaceted regulation of the tumor's immunosuppressive environment, as a result of which ICI therapy has the potential to achieve better antitumor efficacy. The current review has presented evidence of PDT's ability to modulate the level of immune checkpoints and the effectiveness of the association of PDT with ICIs and other molecules in inducing an effective immune response against cancer cells. However, these studies are at an early stage and many more observations need to be made to confirm their efficacy. The new research directions indicated may contribute to the development of further strategies.
Collapse
Affiliation(s)
- David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College, The Rzeszów University, 35-959 Rzeszów, Poland
| | - Agnieszka Przygórzewska
- English Division Science Club, Medical College of The Rzeszów University, 35-025 Rzeszów, Poland;
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of The Rzeszów University, 35-025 Rzeszów, Poland;
| |
Collapse
|
13
|
Franzese O, Ancona P, Bianchi N, Aguiari G. Apoptosis, a Metabolic "Head-to-Head" between Tumor and T Cells: Implications for Immunotherapy. Cells 2024; 13:924. [PMID: 38891056 PMCID: PMC11171541 DOI: 10.3390/cells13110924] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Induction of apoptosis represents a promising therapeutic approach to drive tumor cells to death. However, this poses challenges due to the intricate nature of cancer biology and the mechanisms employed by cancer cells to survive and escape immune surveillance. Furthermore, molecules released from apoptotic cells and phagocytes in the tumor microenvironment (TME) can facilitate cancer progression and immune evasion. Apoptosis is also a pivotal mechanism in modulating the strength and duration of anti-tumor T-cell responses. Combined strategies including molecular targeting of apoptosis, promoting immunogenic cell death, modulating immunosuppressive cells, and affecting energy pathways can potentially overcome resistance and enhance therapeutic outcomes. Thus, an effective approach for targeting apoptosis within the TME should delicately balance the selective induction of apoptosis in tumor cells, while safeguarding survival, metabolic changes, and functionality of T cells targeting crucial molecular pathways involved in T-cell apoptosis regulation. Enhancing the persistence and effectiveness of T cells may bolster a more resilient and enduring anti-tumor immune response, ultimately advancing therapeutic outcomes in cancer treatment. This review delves into the pivotal topics of this multifaceted issue and suggests drugs and druggable targets for possible combined therapies.
Collapse
Affiliation(s)
- Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy;
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy;
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via F. Mortara 74, 44121 Ferrara, Italy;
| |
Collapse
|
14
|
Pardeshi S, Mohite P, Rajput T, Puri A. The Nanotech Potential of Curcumin in Pharmaceuticals: An Overview. Curr Drug Discov Technol 2024; 21:e260723219113. [PMID: 37493163 DOI: 10.2174/1570163820666230726125809] [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: 01/28/2023] [Revised: 03/02/2023] [Accepted: 03/10/2023] [Indexed: 07/27/2023]
Abstract
It is safe to use Curcumin as a cosmetic and therapeutic ingredient in pharmaceutical products. For the uses mentioned above and for fundamental research, it is essential to obtain pure Curcumin from plant sources. There is a requirement for effective extraction and purification techniques that adhere to green chemistry standards for efficiency improvement, process safety, and environmental friendliness. Several outstanding studies have looked into the extraction and purification of Curcumin. This review thoroughly covers the currently available curcumin extraction, synthesis, and transformation techniques. Additionally, Curcumin's poor solubility and low absorption in the human body have limited its potential for pharmaceutical use. However, recent developments in novel curcumin formulations utilizing nanotechnology delivery methods have provided new approaches to transport and maximize the human body's curcumin absorption efficiency. In this review, we explore the various curcumin nanoformulations and the potential medicinal uses of nano curcumin. Additionally, we review the necessary future research directions to recommend Curcumin as an excellent therapeutic candidate.
Collapse
Affiliation(s)
- Sagar Pardeshi
- Department of Pharmaceutics AET's St. John Institute of Pharmacy and Research, Manor Road, Palghar, Maharashtra- 401404, India
| | - Popat Mohite
- Department of Pharmaceutical Chemistry, AET's St. John Institute of Pharmacy and Research, Manor Road, Palghar, Maharashtra-401404, India
| | - Tanavirsing Rajput
- Department of Pharmaceutical Chemistry, AET's St. John Institute of Pharmacy and Research, Manor Road, Palghar, Maharashtra-401404, India
| | - Abhijeet Puri
- Department of Pharmacognosy, AET's St. John Institute of Pharmacy and Research, Manor Road, Palghar, Maharashtra-401404, India
| |
Collapse
|
15
|
Qiu Z, Lu Z, Huang J, Zhong Y, Yan N, Kong R, Cheng H. Self-reinforced photodynamic immunostimulator to downregulate and block PD-L1 for metastatic breast cancer treatment. Biomaterials 2023; 303:122392. [PMID: 37984245 DOI: 10.1016/j.biomaterials.2023.122392] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/22/2023] [Accepted: 11/04/2023] [Indexed: 11/22/2023]
Abstract
Tumor cells overexpress programmed cell death ligand 1 (PD-L1) to impede immune responses and escape immune elimination. Development of effective combination regimens to sensitize immunotherapy is promising but always challenging. Herein, a self-reinforced photodynamic immunostimulator (designated as PCS) is constructed for metastatic breast cancer treatment through simultaneous downregulation and blockade of PD-L1. Specifically, PCS is prepared by encapsulating signal transducer and activator of transcription 3 (STAT3) inhibitor (Stattic) into photosensitizer (protoporphyrin IX) modified PD-L1 blockade peptide (CVRARTR) through drug self-assembly. PCS can facilitate the targeted drug accumulation in PD-L1 overexpressed breast cancer cells to block PD-L1 and inhibit the phosphorylation of STAT3 to downregulate PD-L1. Moreover, PCS increases intracellular oxidative stress to show a robust anti-proliferation effect through photodynamic therapy (PDT), which also triggers an immunogenic cell death (ICD) to expose the immunostimulatory signals. Consequently, the efficient PD-L1 inhibition and robust PDT of PCS synergistically suppress the malignant growth of breast cancer, and concurrently activate the systemic anti-tumor immunity for metastatic inhibition with no obvious side effects. Such a photodynamic immunostimulator may provide an effective combination regimen for therapies activated immunotherapy against metastatic breast cancer.
Collapse
Affiliation(s)
- Ziwen Qiu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Zhenming Lu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Jiaqi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Yingtao Zhong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Ni Yan
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Renjiang Kong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, PR China
| | - Hong Cheng
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
16
|
Amiri M, Molavi O, Sabetkam S, Jafari S, Montazersaheb S. Stimulators of immunogenic cell death for cancer therapy: focusing on natural compounds. Cancer Cell Int 2023; 23:200. [PMID: 37705051 PMCID: PMC10500939 DOI: 10.1186/s12935-023-03058-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
A growing body of evidence indicates that the anticancer effect of the immune system can be activated by the immunogenic modulation of dying cancer cells. Cancer cell death, as a result of the activation of an immunomodulatory response, is called immunogenic cell death (ICD). This regulated cell death occurs because of increased immunogenicity of cancer cells undergoing ICD. ICD plays a crucial role in stimulating immune system activity in cancer therapy. ICD can therefore be an innovative route to improve anticancer immune responses associated with releasing damage-associated molecular patterns (DAMPs). Several conventional and chemotherapeutics, as well as preclinically investigated compounds from natural sources, possess immunostimulatory properties by ICD induction. Natural compounds have gained much interest in cancer therapy owing to their low toxicity, low cost, and inhibiting cancer cells by interfering with different mechanisms, which are critical in cancer progression. Therefore, identifying natural compounds with ICD-inducing potency presents agents with promising potential in cancer immunotherapy. Naturally derived compounds are believed to act as immunoadjuvants because they elicit cancer stress responses and DAMPs. Acute exposure to DAMP molecules can activate antigen-presenting cells (APCs), such as dendritic cells (DCs), which leads to downstream events by cytotoxic T lymphocytes (CTLs) and natural killer cells (NKs). Natural compounds as inducers of ICD may be an interesting approach to ICD induction; however, parameters that determine whether a compound can be used as an ICD inducer should be elucidated. Here, we aimed to discuss the impact of multiple ICD inducers, mainly focusing on natural agents, including plant-derived, marine molecules, and bacterial-based compounds, on the release of DAMP molecules and the activation of the corresponding signaling cascades triggering immune responses. In addition, the potential of synthetic agents for triggering ICD is also discussed.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahnaz Sabetkam
- Department of Anatomy, Faculty of Medicine, university of Kyrenia, Kyrenia, Northern Cyprus
- Department of Anatomy and histopathology, Faculty of medicine, Tabriz medical sciences, Islamic Azad University, Tabriz, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Oliyapour Y, Dabiri S, Molavi O, Hejazi MS, Davaran S, Jafari S, Montazersaheb S. Chrysin and chrysin-loaded nanocarriers induced immunogenic cell death on B16 melanoma cells. Med Oncol 2023; 40:278. [PMID: 37624439 DOI: 10.1007/s12032-023-02145-z] [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: 06/17/2023] [Accepted: 07/29/2023] [Indexed: 08/26/2023]
Abstract
Induction of immunogenic cell death (ICD) is a promising strategy for cancer immunotherapy. Chrysin, which has potential anticancer effects, faces limitations in clinical applications due to its poor water solubility. This study aimed to formulate chrysin with PEG-poly(α-benzylcarboxylate-ε-caprolactone) (PBCL) nanoparticles (NPs) and assess their anticancer and ICD-inducing potency in melanoma cells, comparing with free chrysin. The co-solvent evaporation method was employed to develop chrysin-loaded NPs. UV spectroscopy, dynamic light scattering, and the dialysis bag method were used to evaluate the encapsulation efficiency (EE), particle size, polydispersity index (PDI), and drug release profile, respectively. The anticancer effects of the drugs were assessed using the MTT and trypan blue exclusion assays. Flow cytometry was employed to evaluate apoptosis and calreticulin (CRT) expression. ELISA and western blotting were used to detect heat shock protein 90 (HSP90), Annexin A1, GRP78 (Glucose-related protein78), and activated protein kinase R-like endoplasmic reticulum kinase (p-PERK). Chrysin-loaded PEG-PBCL NPs (chrysin-PEG-PBCL) showed an EE of 97 ± 1%. Chrysin-PEG-PBCL was 38.18 ± 3.96 nm in size, with a PDI being 0.62 ± 0.23. Chrysin-PEG-PBCL showed an initial burst release, followed by sustained release over 24 h. Chrysin-PEG-PBCL exhibited a significantly stronger anticancer effect in B16 cells. Chrysin-PEG-PBCL was found to be more potent in inducing apoptosis. Both free chrysin and chrysin NPs induced ICD as indicated by an increase in the levels of ICD biomarkers. Interestingly, chrysin NPs were found to be more potent inducers of ICD than the free drug. These findings demonstrate that chrysin and chrysin-PEG-PBCL NPs can induce ICD in B16 cells. PEG-PBCL NPs significantly enhanced the potency of chrysin in inducing ICD compared to its free form.
Collapse
Affiliation(s)
- Yasaman Oliyapour
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sheida Dabiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Mohammad Saeid Hejazi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614731, Iran
| | - Soodabeh Davaran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614711, Iran.
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614731, Iran.
| |
Collapse
|
18
|
Pelka S, Guha C. Enhancing Immunogenicity in Metastatic Melanoma: Adjuvant Therapies to Promote the Anti-Tumor Immune Response. Biomedicines 2023; 11:2245. [PMID: 37626741 PMCID: PMC10452223 DOI: 10.3390/biomedicines11082245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Advanced melanoma is an aggressive form of skin cancer characterized by low survival rates. Less than 50% of advanced melanoma patients respond to current therapies, and of those patients that do respond, many present with tumor recurrence due to resistance. The immunosuppressive tumor-immune microenvironment (TIME) remains a major obstacle in melanoma therapy. Adjuvant treatment modalities that enhance anti-tumor immune cell function are associated with improved patient response. One potential mechanism to stimulate the anti-tumor immune response is by inducing immunogenic cell death (ICD) in tumors. ICD leads to the release of damage-associated molecular patterns within the TIME, subsequently promoting antigen presentation and anti-tumor immunity. This review summarizes relevant concepts and mechanisms underlying ICD and introduces the potential of non-ablative low-intensity focused ultrasound (LOFU) as an immune-priming therapy that can be combined with ICD-inducing focal ablative therapies to promote an anti-melanoma immune response.
Collapse
Affiliation(s)
- Sandra Pelka
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Urology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute of Onco-Physics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
19
|
Lee Y, Shinn J, Xu C, Dobson HE, Neamati N, Moon JJ. Hyaluronic acid-bilirubin nanomedicine-based combination chemoimmunotherapy. Nat Commun 2023; 14:4771. [PMID: 37553327 PMCID: PMC10409794 DOI: 10.1038/s41467-023-40270-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/20/2023] [Indexed: 08/10/2023] Open
Abstract
Despite significant advances in immune checkpoint blockade (ICB), immunosuppression mediated by tumor-associated myeloid cells (TAMCs) poses a major barrier to cancer immunotherapy. In addition, while immunogenic cell death (ICD) provides a viable approach to inducing anti-tumor immune response, it remains unknown how to effectively trigger ICD while addressing immunosuppressive TAMCs. Here, we show that SC144, a gp130 inhibitor that blocks the IL-6/gp130/STAT3 pathway, induces ICD of tumor cells and polarizes macrophages to M1-phenotype in vitro. However, as SC144 also induces killing of CD8+ T-cells, we sought to deliver SC144 selectively to tumor cells and TAMCs. Toward this goal, we have developed hyaluronic acid-bilirubin nanoparticles (HABN) that accumulate in CD44hi tumor cells and TAMCs. Systemic administration of SC144 loaded in HABN (SC144@HABN) induces apoptosis and ICD of tumor cells, increases the ratio of M1-like to M2-like macrophages, and decreases the frequency of myeloid-derived suppressor cells and CD4+ regulatory T-cells, while promoting anti-tumor CD8+ T-cells. Moreover, SC144@HABN combined with anti-PD-L1 ICB efficiently eliminates MC38 tumors and ICB-resistant 4T1 tumors. Overall, our work demonstrates a therapeutic strategy based on coordinated ICD induction and TAMC modulation and highlights the potential of combination chemoimmunotherapy.
Collapse
Affiliation(s)
- Yonghyun Lee
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul, 03760, South Korea.
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea.
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Jongyoon Shinn
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul, 03760, South Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Cheng Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hannah E Dobson
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
20
|
Li L, Mou Y, Zhai Q, Yan C, Zhang X, Du M, Li Y, Wang Q, Xiao Z. PirB negatively regulates the inflammatory activation of astrocytes in a mouse model of sleep deprivation. Neuropharmacology 2023; 235:109571. [PMID: 37146940 DOI: 10.1016/j.neuropharm.2023.109571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/16/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Reactive astrocytes play a potential regulatory role in sleep deprivation (SD). Paired immunoglobulin-like receptor B (PirB) is expressed in reactive astrocytes, suggesting that PirB may participate in regulating the inflammatory response of astrocytes. We used lentiviral and adeno-associated viral approaches to interfere with the expression of PirB in vivo and in vitro. C57BL/6 mice were sleep deprived for 7 days and neurological function was measured via behavioral tests. We found that overexpressed PirB in SD mice could decrease the number of neurotoxic reactive astrocytes, alleviate cognitive deficits, and promote reactive astrocytes tended to be neuroprotective state. IL-1α, TNFα, and C1q were used to induce neurotoxic reactive astrocytes in vitro. Overexpression of PirB relieved the toxicity of neurotoxic astrocytes. Silencing PirB expression had the opposite effect and exacerbated the transition of reactive astrocytes to a neurotoxic state in vitro. Moreover, PirB-impaired astrocytes demonstrated STAT3 hyperphosphorylation which could be reversed by stattic (p-STAT3 inhibitor). Furthermore, Golgi-Cox staining confirmed that dendrite morphology defects and synapse-related protein were significantly increased in PirB-overexpressed SD mice. Our data demonstrated that SD induced neurotoxic reactive astrocytes and contributed to neuroinflammation and cognitive deficits. PirB performs a negative regulatory role in neurotoxic reactive astrocytes via the STAT3 signaling pathway in SD.
Collapse
Affiliation(s)
- Liya Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Yan Mou
- The Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Qian Zhai
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xin Zhang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Mengyu Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Zhaoyang Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
| |
Collapse
|
21
|
Bagheri A, Radman G, Aria N, Rezaei F, Khajenouri M, Ghiabi S, Bagheri Y. The Effects of Quercetin on Apoptosis and Antioxidant Activity in a Renal Ischemia/Reperfusion Injury Animal Model. Drug Res (Stuttg) 2023. [PMID: 36972618 DOI: 10.1055/a-1999-7600] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Renal ischemia-reperfusion injury (IRI) is considered as one of the most prevalent causes of acute kidney injury (AKI), which can happen in various clinical situations including hypovolemic shock, injury, thrombo-embolism, and after a kidney transplant. This paper aims to evaluate the reno-protective effects of Quercetin in induced ischemia/reperfusion injury by regulating apoptosis-related proteins, inflammatory cytokines, MMP-2, MMP-9, and nuclear factor kappa-light-chain-enhancer inactivated B cells (NF-kB) in rats. The male Wistar rats (n=32) were randomly divided into Sham, untreated IR, and Quercetin-treated IR (gavage and intraperitoneal). Quercetin was given orally and intraperitoneally one hour before inducing ischemia-reperfusion injury . After reperfusion, blood samples and kidneys were collected to assess renal function and inflammatory cytokines, apoptotic signaling proteins, and antioxidants. Urea, creatinine, and MDA levels improved in Quercetin-treated groups with different administration methods. In addition, the activities of other antioxidant in Quercetin-treated rats were higher than those in the IR group. Further, Quercetin inhibited NF-kB signaling, apoptosis-associated factors and produced matrix metalloproteinase protein in the kidneys of rats. Based on the findings, the antioxidant, anti-inflammatory, and anti-apoptotic effects of the Quercetin diminished renal ischemia-reperfusion injury in the rats significantly. It is suggested that a single dosage of Quercetin have a reno-protective impact in the case of renal I/R injury.
Collapse
Affiliation(s)
- Amin Bagheri
- Department of Urology, Sina Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghazal Radman
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Negar Aria
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Rezaei
- Faculty of Pharmacy in Hradec Kralove, Charles University, Prague, Czech Republic
| | - Mohammad Khajenouri
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Shamim Ghiabi
- Department of Medical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
| | - Yasin Bagheri
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Exosomal PD-L1 confers chemoresistance and promotes tumorigenic properties in esophageal cancer cells via upregulating STAT3/miR-21. Gene Ther 2023; 30:88-100. [PMID: 35440807 DOI: 10.1038/s41434-022-00331-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/12/2022] [Accepted: 03/02/2022] [Indexed: 11/08/2022]
Abstract
Chemotherapy resistance remains a major obstacle in the treatment of esophageal cancer. Previous researches have shown that an increase in exosomal PD-L1 expression was positively associated with a more advanced clinical stage, a poorer prognosis as well as drug resistance in patients with esophageal squamous cell carcinoma (ESCC). To explore the role of exosomal PD-L1 in ESCC, we performed bioinformatics analysis as well as several in vitro/in vivo functional experiments in a parental sensitive cell line EC-9706 and its derivative, a paclitaxel-resistant subline EC-9706R, and found that the exosomal PD-L1 from EC-9706R was higher than that from EC-9706. Moreover, exosomes from EC-9706R significantly increased invasion, migration and chemoresistance of EC-9706. Anti-PD-L1 treatment in combination with chemotherapy also led to reduced tumor burden in vivo. Inhibition of the release of exosomes by GW4869 or inhibition of STAT3 phosphorylation by stattic could effectively reverse the resistance to paclitaxel mediated by exosomal PD-L1. Furthermore, we found that PD-L1, miR-21, and multidrug resistance (MDR1) gene are involved in the process of exosomal transfer. Moreover, PD-L1 could enhance miR-21 expression by increasing the enrichment of STAT3 on miR-21 promoter. Our results suggested that exosomal PD-L1 may contribute to drug resistance to paclitaxel by regulating the STAT3/miR-21/PTEN/Akt axis and promote tumorigenic phenotype. This study provides a novel potential therapeutic approach to reverse chemoresistance and tumor progression through exosomal PD-L1 in ESCC patients.
Collapse
|
23
|
Jafari S, Heydarian S, Lai R, Mehdizadeh Aghdam E, Molavi O. Silibinin induces immunogenic cell death in cancer cells and enhances the induced immunogenicity by chemotherapy. BIOIMPACTS : BI 2023; 13:51-61. [PMID: 36816998 PMCID: PMC9923812 DOI: 10.34172/bi.2022.23698] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 11/09/2022]
Abstract
Introduction: Silibinin is a natural flavonoid compound known to induce apoptosis in cancer cells. Despite silibinin's safety and efficacy as an anticancer drug, its effects on inducing immunogenic cell death (ICD) are largely unknown. Herein, we have evaluated the stimulating effects of silibinin on ICD in cancer cells treated with silibinin alone or in combination with chemotherapy. Methods: The anticancer effect of silibinin, alone or in combination with doxorubicin or oxaliplatin (OXP), was assessed using the MTT assay. Compusyn software was used to analyze the combination therapy data. Western blotting was conducted to examine the level of STAT3 activity. Flow cytometry was used to analyze calreticulin (CRT) and apoptosis. The heat shock protein (HSP70), high mobility group box protein1 (HMGB1), and IL-12 levels were assessed by ELISA. Results: Compared to the negative control groups, silibinin induced ICD in CT26 and B16F10 cells and significantly enhanced the induction of this type of cell death by doxorubicin, and these changes were allied with substantial increases in the level of damage-associated molecular patterns (DAMPs) including CRT, HSP70, and HMGB1. Furthermore, conditioned media from cancer cells exposed to silibinin and doxorubicin was found to stimulate IL-12 secretion in dendritic cells (DCs), suggesting the link of this treatment with the induction of Th1 response. Silibinin did not augment the ICD response induced by OXP. Conclusion: Our findings showed that silibinin can induce ICD and it potentiates the induction of this type of cell death induced by chemotherapy in cancer cells.
Collapse
Affiliation(s)
- Sevda Jafari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Heydarian
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raymond Lai
- Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Elnaz Mehdizadeh Aghdam
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding author: Ommoleila Molavi,
| |
Collapse
|
24
|
Jie Y, Yang X, Chen W. Pulsatilla Decoction Combined with 5-Fluorouracil Triggers Immunogenic Cell Death in Colorectal Cancer Cells. Cancer Biother Radiopharm 2022; 37:945-954. [PMID: 34042519 DOI: 10.1089/cbr.2020.4369] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Our research is designed to explore the role of 5-FU and Pulsatilla decoction (PD) through modulation of Immunogenic cell death (ICD) for the co-treatment of Colorectal cancer (CRC). Materials and Methods: Cell viability was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT) assays. Cell apoptosis was assessed using flow cytometry. Phosphorylation of STAT3 and expression of Mcl-1 and Bcl-xl were measured by Western blot assays. The levels of ATP and HMGB1 in the supernatants of the culture medium were analyzed by ATP assays and the HMGB1 enzyme linked immunosorbent assay kit. The cell surface levels of CRT were measured by immunofluorescence assays. The tumor growth was analyzed in mice. Results: PD increased 5-FU-induced ICD in CRC cells, as demonstrated by the extracellular levels of adenosine triphosphate (ATP) and high-mobility group box 1 (HMGB1), and the surface levels of calreticulin (CRT). Our mechanism study showed that PD promoted 5-FU-induced ICD by inactivating signal transducer and activator of transcription 3 (STAT3). Furthermore, the co-treatment of 5-FU and PD further promoted 5-FU-induced CRT expression and T cell infiltration in vivo. Tumorigenicity analysis revealed that 5-FU combined with PD notably reduced tumor growth. Conclusion: This study indicated that PD enhances 5-FU-induced ICD and anti-tumor effect in CRC by inactivating STAT3. The combined application of 5-FU with PD may improve the anti-tumor activity of 5-FU in CRC.
Collapse
Affiliation(s)
- Yanghua Jie
- Radiotherapy Center, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, China
| | - Xiaobei Yang
- Department of Anorectal, Urumqi City Hospital of Traditional Chinese Medicine, Urumqi, China
| | - Weidong Chen
- Department of Anorectal, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, China
| |
Collapse
|
25
|
Xie D, Wang Q, Wu G. Research progress in inducing immunogenic cell death of tumor cells. Front Immunol 2022; 13:1017400. [PMID: 36466838 PMCID: PMC9712455 DOI: 10.3389/fimmu.2022.1017400] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/02/2022] [Indexed: 08/29/2023] Open
Abstract
Immunogenic cell death (ICD) is a regulated cell death (RCD) pathway. In response to physical and chemical signals, tumor cells activate specific signaling pathways that stimulate stress responses in the endoplasmic reticulum (ER) and expose damage-associated molecular patterns (DAMPs), which promote antitumor immune responses. As a result, the tumor microenvironment is altered, and many tumor cells are killed. The ICD response in tumor cells requires inducers. These inducers can be from different sources and contribute to the development of the ICD either indirectly or directly. The combination of ICD inducers with other tumor treatments further enhances the immune response in tumor cells, and more tumor cells are killed; however, it also produces side effects of varying severity. New induction methods based on nanotechnology improve the antitumor ability and significantly reduces side effects because they can target tumor cells precisely. In this review, we introduce the characteristics and mechanisms of ICD responses in tumor cells and the DAMPs associated with ICD responses, summarize the current methods of inducing ICD response in tumor cells in five distinct categories: chemical sources, physical sources, pathogenic sources, combination therapies, and innovative therapies. At the same time, we introduce the limitations of current ICD inducers and make a summary of the use of ICD responses in clinical trials. Finally, we provide an outlook on the future of ICD inducer development and provide some constructive suggestions.
Collapse
Affiliation(s)
| | - Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
26
|
Jafari S, Bakhshaei A, Eskandani M, Molavi O. Silibinin-Loaded Nanostructured Lipid Carriers for Growth Inhibition of Cisplatin-Resistant Ovarian Cancer Cells. Assay Drug Dev Technol 2022; 20:339-348. [DOI: 10.1089/adt.2022.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Sevda Jafari
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atabak Bakhshaei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology (RCPN), Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Mansoor A, Khurshid Z, Khan MT, Mansoor E, Butt FA, Jamal A, Palma PJ. Medical and Dental Applications of Titania Nanoparticles: An Overview. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12203670. [PMID: 36296859 PMCID: PMC9611494 DOI: 10.3390/nano12203670] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 05/25/2023]
Abstract
Currently, titanium oxide (TiO2) nanoparticles are successfully employed in human food, drugs, cosmetics, advanced medicine, and dentistry because of their non-cytotoxic, non-allergic, and bio-compatible nature when used in direct close contact with the human body. These NPs are the most versatile oxides as a result of their acceptable chemical stability, lower cost, strong oxidation properties, high refractive index, and enhanced aesthetics. These NPs are fabricated by conventional (physical and chemical) methods and the latest biological methods (biological, green, and biological derivatives), with their advantages and disadvantages in this epoch. The significance of TiO2 NPs as a medical material includes drug delivery release, cancer therapy, orthopedic implants, biosensors, instruments, and devices, whereas their significance as a dental biomaterial involves dentifrices, oral antibacterial disinfectants, whitening agents, and adhesives. In addition, TiO2 NPs play an important role in orthodontics (wires and brackets), endodontics (sealers and obturating materials), maxillofacial surgeries (implants and bone plates), prosthodontics (veneers, crowns, bridges, and acrylic resin dentures), and restorative dentistry (GIC and composites).
Collapse
Affiliation(s)
- Afsheen Mansoor
- Department of Dental Material Sciences, School of Dentistry, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad 44080, Pakistan
| | - Zohaib Khurshid
- Department of Prosthodontics and Dental Implantology, College of Dentistry, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Muhammad Talal Khan
- Department of Dental Biomaterials, Bakhtawar Amin Medical and Dental College, Multan 60650, Pakistan;
| | - Emaan Mansoor
- Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan;
| | - Faaz Ahmad Butt
- Department of Materials Engineering, NED University of Engineering & Technology, Karachi 74200, Pakistan;
| | - Asif Jamal
- Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Paulo J. Palma
- Center for Innovation and Research in Oral Sciences (CIROS), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Institute of Endodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| |
Collapse
|
28
|
Malekinejad Z, Baghbanzadeh A, Nakhlband A, Baradaran B, Jafari S, Bagheri Y, Raei F, Montazersaheb S, Farahzadi R. Recent clinical findings on the role of kinase inhibitors in COVID-19 management. Life Sci 2022; 306:120809. [PMID: 35841979 PMCID: PMC9278000 DOI: 10.1016/j.lfs.2022.120809] [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: 05/18/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022]
Abstract
The highly pathogenic, novel coronavirus disease (COVID-19) outbreak has emerged as a once-in-a-century pandemic with poor consequences, urgently calling for new therapeutics, cures, and supportive interventions. It has already affected over 250 million people worldwide; thereby, there is a need for novel therapies to alleviate the related complications. There is a paradigm shift in developing drugs and clinical practices to combat COVID-19. Several clinical trials have been performed or are testing diverse pharmacological interventions to alleviate viral load and complications such as cytokine release storm (CRS). Kinase-inhibitors have appeared as potential antiviral agents for COVID-19 patients due to their efficacy against CRS. Combination of kinase inhibitors with other therapies can achieve more efficacy against COVID-19. Based on the pre-clinical trials, kinase inhibitors such as Janus kinase-signal transducer and activator of transcription (JAK/STAT) inhibitors, Brutton's tyrosin kinase (BTK) inhibitors, p38 mitogen-activated protein kinases (p38 MAPK) inhibitors, Glycogen synthase kinase 3 (GSK-3) inhibitors can be a promising strategy against COVID-19. Kinase inhibitors possess crucial pharmacological properties for a successful re-purposing in terms of dual anti-inflammatory and anti-viral effects. This review will address the current clinical evidence and the newest discovery regarding the application of kinase inhibitors in COVID-19. An outlook on ongoing clinical trials (clinicaltrials.gov) and unpublished data is also presented here. Besides, Kinase inhibitors' function on COVID-19-mediated CRS is discussed.
Collapse
Affiliation(s)
- Zahra Malekinejad
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ailar Nakhlband
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Jafari
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yasin Bagheri
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Raei
- Departement of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Rodrigues MC, Morais JAV, Ganassin R, Oliveira GRT, Costa FC, Morais AAC, Silveira AP, Silva VCM, Longo JPF, Muehlmann LA. An Overview on Immunogenic Cell Death in Cancer Biology and Therapy. Pharmaceutics 2022; 14:pharmaceutics14081564. [PMID: 36015189 PMCID: PMC9413301 DOI: 10.3390/pharmaceutics14081564] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
Immunogenic cell death (ICD) is a modality of regulated cell death that is sufficient to promote an adaptive immune response against antigens of the dying cell in an immunocompetent host. An important characteristic of ICD is the release and exposure of damage-associated molecular patterns, which are potent endogenous immune adjuvants. As the induction of ICD can be achieved with conventional cytotoxic agents, it represents a potential approach for the immunotherapy of cancer. Here, different aspects of ICD in cancer biology and treatment are reviewed.
Collapse
Affiliation(s)
- Mosar Corrêa Rodrigues
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - José Athayde Vasconcelos Morais
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Rayane Ganassin
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Giulia Rosa Tavares Oliveira
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Fabiana Chagas Costa
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Amanda Alencar Cabral Morais
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Ariane Pandolfo Silveira
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Victor Carlos Mello Silva
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - João Paulo Figueiró Longo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
| | - Luis Alexandre Muehlmann
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-275, Brazil; (M.C.R.); (J.A.V.M.); (R.G.); (G.R.T.O.); (F.C.C.)
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (A.A.C.M.); (A.P.S.); (V.C.M.S.); (J.P.F.L.)
- Correspondence:
| |
Collapse
|
30
|
Li Y, Song Z, Han Q, Zhao H, Pan Z, Lei Z, Zhang J. Targeted inhibition of STAT3 induces immunogenic cell death of hepatocellular carcinoma cells via glycolysis. Mol Oncol 2022; 16:2861-2880. [PMID: 35665592 PMCID: PMC9348600 DOI: 10.1002/1878-0261.13263] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/15/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022] Open
Abstract
In hepatocellular carcinoma (HCC), the signal transducer and activator of transcription 3 (STAT3) is present in an overactive state that is closely related to tumour development and immune escape. STAT3 inhibition reshapes the tumour immune microenvironment, but the underlying mechanisms have not been fully clarified. We found that STAT3 inhibition could induce immunogenic cell death (ICD) of HCC cells via translocation of the “eat me” molecule calreticulin to the cell surface and a significant reduction in the expression of the “don’t eat me” molecule leucocyte surface antigen CD47. STAT3 inhibition promoted dendritic cell (DC) activation and enhanced the recognition and phagocytosis of HCC cells by macrophages. Furthermore, STAT3 inhibition prevented the expression of key glycolytic enzymes, facilitating the induction of ICD in HCC. Interestingly, STAT3 directly regulated the transcription of CD47 and solute carrier family 2 member 1 (SLC2A1; also known as GLUT1). In subcutaneous and orthotopic transplantation mouse tumour models, the STAT3 inhibitor napabucasin prevented tumour growth and induced the expression of calreticulin and the protein disulfide isomerase family A member 3 (PDIA3; also known as ERp57) but suppressed that of CD47 and GLUT1. Meanwhile, the amount of tumour‐infiltrated DCs and macrophages increased, along with the expression of costimulatory molecules. More CD4+ and CD8+ T cells accumulated in tumour tissues, and CD8+ T cells had lower expression of checkpoint molecules such as lymphocyte activation gene 3 protein (LAG‐3) and programmed cell death protein 1 (PD‐1). Significantly, the antitumour immune memory response was induced by treatment targeting STAT3. These findings provide a new mechanism for targeting STAT3‐induced ICD in HCC, and confirms STAT3 as a potential target for the treatment of HCC via reshaping the tumour immune microenvironment.
Collapse
Affiliation(s)
- Ya Li
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhenwei Song
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhaoyi Pan
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhengyang Lei
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
31
|
Ejigah V, Mandala B, Akala EO. Nanotechnology in the development of small and large molecule tyrosine kinase inhibitors and immunotherapy for the treatment of HER2-positive breast cancer. JOURNAL OF CANCER & METASTASIS RESEARCH 2022; 4:6-22. [PMID: 38966076 PMCID: PMC11223443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
The HER2 receptor tyrosine kinase is a member of the epidermal growth factor receptor family which includes EGFR, HER3 and HER4. They are known to play critical roles in both normal development and cancer. A subset of breast cancers is associated with the HER2 gene, which is amplified and/or overexpressed in 20-25% of invasive breast cancers and is correlated with tumor resistance to chemotherapy, Metastatic Breast Cancer (MBC) and poor patient survival. The advent of receptor tyrosine kinase inhibitors has improved the prognosis of HER2-postive breast cancers; however, HER2+MBC invariably progresses (acquired resistance or de novo resistance). The monoclonal antibody-based drugs (large molecule TKIs) target the extracellular binding domain of HER2; while the small molecule TKIs act intracellularly to inhibit proliferation and survival signals. We reviewed the modes of action of the TKIs with a view to showing which of the TKIs could be combined in nanoparticles to benefit from the power of nanotechnology (reduced toxicity, improved solubility of hydrophobic drugs, long circulation half-lives, circumventing efflux pumps and preventing capture by the reticuloendothelial system (mononuclear phagocyte system). Nanotherapeutics also mediate the synchronization of the pharmacokinetics and biodistribution of multiple drugs incorporated in the nanoparticles. Novel TKIs that are currently under investigation with or without nanoparticle delivery are mentioned, and nano-based strategies to improve their delivery are suggested. Immunotherapies currently in clinical practice, clinical trials or at the preclinical stage are discussed. However, immunotherapy only works well in relatively small subsets of patients. Combining nanomedicine with immunotherapy can boost therapeutic outcomes, by turning "cold" non-immunoresponsive tumors and metastases into "hot" immunoresponsive lesions.
Collapse
Affiliation(s)
- Victor Ejigah
- Department of Pharmaceutical Sciences, College of Pharmacy Howard University Washington DC, Center for Drug Research and Development (CDRD), USA
| | - Bharathi Mandala
- Department of Pharmaceutical Sciences, College of Pharmacy Howard University Washington DC, Center for Drug Research and Development (CDRD), USA
| | - Emmanuel O Akala
- Department of Pharmaceutical Sciences, College of Pharmacy Howard University Washington DC, Center for Drug Research and Development (CDRD), USA
| |
Collapse
|
32
|
Jin W, Zhang T, Zhou W, He P, Sun Y, Hu S, Chen H, Ma X, Peng Y, Yi Z, Liu M, Chen Y. Discovery of 2-Amino-3-cyanothiophene Derivatives as Potent STAT3 Inhibitors for the Treatment of Osteosarcoma Growth and Metastasis. J Med Chem 2022; 65:6710-6728. [PMID: 35476936 DOI: 10.1021/acs.jmedchem.2c00004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Osteosarcoma is one of the most common malignant bone tumors. However, the treatment and clinical outcomes of osteosarcoma have hardly changed over the past three decades due to the comprehensive heterogeneity and higher rate of mutation of osteosarcoma. Recent studies have shown that STAT3 has the potential to suppress the proliferation and metastasis of osteosarcoma. In this study, a novel class of 2-amino-3-cyanothiophene derivatives were designed and synthesized to inhibit osteosarcoma by targeting STAT3. Representative compound 6f showed potent antiproliferative effects against osteosarcoma cells, directly bound to the STAT3 SH2 domain with a KD of 0.46 μM, and inhibited the phosphorylation of STAT3 Y705 in a dose-dependent manner. Furthermore, compound 6f promoted osteosarcoma cell apoptosis in vitro and significantly suppressed the growth and metastasis of osteosarcoma in vivo. These findings demonstrate that targeting STAT3 may be a feasible therapeutic strategy for the treatment of metastatic osteosarcoma.
Collapse
Affiliation(s)
- Wangrui Jin
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Tao Zhang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wenbo Zhou
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Peng He
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yue Sun
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shijia Hu
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Huang Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xinglong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yangrui Peng
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
33
|
Yang C, Ming Y, Zhou K, Hao Y, Hu D, Chu B, He X, Yang Y, Qian Z. Macrophage Membrane-Camouflaged shRNA and Doxorubicin: A pH-Dependent Release System for Melanoma Chemo-Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9768687. [PMID: 35233535 PMCID: PMC8851070 DOI: 10.34133/2022/9768687] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/12/2021] [Indexed: 02/05/2023]
Abstract
Improving the efficacy of melanoma treatment remains an important global challenge. Here, we combined chemotherapy with protein tyrosine phosphatase nonreceptor type 2(Ptpn2) based immunotherapy in an effort to address this challenge. Short-hairpin RNA (shRNA) targeting Ptpn2 was coencapsulated with doxorubicin (DOX) in the cell membrane of M1 macrophages (M1HD@RPR). The prepared nanoparticles (NPs) were effectively phagocytosed by B16F10 cells and M1 macrophages, but not by M0 macrophages. Hence, NP evasion from the reticuloendothelial system (RES) was improved and NP enrichment in tumor sites increased. M1HD@RPR can directly kill tumor cells and stimulate immunogenic cell death (ICD) by DOX and downregulate Ptpn2. It can promote M1 macrophage polarization and dendritic cell maturation and increase the proportion of CD8+ T cells. M1HD@RPR killed and inhibited the growth of primary melanoma and lung metastatic tumor cells without harming the surrounding tissue. These findings establish M1HD@RPR as a safe multifunctional nanoparticle capable of effectively combining chemotherapy and gene immunotherapies against melanoma.
Collapse
Affiliation(s)
- Chengli Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China.,Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, China
| | - Yang Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Kai Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Ying Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Danrong Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Bingyang Chu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Yun Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| |
Collapse
|
34
|
Widjaya AS, Liu Y, Yang Y, Yin W, Liang J, Jiang Y. Tumor-permeable smart liposomes by modulating the tumor microenvironment to improve the chemotherapy. J Control Release 2022; 344:62-79. [PMID: 35182612 DOI: 10.1016/j.jconrel.2022.02.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 12/14/2022]
Abstract
Low levels of accumulation and permeability in tumors are two primary reasons for the limited efficacy of conventional antineoplastic nanodrugs. In the present study, based on an original corosolic acid liposome (CALP) carrier with the functions of cell penetration, tumor permeability and anti-inflammation developed by our previous work, a versatile PTX/CALP was achieved by CALP loading paclitaxel (PTX). Compared to conventional PTX liposomes (PTX/LP) prepared by cholesterol and phospholipid, PTX/CALP exhibited extremely increasing cellular uptake and cytotoxicity in vitro, and in vivo enhancing the accumulation and permeability of tumor, thus significantly improving the antitumor efficacy. Further evidence indicated that PTX/CALP conspicuously promoted the recruitment of CD8+ T cells as well as reduced the infiltration of regulatory T cells and M2 macrophages into tumor by inducing enhanced immunogenic cell death (ICD) and down-regulating the inflammation level. Therefore, the improvement of efficacy was also attributed to the superiorities of PTX/CALP in modulating the inflammatory and immunosuppressive tumor microenvironment. Overall, the smart PTX liposomes based on the multi-functional CALP carrier without any modification could overcome the harsh tumor biological barriers, enhance the induction of ICD and then achieve satisfactory efficacy, suggesting its promising potentials in industrial transfer and clinical application.
Collapse
Affiliation(s)
- Andy Samuel Widjaya
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yunhu Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yueying Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weiwei Yin
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianying Liang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yanyan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
35
|
Emerging Therapeutic Agents for Colorectal Cancer. Molecules 2021; 26:molecules26247463. [PMID: 34946546 PMCID: PMC8707340 DOI: 10.3390/molecules26247463] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
There are promising new therapeutic agents for CRC patients, including novel small-molecule inhibitors and immune checkpoint blockers. We focused on emerging CRC’s therapeutic agents that have shown the potential for progress in clinical practice. This review provides an overview of tyrosine kinase inhibitors targeting VEGF and KIT, BRAF and MEK inhibitors, TLR9 agonist, STAT3 inhibitors, and immune checkpoint blockers (PD1/PDL-1 inhibitors), for which recent advances have been reported. These new agents have the potential to provide benefits to CRC patients with unmet medical needs.
Collapse
|
36
|
Rahimi MM, Bagheri A, Bagheri Y, Fathi E, Bagheri S, Nia AV, Jafari S, Montazersaheb S. Renoprotective effects of prazosin on ischemia-reperfusion injury in rats. Hum Exp Toxicol 2021; 40:1263-1273. [PMID: 33559503 DOI: 10.1177/0960327121993224] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Renal ischemia-reperfusion (IR) injury is one of the main leading causes of acute kidney injury associated with inflammation, oxidative stress and cell apoptosis. We studied the effects of prazosin, as a specific blocker of α1-AR, on renal IR injury. METHODS Rats were divided into normal control; untreated IR and prazosin-treated IR (1 mg/kg body weight). Prazosin was administered by intraperitoneal injection 30 min prior to IR induction. The level of urea/creatinine and oxidative factors were detected by colorimetric methods. Apoptosis-associated factors, inflammatory, and signaling proteins were analyzed in renal tissue. The abnormalities of renal histopathology were detected by immunohistochemistry. RESULTS Administration of prazosin to IR rats ameliorated serum urea and creatinine and IR-induced histopathological damages. Lipid peroxidation was significantly improved after treatment by prazosin in IR injury rats, however, antioxidant status was not affected. Rats subjected to IR injury activated Bax protein and NF-κB mediated inflammatory response. Moreover, treatment with prazosin inhibited renal NF-κB activation, resulting in a significant decline in pro-inflammatory cytokine of IL-6. CONCLUSION These findings suggest that prazosin could be a good candidate to attenuate renal IR injury due to its ability to modulate renal function, apoptosis and inflammation.
Collapse
Affiliation(s)
- M M Rahimi
- Kidney Research Center, 48432Tabriz University of Medical Sciences, Tabriz, Iran
| | - A Bagheri
- Department of Urology, Sina Hospital, 48432Tabriz University of Medical Sciences, Tabriz, Iran
| | - Y Bagheri
- Young Researchers and Elite Club, 201583Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - E Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, 56947University of Tabriz, Tabriz, Iran
| | - S Bagheri
- 475027Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - A V Nia
- 475027Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - S Jafari
- Kidney Research Center, 48432Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, 48432Tabriz University of Medical Sciences, Tabriz, Iran
| | - S Montazersaheb
- Molecular Medicine Research Center, 48432Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
37
|
Xuan Y, Guan M, Zhang S. Tumor immunotherapy and multi-mode therapies mediated by medical imaging of nanoprobes. Theranostics 2021; 11:7360-7378. [PMID: 34158855 PMCID: PMC8210602 DOI: 10.7150/thno.58413] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/14/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy is an effective tumor treatment strategy that has several advantages over conventional methods such as surgery, radiotherapy and chemotherapy. Studies show that multifunctional nanoprobes can achieve multi-mode image-guided multiple tumor treatment modes. The tumor cells killed by chemotherapies or phototherapies release antigens that trigger an immune response and augment the effects of tumor immunotherapy. Thus, combining immunotherapy and multifunctional nanoprobes can achieve early cancer diagnosis and treatment. In this review, we have summarized the current research on the applications of multifunctional nanoprobes in image-guided immunotherapy. In addition, image-guided synergistic chemotherapy/photothermal therapy/photodynamic therapy and immunotherapy have also been discussed. Furthermore, the application potential and clinical prospects of multifunctional nanoprobes in combination with immunotherapy have been assessed.
Collapse
Affiliation(s)
| | | | - Shubiao Zhang
- Key Lab of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, 116600, China
| |
Collapse
|
38
|
Li H, Qin S, Liu Y, Chen Z, Ren Z, Xiong J, Meng Z, Zhang X, Wang L, Zhang X, Zou J. Camrelizumab Combined with FOLFOX4 Regimen as First-Line Therapy for Advanced Hepatocellular Carcinomas: A Sub-Cohort of a Multicenter Phase Ib/II Study. Drug Des Devel Ther 2021; 15:1873-1882. [PMID: 33976538 PMCID: PMC8106453 DOI: 10.2147/dddt.s304857] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors and chemotherapy can synergistically increase efficacy in a variety of malignancies. We conducted this phase Ib/II study to assess the safety and efficacy of anti-PD-1 antibody camrelizumab in combination with FOLFOX4 for treatment-naive advanced hepatocellular carcinoma (aHCC). METHODS This open-label, multicenter phase Ib/II study (NCT03092895) enrolled patients with aHCC and without prior systemic treatment for treatment with camrelizumab (3 mg/kg) and FOLFOX4 every two weeks. First, six patients were enrolled, followed by an additional 28 patients after dose-limiting toxicity cases were determined to be <33% of patients. The primary endpoint was tolerability and safety of treatment. RESULTS A total of 34 aHCC patients were enrolled and received study treatment. No dose-limiting toxicity were observed in the first six patients enrolled. Twenty-nine (85.3%) of the total 34 patients had grade ≥3 treatment-related adverse events (TRAEs), with the most common ones being decreased neutrophil count (55.9%) and decreased white blood cell count (38.2%). No TRAEs-related deaths occurred. The objective response and disease control rate were 29.4% (95% CI, 15.1-47.5) and 79.4% (95% CI, 62.1-91.3), respectively. The median duration of response, progression-free survival, and overall survival was 6.9 months (range, 3.3-11.5), 7.4 months (95% CI, 3.9-9.2), and 11.7 months (95% CI, 8.2-22.0), respectively. CONCLUSION Camrelizumab combined with FOLFOX4 for first-line treatment of patients with aHCC showed good safety and tolerability, with promising preliminary antitumor activity.
Collapse
Affiliation(s)
- Hui Li
- Department of Medical Oncology Center, Bayi Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Shukui Qin
- Department of Medical Oncology Center, Bayi Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Ying Liu
- Department of Oncology, Cancer Hospital of Henan Province, Zhengzhou, People’s Republic of China
| | - Zhendong Chen
- Department of Clinical Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Zhenggang Ren
- Department of Clinical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Jianping Xiong
- Department of Clinical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Zhiqiang Meng
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
| | - Xiao Zhang
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, People’s Republic of China
| | - Linna Wang
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, People’s Republic of China
| | - Xiaojing Zhang
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, People’s Republic of China
| | - Jianjun Zou
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, People’s Republic of China
| |
Collapse
|
39
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Rahmani Moghadam E, Raei M, Kalantari M, Tavakol S, Mohammadinejad R, Najafi M, Tay FR, Makvandi P. Progress in Natural Compounds/siRNA Co-delivery Employing Nanovehicles for Cancer Therapy. ACS COMBINATORIAL SCIENCE 2020; 22:669-700. [PMID: 33095554 PMCID: PMC8015217 DOI: 10.1021/acscombsci.0c00099] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Chemotherapy using natural compounds, such as resveratrol, curcumin, paclitaxel, docetaxel, etoposide, doxorubicin, and camptothecin, is of importance in cancer therapy because of the outstanding therapeutic activity and multitargeting capability of these compounds. However, poor solubility and bioavailability of natural compounds have limited their efficacy in cancer therapy. To circumvent this hurdle, nanocarriers have been designed to improve the antitumor activity of the aforementioned compounds. Nevertheless, cancer treatment is still a challenge, demanding novel strategies. It is well-known that a combination of natural products and gene therapy is advantageous over monotherapy. Delivery of multiple therapeutic agents/small interfering RNA (siRNA) as a potent gene-editing tool in cancer therapy can maximize the synergistic effects against tumor cells. In the present review, co-delivery of natural compounds/siRNA using nanovehicles are highlighted to provide a backdrop for future research.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty
of Engineering and Natural Sciences, Sabanci
University, Orta Mahalle,
Üniversite Caddesi No. 27, Orhanlı,
Tuzla, 34956 Istanbul, Turkey
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Ali Zarrabi
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Kiavash Hushmandi
- Department
of Food Hygiene and Quality Control, Division of Epidemiology &
Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran
| | - Farid Hashemi
- Department
of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department
of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Mehdi Raei
- Health Research
Center, Life Style Institute, Baqiyatallah
University of Medical Sciences, Tehran 1435916471, Iran
| | - Mahshad Kalantari
- Department
of Genetics, Tehran Medical Sciences Branch, Azad University, Tehran 19168931813, Iran
| | - Shima Tavakol
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 1449614525, Iran
| | - Reza Mohammadinejad
- Pharmaceutics
Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Masoud Najafi
- Medical
Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Radiology
and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Franklin R. Tay
- College
of Graduate Studies, Augusta University, Augusta, Georgia 30912, United States
| | - Pooyan Makvandi
- Istituto
Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa Italy
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, 14496-14535 Tehran, Iran
| |
Collapse
|
40
|
Jafari S, Mahyad B, Hashemzadeh H, Janfaza S, Gholikhani T, Tayebi L. Biomedical Applications of TiO 2 Nanostructures: Recent Advances. Int J Nanomedicine 2020; 15:3447-3470. [PMID: 32523343 PMCID: PMC7234979 DOI: 10.2147/ijn.s249441] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
Titanium dioxide (TiO2) nanostructures are one of the most plentiful compounds that have emerged in various fields of technology such as medicine, energy and biosensing. Various TiO2 nanostructures (nanotubes [NTs] and nanowires) have been employed in photoelectrochemical (PEC) biosensing applications, greatly enhancing the detection of targets. TiO2 nanostructures, used as reinforced material or coatings for the bare surface of titanium implants, are excellent additive materials to compensate titanium implants deficiencies-like poor surface interaction with surrounding tissues-by providing nanoporous surfaces and hierarchical structures. These nanostructures can also be loaded by diversified drugs-like osteoporosis drugs, anticancer and antibiotics-and used as local drug delivery systems. Furthermore, TiO2 nanostructures and their derivatives are new emerging antimicrobial agents to overcome human pathogenic microorganisms. However, like all other nanomaterials, toxicity and biocompatibility of TiO2 nanostructures must be considered. This review highlights recent advances, along with the properties and numerous applications of TiO2-based nanostructure compounds in nano biosensing, medical implants, drug delivery and antibacterial fields. Moreover, in the present study, some recent advances accomplished on the pharmaceutical applications of TiO2 nanostructures, as well as its toxicity and biocompatibility, are presented.
Collapse
Affiliation(s)
- Sevda Jafari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Baharak Mahyad
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, Islamic Republic of Iran
| | - Hadi Hashemzadeh
- Department of Nanobiotechnology, Tarbiat Modares University, Tehran, 14117, Islamic Republic of Iran
| | - Sajjad Janfaza
- Department of Nanobiotechnology, Tarbiat Modares University, Tehran, 14117, Islamic Republic of Iran
| | - Tooba Gholikhani
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI53233, USA
| |
Collapse
|
41
|
Zahednezhad F, Zakeri-Milani P, Shahbazi Mojarrad J, Valizadeh H. The latest advances of cisplatin liposomal formulations: essentials for preparation and analysis. Expert Opin Drug Deliv 2020; 17:523-541. [DOI: 10.1080/17425247.2020.1737672] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Fahimeh Zahednezhad
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Science, Iran
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Shahbazi Mojarrad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Science, Iran
| |
Collapse
|